Archives

TSH Receptor Mutations and Diseases

ABSTRACT

The thyroid-stimulating hormone (TSH) receptor (TSHR) is a member of the glycoprotein hormone receptors (GPHRs), a sub-group of class A G protein-coupled receptors (GPCRs). TSHR and its ligand thyrotropin are of essential importance for growth and function of the thyroid gland. The TSHR activates different G-protein subtypes and signaling pathways, of which Gs and Gq induced signaling are of highest importance in the thyroid gland. A proper interplay between TSH and TSHR is pivotal for thyroid growth and regulated production and release of thyroid hormones (TH). Autoimmune (antibody binding) or non-autoimmune (occurrence of mutants) TSHR dysfunctions are the underlying cause of several pathologies, including rare cancer-development. The sequential processes of TSHR binding, signal transduction across the cell-membrane and activation of intracellular effectors involve elaborate specific structural properties of the receptor and several interacting proteins. In consequence, different pathogenic mutations at TSHR or TSH may have diverse impact on particular molecular functions, but finally result in either hypo- or hyperthyroid states accompanied or not by various growth anomalies. We here summarize current knowledge regarding naturally occurring TSHR mutations, associated diseases and related molecular pathogenic mechanisms at the level of TSHR structure and function. For complete coverage of this and related areas in Endocrinology, visit our free web-books, www.endotext.org and www.thyroidmanager.org

 

 

GAIN-OF-FUNCTION MUTATIONS

On a theoretical basis, for a hormone receptor, “gain-of-function” may have several meanings: (i) activation in the absence of ligand (constitutively); (ii) increased sensitivity to its normal agonist; (iii) increased, or de novo sensitivity to an allosteric modulator; (iv) broadening of its specificity. When the receptor is part of a chemostat, as is the case for the TSHR, the first situation is expected to cause tissue autonomy, whereas the second would simply cause adjustment of TSH to a lower value. In the third and fourth cases, inappropriate stimulation of the target will occur because the illegitimate agonists or modulators are not expected to be subject to the normal negative feedback. If a gain-of-function mutation of the first category occurs in a single cell normally expressing the receptor (somatic mutation), it will become symptomatic only if the regulatory cascade controlled by the receptor is mitogenic in this particular cell type or, during development, if the mutation affects a progenitor contributing significantly to the final organ. Autonomous activity of the receptor will cause clonal expansion of the mutated cell. If the regulatory cascade also positively controls function, the resulting tumor may progressively take over the function of the normal tissue and ultimately result in autonomous hyperfunction. If the mutation is present in all cells of an organism (germline mutation) autonomy will be displayed by the whole organ.

From what we know of thyroid cell physiology it is easy to predict the phenotypes associated with gain-of-function of the cAMP-dependent regulatory cascade. Two observations provide pertinent models of this situation. Transgenic mice made to express ectopically the adenosine A2a receptor in their thyroid display severe hyperthyroidism associated with thyroid hyperplasia (1). As the A2a adenosine receptor is coupled to Gs and displays constitutive activity due to its continuous stimulation by ambient adenosine (2), this model mimics closely the situation expected for a gain-of-function germline mutation of the TSHR. Patients with the McCune-Albright syndrome are mosaïc for mutations in the Gs protein (Gsp mutations) leading to the constitutive stimulation of adenylyl-cyclase (3). Hyperfunctioning thyroid adenomas develop in these patients from cells harboring the mutation, making them a model for gain-of-function somatic mutations of the TSHR. A transgenic model in which Gsp mutations are targeted for expression in the mouse thyroid has been constructed. Though with a less dramatic phenotype this represents also a pertinent model for a gain-of-function of the cAMP regulatory cascade (4).

Since the TSHR is capable of activating both Gs and Gq (though with lower potency) the question arises whether mutations with a different effect on the two cascades would be associated with different phenotypes. Studies in mice (5) and rare patients (6) suggests that activation of Gq may be required to observe goitrogenesis in patients with non-autoimmune familial hyperthyroidism. However, when tested in transfected non-thyroid cells, all identified gain-of-function mutations of the TSHR stimulate constitutively Gs, with only a minority capable of stimulating both Gs and Gq (7,8). Also, thyroid adenomas or multinodular goitre are frequent in McCune Albright syndrome, which is characterized by pure Gs stimulation (9).

Familial non-autoimmune hyperthyroidism or hereditary toxic thyroid hyperplasia

 

The major cause of hyperthyroidism in adults is Graves' disease in which an autoimmune reaction is mounted against the thyroid gland and auto-antibodies are produced that recognize and stimulate the TSHR (10,11). This may explain why the initial description by the group of Leclère of a family showing segregation of thyrotoxicosis as an autosomal dominant trait in the absence of signs of autoimmunity was met with skepticism (12). Re-investigation of this family together with another family from Reims identified two mutations of the TSHR gene, which segregated in perfect linkage with the disease (13). A series of additional families have been studied since (14-28) [Figure 1 and Table 1, For a completed list of naturally occurring TSHR single amino acid substitutions with their functional characteristics, see the glycoprotein hormone receptor information resource “SSFA” (29,30) available under: http://www.ssfa-gphr.de]. The functional characteristics of these mutant receptors confirm that they are constitutively stimulated (see below). This nosological entity, hereditary toxic thyroid hyperplasia (HTTH), sometimes called Leclère’s disease, is characterized by the following clinical characteristics: autosomal dominant transmission; hyperthyroidism with a variable age of onset (from infancy to adulthood, even within a given family); hyperplastic goiter of variable size, but with a steady growth; absence of clinical or biological stigmata of auto-immunity. An observation common to most cases is the need for drastic ablative therapy (surgery or radioiodine) in order to control the disease, once the patient has become hyperthyroid (13,31). The autonomous nature of the thyroid tissue from these patients has been elegantly demonstrated by grafting in nude mice (32). Contrary to tissue from Graves' disease patients, HTTH cells continue to grow in the absence of stimulation by TSH or TSAb.

The prevalence of hereditary toxic thyroid hyperplasia is difficult to estimate. It is likely that many cases are still mistaken for Graves' disease. This may be explained by the high frequency of thyroid auto-antibodies (anti-thyroglobulin, anti-thyroperoxidase) in the general population. It is expected that wider knowledge of the existence of the disease will lead to better diagnosis. This is not a purely academic problem, since presymptomatic diagnosis in children of affected families may prevent the developmental or psychological complications associated with infantile or juvenile hyperthyroidism (for a review, see (33)). A country-wide screening for the condition has been performed in Denmark. It was found in one out of 121 patients with juvenile thyrotoxicosis (0.8%; 95% CI: 0.02-4.6%), which corresponds to one in 17 patients with presumed non-autoimmune juvenile thyrotoxicosis (6%; 95% CI:0.15-28.69) (34).

 Sporadic toxic thyroid hyperplasia

Cases with toxic thyroid hyperplasia have been described in children born from unaffected parents (35-39). Conspicuously, congenital hyperthyroidism was present in most of the cases and required aggressive treatment. Mutations of one TSHR allele were identified in the children, but were absent in the parents. As paternity was confirmed by mini- or microsatellite testing, these cases qualify as true neo-mutations. When comparing the amino acid substitutions implicated in hereditary and sporadic cases, for the majority, they do not overlap (see Table 1). Whereas most of the sporadic cases harbor mutations that are also found in toxic adenomas, most of the hereditary cases have "private" mutations. Although there may be exceptions, the analysis of the functional characteristics of the individual mutant receptors in COS cells, and the clinical course of individual patients, suggest an explanation for this observation: "sporadic" and somatic mutations seem to have a stronger activating effect than "hereditary" mutations (40). From their severe phenotypes, it is likely that newborns with neo-mutations would not have survived, if not treated efficiently. On the contrary, from inspection of the available pedigrees, it seems that the milder phenotype of patients with “hereditary" mutations has only limited effect on reproductive fitness. The fact that "hereditary" mutations are rarely observed in toxic adenomas is compatible with the suggestion that they would cause extremely slow tissue growth and, accordingly, would rarely cause thyrotoxicosis if somatic. There is, however, no a priori reason for neo-mutations to cause stronger gain-of-function than hereditary mutations. Accordingly, an activating mutation of the TSHR gene has been found in a six month child with subclinical hyperthyroidism presenting with weight loss as the initial symptom (41).

 

 Somatic mutations: autonomous toxic adenomas

 

  Soon after mutations of Gαs had been found in adenomas of the pituitary somatotrophs (42), similar mutations (also called Gsp mutations) were found in some toxic thyroid adenomas and follicular carcinomas (43-46). The mutated residues (Arg201, Glu227) are homologous to those found mutated in the ras proto-oncogenes: i.e. the mutations decrease the endogenous GTPase activity of the G protein, resulting in a constitutively active molecule. Toxic adenomas were found to be a fruitful source of somatic mutations activating the TSHR, probably because the phenotype is very conspicuous and easy to diagnose (47). Whereas mutations are distributed all along the serpentine portion of the receptor and even in the extracellular amino-terminal domain (48-54), there is clearly a hotspot at the cytoplasmic end of the sixth transmembrane segment (see Figure 1). The clustering reflects the pivotal role of this portion in the activation mechanism observed in the TSHR and in class A GPCRs generally [e.g. (55-61)].

Despite some dispute about the prevalence of TSHR mutations in toxic adenomas (which may be due to different origin of patients (62,63) or different sensitivity of the methodology) the current consensus is that activating mutations of the TSHR are the major cause of solitary toxic adenomas and account for about 60 to 80% of cases (15,7,64-66). Contrary to initial suggestions (63), the same percentage of activating TSHR mutations is observed in Japan, an iodine-sufficient country with low prevalence of toxic adenomas (67). In some patients with a multinodular goiter and two zones of autonomy at scintigraphy, a different mutation of the TSHR was identified in each nodule (68,36,69,70). This indicates that the pathophysiological mechanism responsible for solitary toxic adenomas can be at work on a background of multinodular goiter.

Table 1

 

CODONS Substitution

Somatic

mutation

Germline

neo- mutation

Germline

familial

Cancer

Stimulation

of basal cAMP

Stimulation of IP/Ca
Ser 281 Asn +       + -
    Thr +       + -
    Ile   +     + -
Asp 403 deletion +       + nd
Asn 406 Ser     +   + nd
Ser 425 Ile +       + -
Ala 428 Val   +     nd nd
Gly 431 Ser     +   + +
Met 453 Thr + +   + + -
Met 463 Val     +   + -
Ala 485 Val     +   + -
Ile 486 Phe +       + +
    Met +       + ±
    Asn   +     + -
Ser 505 Arg     +   + -
    Asn   +     + -
Val 509 Ala     +   + -
Leu 512 Arg +       + nd
    Gln +       + nd
Ile 568 Thr + +     + ±
    Phe +       + ±
Glu 575 Lys     +   + nd
Ala 593 Asn +       + nd
Val 597 Leu   +     + nd
    Phe         + nd
Y613-F631 deletion           -
Tyr 601 Asn +       + -
Asp 617 Tyr     +   + ±
Asp 619 Gly +       + -
Thr 620 Ile +     + + nd
Ala 623 Ile +       + ±
    Val +   +   + -
    Ser +     + + -
    Phe +       + nd
Met 626 Ile     +   + nd
Ala 627 Val +       + nd
Leu 629 Phe +   +   + -
Ile 630 Leu +       + -
Phe 631 Leu +       + -
    Cys + +     + -
    Ile       + + -
Thr 632 Ile + +     + -
Thr 632 Ala +     + + nd
Asp 633 Tyr +     + + -
    Glu +       + -
    His +     + + -
    Ala +       + -
Ile 635 Val +       + nd
Cys 636 Trp     +   + -
    Arg   +     + ±
Pro 639 Ser +   +   + +
Ile 640 Lys +       + nd
Asn 650 Tyr     +   + -
Val 656 Phe +       + -
Del 658-661   +         -
Asn 670 Ser     +   + -
Cys 672 Thr     +   + -
Leu 677 Val       + + nd

 

Table 1: Gain-of-function TSHR mutations. The nature of the mutations is indicated with their origins (somatic, germline sporadic, germline familial, cancer) and effects on intracellular regulatory cascades. nd - not determined; “-“ decreased functional property; “+” enhanced; “+/-“ similar to wild type.

 

In agreement with this notion, activating mutations of the TSHR have been identified in hyperfunctioning areas of multinodular goiter (70,19,65,23). The independent occurrence of two activating mutations in a patient may seem highly improbable at first. However, the multiplicity of the possible targets for activating mutations within the TSHR makes this less unlikely. It is also possible that a mutagenic environment is created in glands exposed to a chronic stimulation by TSH, resulting in H2O2 generation (71,72). Finally the involvement of TSHR mutations in thyroid cancers has been implicated in a limited number of follicular thyroid carcinoma (73-81).

 

Figure 1

Legend to figure 1: Structural model of the TSHR with interacting proteins and highlighted positions for gain-of-function mutations. Left: The model shows different parts of the receptor for which homologous structural information is available. The leucine-rich repeat domain (LRRD) and the hinge region are both harboring determinants for hormone (TSH model (surface) based on the FSH structure (82)) and antibody binding. The hinge region (colored pink) structurally links the LRRD with the serpentine domain made of transmembrane helices (H) 1-7 connected by intracellular (I1-3) and extracellular (E1-3) loops. Three cysteine bridges (yellow spheres) between the C-terminal LRRD and the C-terminus of the hinge region are indicated that are required for correct receptor arrangement and function. Wild type positions of constitutively activating mutations are indicated by side-chain representation (red sticks). Right: The known activating mutations (Table 1) are distributed over the entire serpentine portion of the receptor structure with clustering in the central core and specifically in helix 6. In contrast to other glycoprotein-hormone receptors (GPHRs), naturally occurring activating mutations were also identified in the extracellular loops and in the hinge region (Ser281).

 

Structure-function relationships of the TSHR

An important observation has been that the wild-type TSHR itself displays significant constitutive activity [(83,47) and review (84)]. This characteristic is not exceptional amongst GPCRs (e.g. (85)), but interestingly, it is not shared, at least to the same level, by its close relatives, the human luteinizing hormone/chorionic gonadotropin (LH/CG) receptor (LHCGR) and the human follitropin (FSH) receptor (FSHR). Compared to the TSHR, the LHCGR displays minimal basal activity and the human FSH receptor is totally silent (86). Together with the observation that mutations in residues distributed over most of the serpentine portion of the TSHR are equally effective in activating it (which does not seem to be a general characteristic in all GPCRs) this suggests that the unliganded TSHR might be less constrained than other GPCRs. As a consequence, being already “noisy” it would be more prone to further destabilization by a wide variety of mutations affecting multiple structural elements (Figure 1).

The effect of activating mutations must accordingly be interpreted in terms of “increase in constitutive activity”. Most constitutively active mutant receptors (also referred to as “CAMs”) found in toxic adenomas and/or toxic thyroid hyperplasia share common characteristics: i) they increase the constitutive activity of the receptor toward stimulation of adenylyl cyclase; ii) with a few notable exceptions (see Table 1 and below) (48), they do not display constitutive activity toward the inositol phosphate/diacylglycerol pathway; iii) their expression at the cell surface is decreased (from slightly to severely); iv) most, but not all of them keep responding to TSH for stimulation of cAMP and inositol phosphate generation, with a tendency to do so at decreased median effective concentrations; and v) they bind 125I-labeled bovine TSH with an apparent affinity higher than that of the wild-type receptor. Of note, CAMs with mutations at Ser281 (to Ile) (37) in the extracellular N-terminal part, at Ile486 (to Phe or Met) (48) and Ile568 (to Thr) (48) in the first and second extracellular loops, respectively, and at both Asp633 (to His) (7) and Pro639 (to Ser) (69) in transmembrane helix 6 are exceptional in that in addition to stimulating adenylyl cyclase, they cause constitutive activation of the inositol phosphate pathway. The constitutive activity of these mutants is interesting as it points to positions and structural fragments of the wild type receptor which may be of high relevance for its physiological coupling to both Gs and Gq (Figure 1 and Table 1).

No direct relationship is found between the level of cAMP achieved by different mutants in transfected COS cells and their level of expression at the cell membrane (87), which means that individual mutants have widely different “specific constitutive activity” (measured as the stimulation of cAMP accumulation/receptor number at the cell surface). Although this specific activity may tell us something about the mechanisms of receptor activation, it is not a measure of the actual phenotypic effect of the mutation in vivo. Indeed, one of the relatively mild mutations, observed up to now only in a family with HTTH (Cys672Tyr) (13), is among the strongest according to this criterion.

Differences between the effects of the mutants in transfected COS or HEK293 cells and thyrocytes in vivo render these correlations a difficult exercise. Indeed, most of the activating mutations of the TSHR have been studied by transient expression in COS or HEK293T cells and there is no guarantee that the mutants will function in an identical way in these artificial systems as they do in thyrocytes (88). In thyrocytes, a better relation has been observed between adenylylcyclase stimulation and differentiation than with growth (88). However, the built-in amplification associated with transfection of constructs in COS or HEK 293T cells has the advantage of allowing detection of even slight increases in constitutive activity of certain TSHR mutants.

 

According to a current model of GPCR activation, the receptor would exist under at least two interconverting conformations: R (silent conformations) and R* (active forms) (89,55,90,91) (Figure 2). The unliganded wild type receptor would shuttle between both forms, the equilibrium being in favor of R (90). Binding of the agonistic ligand is believed to stabilize the R* conformation.

 

Figure 2

Legend to figure 2: Left. Schematic representation of the equilibria between inactive (R) and active (R*) conformations of TSHR. The triangles indicate the equilibrium point of the wild type receptor (pink) and hypothetical mutants with increasing constitutive activity (brown, red). The situation of a receptor which would be devoid of basal activity is also indicated (blue triangle). Note that the wild type receptor (pink) has basal activity. Right. The concentration action curves corresponding to the hypothetical mutants and wild type receptors are indicated with the same color code.

 

The resulting R-to-R* transition was supposed to involve a conformational change that modifies the relative position of transmembrane helices to each other, which in turn would translate into conformational changes in the cytoplasmic crevice between the intracellular loops and transmembrane helices interacting with the hetero-trimeric G-protein. This model is strongly supported by solved crystal structures of active GPCR conformations (59,61) reviewed in (92)]. They reveal that receptor activation and signal transduction is characterized by specific movements of transmembrane helices (TMHs) 5, 6 and 7 leading to modification of their distances relative to each other. Helix 6 is a major player in this process, its cytoplasmic end moves away from that of TMH3 by turning around a pivotal helix-kink. The result is an “opening” of the cytoplasmic crevice of the receptor allowing interaction with the G protein (59). (A more detailed description of the activation mechanics, adapted to a model of the TSHR, is given in the legend to Figure 3.) This essential function of TMH6 for determination of an active state (59,61) might explain, why most activating TSHR mutations were found in this particular helix (Figure 1). This conclusion is in accordance with the early concept that the silent form of GPCRs would be submitted to structural constraints requiring the wild-type primary structure of the helix 6 and the connected third intracellular loop (93,90,91), and explains why these constraints could be released by a wide spectrum of amino acid substitutions in this segment as observed also for the TSHR (60).

 

Figure 3

Legend to figure 3: Model of the TSHR structure in complex with TSH and G-protein and illustration of the putative activation mechanism. The receptor is displayed as a backbone cartoon in complex with the hetero-dimeric hormone and a hetero-trimeric G-protein molecule (surface representations). For the serpentine portion of the receptor, the model is based on the solved structure of the β2-adrenergic/Gs crystal (61). The ectodomain (in complex with TSH) and the hinge region were modeled based on a determined and homologous FSHR-FSH structure (82). A selection of residues affected by known activating mutations are shown as red sticks and identified by their position in the primary structure of the protein (see Figure 1). Their positions tentatively illustrate the “path” followed by the activation signal, from outside the membrane (in the ectodomain) to the cytoplasmic surface of the receptor, via the transmembrane helices. Briefly, the hormone binds to both the Leucine rich repeat domain (LRRD) and the hinge region (e.g. sulfated tyrosine 385 (sTyr)). This initial signal is transduced into a conformational change of a module (intramolecular agonist) constituted by the “hinge” region of the ectodomain and the exoloops (E1-3) of the serpentine domain. In favor of this model, several residues belonging to this module (Ser281 in the ectodomain; Asp403 and Asn406 at the ectodomain-serpentine domain border; Ile486, Ile568, Val656 in the exoloops) can activate the receptor constitutively when mutated. Together with the observation that a truncated receptor devoid of ectodomain displays significant increase in constitutive activity, this suggests that activation of the TSHR involves switching of specific extracellular portions from a tethered inverse agonist (maintenance of the basal state) to an intramolecular agonist (94). The resulting structural changes affecting the exoloops are expected to be directly conveyed to the transmembrane helices with the resulting breakage of silencing locks (arrows). From comparison of the inactive and active rhodopsin (59) or beta-2 adrenoreceptor structures (61), the largest spatial movement affects TMH6, involving a combination of horizontal and rotational (wound arrow) movements around a pivotal helix-kink at a proline (TSHR Pro639). These global changes result in the partial “opening” of the intra-helical crevice on the cytoplasmic side of the receptor (horizontal double-head arrows), allowing complete binding and activation of G-proteins.

 

In addition to the release of structural locks stabilizing the inactive conformation of GPCRs, activation of the GPHRs has been shown to involve a triggering mechanism exerted by an “intramolecular agonist” constituted by segments of the exoloops and the C-terminal portion of their ectodomain (94-96). According to this model, it is this module, activated by the binding of TSH, thyroid stimulating auto-antibodies, thyrostimulin (97,98) or mutations (see below) which would be the immediate agonist of the serpentine portion of the receptor (94,95,99,96,100,101). In all cases, however, mutations are expected to affect the local three dimensional structure of the receptor with a resulting global effect on its activation state. Amongst these are modification of “knob and hole” interactions (e.g. by repulsion) in tightly packed local microdomains and breakage, or creation of intramolecular interactions by changing the biophysical characteristics of side chains (e.g. (6)). As exclusive examples of these, mutations at Asp633 (57,102) or Asp619 (47) are expected to break interhelical locks between transmembrane helices 6 and 7 or 3, respectively. Interestingly, even mutations affecting an important residue of the trigger in the ectodomain (Ser281) seem also to be responsible for a “loss-of-local structure”. Indeed, substitution of the wild type residue (serine) by almost any amino acid results in constitutive activation (103,104). This implicates that predictions of phenotype-genotype relationships must always be considered with much caution if they are not backed by detailed structural and functional knowledge.

 

Familial gestational hyperthyroidism

Some degree of stimulation of the thyroid gland by human chorionic gonadotrophin (hCG) is commonly observed during early pregnancy. It is usually responsible for decrease in serum thyrotropin with an increase in free thyroxine concentrations that remains within the normal range (for references see (105)). When the concentrations of hCG are abnormally high, like in molar pregnancy, true hyperthyroidism may ensue. The pathophysiological mechanism is believed to be the promiscuous stimulation of the TSHR by excess hCG, as suggested by the rough direct or inverse relation between serum hCG and free T4 or TSH concentrations, respectively (106,105). A convincing rationale is provided by the close structural relationships of the glycoprotein hormones and their receptors, respectively (107).

A new syndrome has been described in 1998 in a family with dominant transmission of hyperthyroidism limited to pregnancy (Figure 4) (108). The proposita and her mother had severe thyrotoxicosis together with hyperemesis gravidarum during the course of each of their pregnancies. When non pregnant they were clinically and biologically euthyroid. Both patients were heterozygous for a K183R mutation in the extracellular amino-terminal domain (Figure 3) of the TSHR gene. When tested by transient transfection in COS cells, the mutant receptor displayed normal characteristics towards TSH. However, providing a convincing explanation to the phenotype, it showed higher sensitivity to stimulation by hCG, when compared with wild type TSHR (108).

The amino acid substitution responsible for the promiscuous stimulation of the TSHR by hCG is surprisingly conservative. Also surprising is the observation that residue 183 is a lysine in both the TSH and LH/CG receptors. When placed on the available three dimensional model of the hormone-binding domain of the TSHR (109), residue 183 belongs to one of the beta-sheets which constitute the putative surface of interaction with the hormones (Figure 3).

 

Figure 4

Legend to figure 4: Familial gestational hyperthyroidism secondary to mutation of the TSHR gene. Upper panel displays the pedigree, with the two ladies affected, together with a “snake plot “of the TSHR, with the mutation indicated. Lower right panel illustrates the increased sensitivity of the K183R mutant TSHR vis-à-vis hCG. Binding of TSH, or hCG to the ectodomain of the TSHR, according to models based on crystallographic data from (110,82) is visualized in figure 3.

Detailed analysis of the effect of the K183R mutation by site-directed mutagenesis indicated that any amino acid substitution at this position confers a slight increase in stability to the illegitimate hCG/TSHR complex (111). This increase in stability would be enough to cause signal transduction by the hCG concentrations achieved in pregnancy, but not by the LH concentrations observed after menopause. Indeed, the mother of the proposita remained euthyroid after menopause. This finding is compatible with a relatively modest gain-of-function of the K183R mutant upon stimulation by hCG. A second family with the same phenotype has recently been identified. Interestingly, the mutation affects the same residue (K183N) (112).

 

Contrary to other mammals, human and primates rely on chorionic gonadotropin for maintenance of corpus luteum in early pregnancy (113). The frequent partial suppression of TSH observed at peak hCG levels during normal pregnancy indicates that evolution has selected physiological mechanisms operating very close to the border of thyrotoxicosis. This may provide a rationale to the observation that, in comparison to other species, the glycoprotein hormones of primates display a lower biological activity due to positive selection by evolution of specific amino acid substitutions in their alpha-subunits (114). Up to now no spontaneous mutation has been identified which would increase the bioactivity of hCG. An interesting parallel may be drawn between familial gestational hyperthyroidism and cases of spontaneous ovarian hyperstimulation syndrome (sOHSS) (86,115). In sOHSS, mutations of the FSH receptor gene render the receptor abnormally sensitive to hCG. The result is recurrent hyperstimulation of the ovary, on the occasion of each pregnancy.

 

LOSS OF FUNCTION MUTATIONS

Loss-of-function mutations in the TSHR gene are expected to cause a syndrome of “resistance to TSH.” The expected phenotype is likely to resemble that of patients with mutations in TSH itself. These mutations have been described early because of the prior availability of information on TSH alpha and beta genes (114). Mouse models of resistance to TSH are available as natural (hyt/hyt mouse) (116) or experimental TSHR mutant lines (117,118). Interestingly, and contrary to the situation in human (see below), the thyroid of newborn TSHR knockout mice is of normal size. As expected, the homozygote animals displayed profound hypothyroidism. Their thyroids do not express the sodium-iodide symporter, but showed significant (non-iodinated) thyroglobulin production. From this information one would expect patients with two TSHR mutated alleles to exhibit a degree of hypothyroidism in accordance with the extent of the loss-of-function, going from mild, compensated, hypothyroidism, to profound neonatal hypothyroidism with absent iodide trapping. Heterozygous carriers are expected to be normal or display minimal increase in plasma TSH.

 

Clinical cases with the mutations identified

A few patients with convincing resistance to TSH had been described before molecular genetics permitted identification of the mutations (119,120). The first cases described in molecular terms were euthyroid siblings with elevated TSH (121). Sequencing of the TSHR gene identified a different mutation in each allele of the affected individuals, which made them compound heterozygotes. The substitutions were in the extracellular amino-terminal portion of the receptor (maternal allele, P162A; paternal allele, I167N). The functional characteristics of the mutant receptors showed that the paternal allele was virtually completely non-functional, whereas the maternal allele displayed an increase in the median effective TSH concentration for stimulation of cAMP production. Current knowledge of the structure of part of the ectodomain of the receptor allows to establish structure-function relationships for mutations affecting this portion of the receptor (122,109,123-126,101).

A large number of familial cases with loss-of-function mutations of the TSHR have been identified in the course of screening programs for congenital hypothyroidism (127-140) [(Figure 5, For a complete list of naturally occurring and side-directed TSHR single amino acid substitutions with their functional characteristics see the GPHR information resource “SSFA” available under: http://www.ssfa-gphr.de (29,30)]. Some of the patients displayed the usual criteria for congenital hypothyroidism, including high TSH, low free T4, and undetectable trapping of 99Tc. In some cases, plasma thyroglobulin levels were normal or high. The patients can be compound heterozygotes for complete loss of function mutations (129), or homozygotes, born to consanguineous (128) or apparently unrelated parents (134).

 

Figure 5

Legend to figure 5: Structural model of the TSHR with indication of loss-of-function mutations. The location and substitutions responsible for known loss-of-function mutations (side-chains as blue sticks) are indicated on a three-dimensional receptor model. Single letter abbreviations of amino-acids are used. In contrast to activating mutations, many inactivating mutations are located also in the LRRD. As observable in this complex model inactivating mutations can have different molecular effects on TSHR functions dependent on their localization, like diminishing hormone binding (location in the LRRD, e.g. R109Q), G-protein binding (intracellular localization, e.g. M527T, R531W), leading to a decreased receptor cell surface expression by modification of the three-dimensional structure (e.g. mutations at extracellular cysteines which interrupts stabilizing disulfide bridges, e.g. C390W), or interrupting the signal transport in the serpentine domain (located at the helices, e.g. A593V).

 

In agreement with the phenotype of knock-out mice with homozygous invalidation of the TSHR, patients with complete loss-of-function of the receptor display an in-place, thyroid with completely absent iodide or 99Tc trapping. However, in contrast with the situation in mice, the gland is hypoplastic. Activation of the cAMP pathway, while dispensable for the anatomical development of the gland and thyroglobulin production, is thus absolutely required for expression of the NIS gene and, at least in human, for normal growth of the tissue during fetal life. This explains that in the absence of thyroglobulin measurements or expert echography, loss-of-function mutations of the TSHR may easily be misdiagnosed as thyroid agenesis. In the heterozygous state, complete loss of function mutations of the TSHR is a cause of moderate hyperthyrotropinemia (subclinical hypothyroidism), segregating as an autosomal dominant trait (141).

 

Resistance to thyrotropin not linked to the TSHR gene

Finally, it must be stressed that an autosomal dominant form of partial resistance to TSH has been demonstrated in families in which linkage to the TSHR gene has been excluded (142). A locus has been identified on chromosome 15q25.3-26.1 but the gene responsible for the phenotype has not been identified yet (143).

 

Polymorphisms

A series of single nucleotide polymorphisms affecting the coding sequence have been identified in the TSHR gene. After the initial suggestion that some of these (D36H, P52T, D727E) would be associated with susceptibility to autoimmune thyroid diseases (144-146) the current consensus is that they represent neutral alleles with no pathophysiological significance (147-150). However, a genome-wide study involving a large cohort of patients has recently demonstrated association between non-coding SNPs at the TSHR gene locus and Graves’disease (151,152). The genetic substratum responsible for this association is still under study (152). However, a large meta-analysis of genome wide association studies failed to identify the TSHR as a locus affecting plasma TSH values (153).

One polymorphic residue deserves special mention: position 601 was found to be a tyrosine or a histidine in the two initial reports of TSHR cloning (154,155). Characterization of the two alleles by transfection in COS cells indicated interesting functional differences: the Tyr601 allele displayed readily detectable constitutive activity, whereas the His601 was completely silent; the Tyr601 allele responded to stimulation by TSH by activating both the adenylylcyclase and phospholipase C dependent regulatory cascades, when the His601 allele was only active on the cAMP pathway (156,157). The Tyr601 allele is by far the most frequent in all populations tested. A Tyr601Asn mutation was found in a toxic adenoma. Characterization of the mutant demonstrated increase in constitutive activation of the cAMP regulatory cascade (157), making the 601 residue an interesting target for structure-function studies.

Acknowledgments

Research in the laboratory of the author was supported by the Interuniversity Attraction Poles Programme-Belgian State-Belgian Science Policy (6/14), the Fonds de la Recherche Scientifique Médicale of Belgium, the Walloon Region (program “Cibles”) and the non-for-profit Association Recherche Biomédicale et Diagnostic. This work was supported by the Deutsche Forschungsgemeinschaft (DFG), projects KL2334/2-2 and Cluster of Excellence ‘Unifying Concepts in Catalysis’ (Research Field D3/E3-1) to G.Kl..

 

REFERENCES

1. Ledent C, Dumont JE, Vassart G, et al. Thyroid expression of an A2 adenosine receptor transgene induces thyroid hyperplasia and hyperthyroidism. The EMBO journal 1992; 11:537-542
2. Maenhaut C, Van Sande J, Libert F, et al. RDC8 codes for an adenosine A2 receptor with physiological constitutive activity. Biochemical and biophysical research communications 1990; 173:1169-1178
3. Weinstein LS, Shenker A, Gejman PV, et al. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. The New England journal of medicine 1991; 325:1688-1695
4. Michiels FM, Caillou B, Talbot M, et al. Oncogenic potential of guanine nucleotide stimulatory factor alpha subunit in thyroid glands of transgenic mice. Proceedings of the National Academy of Sciences of the United States of America 1994; 91:10488-10492
5. Kero J, Ahmed K, Wettschureck N, et al. Thyrocyte-specific Gq/G11 deficiency impairs thyroid function and prevents goiter development. The Journal of clinical investigation 2007; 117:2399-2407

6. Winkler F, Kleinau G, Tarnow P, et al. A new phenotype of nongoitrous and nonautoimmune hyperthyroidism caused by a heterozygous thyrotropin receptor mutation in transmembrane helix 6. The Journal of clinical endocrinology and metabolism 2010; 95:3605-3610
7. Parma J, Duprez L, Van Sande J, et al. Diversity and prevalence of somatic mutations in the thyrotropin receptor and Gs alpha genes as a cause of toxic thyroid adenomas. The Journal of clinical endocrinology and metabolism 1997; 82:2695-2701
8. Corvilain B, Van Sande J, Dumont JE, et al. Somatic and germline mutations of the TSH receptor and thyroid diseases. Clinical endocrinology 2001; 55:143-158
9. Chanson P, Salenave S, Orcel P. McCune-Albright syndrome in adulthood. Pediatric endocrinology reviews : PER 2007; 4 Suppl 4:453-462
10. Rapoport B, Chazenbalk GD, Jaume JC, et al. The thyrotropin (TSH) receptor: interaction with TSH and autoantibodies. Endocr Rev 1998; 19:673-716

11. Davies TF, Ando T, Lin RY, et al. Thyrotropin receptor-associated diseases: from adenomata to Graves disease. The Journal of clinical investigation 2005; 115:1972-1983
12. Thomas JS, Leclere J, Hartemann P, et al. Familial hyperthyroidism without evidence of autoimmunity. Acta endocrinologica 1982; 100:512-518
13. Duprez L, Parma J, Van Sande J, et al. Germline mutations in the thyrotropin receptor gene cause non-autoimmune autosomal dominant hyperthyroidism. Nature genetics 1994; 7:396-401
14. Tonacchera M, Van Sande J, Cetani F, et al. Functional characteristics of three new germline mutations of the thyrotropin receptor gene causing autosomal dominant toxic thyroid hyperplasia. The Journal of clinical endocrinology and metabolism 1996; 81:547-554
15. Fuhrer D, Holzapfel HP, Wonerow P, et al. Somatic mutations in the thyrotropin receptor gene and not in the Gs alpha protein gene in 31 toxic thyroid nodules. The Journal of clinical endocrinology and metabolism 1997; 82:3885-3891
16. Esapa CT, Duprez L, Ludgate M, et al. A novel thyrotropin receptor mutation in an infant with severe thyrotoxicosis. Thyroid 1999; 9:1005-1010
17. Khoo DH, Parma J, Rajasoorya C, et al. A germline mutation of the thyrotropin receptor gene associated with thyrotoxicosis and mitral valve prolapse in a Chinese family. JClinEndocrinolMetab 1999; 84:1459-1462
18. Fuhrer D, Warner J, Sequeira M, et al. Novel TSHR germline mutation (Met463Val) masquerading as Graves' disease in a large Welsh kindred with hyperthyroidism. Thyroid 2000; 10:1035-1041
19. Tonacchera M, Agretti P, Rosellini V, et al. Sporadic nonautoimmune congenital hyperthyroidism due to a strong activating mutation of the thyrotropin receptor gene. Thyroid 2000; 10:859-863
20. Alberti L, Proverbio MC, Costagliola S, et al. A novel germline mutation in the TSH receptor gene causes non-autoimmune autosomal dominant hyperthyroidism. EurJEndocrinol 2001; 145:249-254
21. Biebermann H, Schoneberg T, Hess C, et al. The first activating TSH receptor mutation in transmembrane domain 1 identified in a family with nonautoimmune hyperthyroidism. The Journal of clinical endocrinology and metabolism 2001; 86:4429-4433
22. Claus M, Maier J, Paschke R, et al. Novel thyrotropin receptor germline mutation (Ile568Val) in a Saxonian family with hereditary nonautoimmune hyperthyroidism. Thyroid 2005; 15:1089-1094
23. Gozu H, Avsar M, Bircan R, et al. Two novel mutations in the sixth transmembrane segment of the thyrotropin receptor gene causing hyperfunctioning thyroid nodules. Thyroid 2005; 15:389-397
24. Nwosu BU, Gourgiotis L, Gershengorn MC, et al. A novel activating mutation in transmembrane helix 6 of the thyrotropin receptor as cause of 25.Nishihara E, Nagayama Y, Amino N, et al. A novel thyrotropin receptor germline mutation (Asp617Tyr) causing hereditary hyperthyroidism. EndocrJ 2007; 54:927-934
26. Akcurin S, Turkkahraman D, Tysoe C, et al. A family with a novel TSH receptor activating germline mutation (p.Ala485Val). Eur J Pediatr 2008; 167:1231-1237
27. Liu Z, Sun Y, Dong Q, et al. A novel TSHR gene mutation (Ile691Phe) in a Chinese family causing autosomal dominant non-autoimmune hyperthyroidism. JHumGenet 2008; 53:475-478
28. Nishihara E, Chen CR, Higashiyama T, et al. Subclinical nonautoimmune hyperthyroidism in a family segregates with a thyrotropin receptor mutation with weakly increased constitutive activity. Thyroid 2010; 20:1307-1314
29. Kleinau G, Brehm M, Wiedemann U, et al. Implications for molecular mechanisms of glycoprotein hormone receptors using a new sequence-structure-function analysis resource. Mol Endocrinol 2007; 21:574-580
30. Kreuchwig A, Kleinau G, Kreuchwig F, et al. Research resource: Update and extension of a glycoprotein hormone receptors web application. Mol Endocrinol 2011; 25:707-712
31. Bircan R, Miehle K, Mladenova G, et al. Multiple relapses of hyperthyroidism after thyroid surgeries in a patient with long term follow-up of sporadic non-autoimmune hyperthyroidism. Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association 2008; 116:341-346
32. Leclere J, Bene MC, Duprez A, et al. Behaviour of thyroid tissue from patients with Graves' disease in nude mice. The Journal of clinical endocrinology and metabolism 1984; 59:175-177
33. Hebrant A, van Staveren WC, Maenhaut C, et al. Genetic hyperthyroidism: hyperthyroidism due to activating TSHR mutations. European journal of endocrinology 2011; 164:1-9
34. Lavard L, Jacobsen BB, Perrild H, et al. Prevalence of germline mutations in the TSH receptor gene as a cause of juvenile thyrotoxicosis. Acta paediatrica 2004; 93:1192-1194
35. Kopp P, van Sande J, Parma J, et al. Brief report: congenital hyperthyroidism caused by a mutation in the thyrotropin-receptor gene. The New England journal of medicine 1995; 332:150-154
36. Holzapfel HP, Wonerow P, von Petrykowski W, et al. Sporadic congenital hyperthyroidism due to a spontaneous germline mutation in the thyrotropin receptor gene. JClinEndocrinolMetab 1997; 82:3879-3884
37. Kopp P, Muirhead S, Jourdain N, et al. Congenital hyperthyroidism caused by a solitary toxic adenoma harboring a novel somatic mutation (serine281-->isoleucine) in the extracellular domain of the thyrotropin receptor. The Journal of clinical investigation 1997; 100:1634-1639
38. Gruters A, Schoneberg T, Biebermann H, et al. Severe congenital hyperthyroidism caused by a germ-line neo mutation in the extracellular portion of the thyrotropin receptor. The Journal of clinical endocrinology and metabolism 1998; 83:1431-1436
39. Karges B, Krause G, Homoki J, et al. TSH receptor mutation V509A causes familial hyperthyroidism by release of interhelical constraints between transmembrane helices TMH3 and TMH5. JEndocrinol 2005; 186:377-385
40. Gozu HI, Lublinghoff J, Bircan R, et al. Genetics and phenomics of inherited and sporadic non-autoimmune hyperthyroidism. Molecular and cellular endocrinology 2010; 322:125-134
41. Pohlenz J, Pfarr N, Kruger S, et al. Subclinical hyperthyroidism due to a thyrotropin receptor (TSHR) gene mutation (S505R). Acta paediatrica 2006; 95:1685-1687
42. Landis CA, Masters SB, Spada A, et al. GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 1989; 340:692-696
43. Lyons J, Landis CA, Harsh G, et al. Two G protein oncogenes in human endocrine tumors. Science 1990; 249:655-659
44. O’Sullivan C, Barton CM, Staddon SL, et al. Activating point mutations of the gsp oncogene in human thyroid adenomas. Molecular carcinogenesis 1991; 4:345-349
45. Suarez HG, du Villard JA, Caillou B, et al. gsp mutations in human thyroid tumours. Oncogene 1991; 6:677-679
46. Goretzki PE, Lyons J, Stacy-Phipps S, et al. Mutational activation of RAS and GSP oncogenes in differentiated thyroid cancer and their biological implications. World journal of surgery 1992; 16:576-581; discussion 581-572
47. Parma J, Duprez L, Van Sande J, et al. Somatic mutations in the thyrotropin receptor gene cause hyperfunctioning thyroid adenomas. Nature 1993; 365:649-651
48. Parma J, Van Sande J, Swillens S, et al. Somatic mutations causing constitutive activity of the thyrotropin receptor are the major cause of hyperfunctioning thyroid adenomas: identification of additional mutations activating both the cyclic adenosine 3',5'-monophosphate and inositol phosphate-Ca2+ cascades. Mol Endocrinol 1995; 9:725-733
49. Duprez L, Parma J, Costagliola S, et al. Constitutive activation of the TSH receptor by spontaneous mutations affecting the N-terminal extracellular domain. FEBS letters 1997; 409:469-474
50. Fuhrer D, Wonerow P, Willgerodt H, et al. Identification of a new thyrotropin receptor germline mutation (Leu629Phe) in a family with neonatal onset of autosomal dominant nonautoimmune hyperthyroidism. JClinEndocrinolMetab 1997; 82:4234-4238
51. Holzapfel HP, Fuhrer D, Wonerow P, et al. Identification of constitutively activating somatic thyrotropin receptor mutations in a subset of toxic multinodular goiters. The Journal of clinical endocrinology and metabolism 1997; 82:4229-4233
52. Parma J, Duprez L, Van Sande J, et al. Diversity and prevalence of somatic mutations in the thyrotropin receptor and Gs alpha genes as a cause of toxic thyroid adenomas. JClinEndocrinolMetab 1997; 82:2695-2701
53. Wonerow P, Chey S, Fuhrer D, et al. Functional characterization of five constitutively activating thyrotrophin receptor mutations. Clinical endocrinology 2000; 53:461-468
54. Castro I, Lima L, Seoane R, et al. Identification and functional characterization of two novel activating thyrotropin receptor mutants in toxic thyroid follicular adenomas. Thyroid 2009; 19:645-649
55. Gether U. Uncovering molecular mechanisms involved in activation of G protein-coupled receptors. Endocr Rev 2000; 21:90-113
56. Govaerts C, Lefort A, Costagliola S, et al. A conserved Asn in transmembrane helix 7 is an on/off switch in the activation of the thyrotropin receptor. J Biol Chem 2001; 276:22991-22999
57. Neumann S, Krause G, Chey S, et al. A free carboxylate oxygen in the side chain of position 674 in transmembrane domain 7 is necessary for TSH receptor activation. Mol Endocrinol 2001; 15:1294-1305
58. Kleinau G, Claus M, Jaeschke H, et al. Contacts between extracellular loop two and transmembrane helix six determine basal activity of the thyroid-stimulating hormone receptor. J Biol Chem 2007; 282:518-525
59. Scheerer P, Park JH, Hildebrand PW, et al. Crystal structure of opsin in its G-protein-interacting conformation. Nature 2008; 455:497-502
60. Kleinau G, Hoyer I, Kreuchwig A, et al. From Molecular Details of the Interplay between Transmembrane Helices of the Thyrotropin Receptor to General Aspects of Signal Transduction in Family A G-protein-coupled Receptors (GPCRs). J Biol Chem 2011; 286:25859-25871
61. Rasmussen SG, DeVree BT, Zou Y, et al. Crystal structure of the beta2 adrenergic receptor-Gs protein complex. Nature 2011; 477:549-555
62. Russo D, Arturi F, Wicker R, et al. Genetic alterations in thyroid hyperfunctioning adenomas. The Journal of clinical endocrinology and metabolism 1995; 80:1347-1351

63. Takeshita A, Nagayama Y, Yokoyama N, et al. Rarity of oncogenic mutations in the thyrotropin receptor of autonomously functioning thyroid nodules in Japan. The Journal of clinical endocrinology and metabolism 1995; 80:2607-2611
64. Trulzsch B, Krohn K, Wonerow P, et al. Detection of thyroid-stimulating hormone receptor and Gsalpha mutations: in 75 toxic thyroid nodules by denaturing gradient gel electrophoresis. Journal of molecular medicine 2001; 78:684-691
65. Georgopoulos NA, Sykiotis GP, Sgourou A, et al. Autonomously functioning thyroid nodules in a former iodine-deficient area commonly harbor gain-of-function mutations in the thyrotropin signaling pathway. European journal of endocrinology 2003; 149:287-292
66. Palos-Paz F, Perez-Guerra O, Cameselle-Teijeiro J, et al. Prevalence of mutations in TSHR, GNAS, PRKAR1A and RAS genes in a large series of toxic thyroid adenomas from Galicia, an iodine-deficient area in NW Spain. European journal of endocrinology 2008; 159:623-631
67. Vanvooren V, Uchino S, Duprez L, et al. Oncogenic mutations in the thyrotropin receptor of autonomously functioning thyroid nodules in the Japanese population. European journal of endocrinology 2002; 147:287-291
68. Duprez L, Hermans J, Van Sande J, et al. Two autonomous nodules of a patient with multinodular goiter harbor different activating mutations of the thyrotropin receptor gene. JClinEndocrinolMetab 1997; 82:306-308
69. Tonacchera M, Chiovato L, Pinchera A, et al. Hyperfunctioning thyroid nodules in toxic multinodular goiter share activating thyrotropin receptor mutations with solitary toxic adenoma. The Journal of clinical endocrinology and metabolism 1998; 83:492-498
70. Tonacchera M, Vitti P, Agretti P, et al. Activating thyrotropin receptor mutations in histologically heterogeneous hyperfunctioning nodules of multinodular goiter. Thyroid 1998; 8:559-564
71. Krohn K, Fuhrer D, Bayer Y, et al. Molecular pathogenesis of euthyroid and toxic multinodular goiter. Endocr Rev 2005; 26:504-524
72. Maier J, van Steeg H, van Oostrom C, et al. Deoxyribonucleic acid damage and spontaneous mutagenesis in the thyroid gland of rats and mice. Endocrinology 2006; 147:3391-3397
73. Russo D, Arturi F, Schlumberger M, et al. Activating mutations of the TSH receptor in differentiated thyroid carcinomas. Oncogene 1995; 11:1907-1911
74. Spambalg D, Sharifi N, Elisei R, et al. Structural studies of the thyrotropin receptor and Gs alpha in human thyroid cancers: low prevalence of mutations predicts infrequent involvement in malignant transformation. The Journal of clinical endocrinology and metabolism 1996; 81:3898-3901
75. Camacho P, Gordon D, Chiefari E, et al. A Phe 486 thyrotropin receptor mutation in an autonomously functioning follicular carcinoma that was causing hyperthyroidism. Thyroid 2000; 10:1009-1012
76. Mircescu H, Parma J, Huot C, et al. Hyperfunctioning malignant thyroid nodule in an 11-year-old girl: pathologic and molecular studies. The Journal of pediatrics 2000; 137:585-587
77. Fuhrer D, Tannapfel A, Sabri O, et al. Two somatic TSH receptor mutations in a patient with toxic metastasising follicular thyroid carcinoma and non-functional lung metastases. Endocrine-related cancer 2003; 10:591-600
78. Niepomniszcze H, Suarez H, Pitoia F, et al. Follicular carcinoma presenting as autonomous functioning thyroid nodule and containing an activating mutation of the TSH receptor (T620I) and a mutation of the Ki-RAS (G12C) genes. Thyroid 2006; 16:497-503
79. Garcia-Jimenez C, Santisteban P. TSH signalling and cancer. Arquivos brasileiros de endocrinologia e metabologia 2007; 51:654-671
80. Nikiforova MN, Wald AI, Roy S, et al. Targeted next-generation sequencing panel (ThyroSeq) for detection of mutations in thyroid cancer. The Journal of clinical endocrinology and metabolism 2013; 98:E1852-1860
81. O'Hayre M, Vazquez-Prado J, Kufareva I, et al. The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer. Nature reviews Cancer 2013; 13:412-424
82. Jiang X, Liu H, Chen X, et al. Structure of follicle-stimulating hormone in complex with the entire ectodomain of its receptor. Proceedings of the National Academy of Sciences of the United States of America 2012; 109:12491-12496
83. Kosugi S, Okajima F, Ban T, et al. Mutation of alanine 623 in the third cytoplasmic loop of the rat thyrotropin (TSH) receptor results in a loss in the phosphoinositide but not cAMP signal induced by TSH and receptor autoantibodies. J Biol Chem 1992; 267:24153-24156
84. Kleinau G, Biebermann H. Constitutive activities in the thyrotropin receptor: regulation and significance. Advances in pharmacology 2014; 70:81-119
85. Berg KA, Harvey JA, Spampinato U, et al. Physiological relevance of constitutive activity of 5-HT2A and 5-HT2C receptors. Trends in pharmacological sciences 2005; 26:625-630
86. Smits G, Olatunbosun O, Delbaere A, et al. Ovarian hyperstimulation syndrome due to a mutation in the follicle-stimulating hormone receptor. The New England journal of medicine 2003; 349:760-766
87. Van Sande J, Parma J, Tonacchera M, et al. Somatic and germline mutations of the TSH receptor gene in thyroid diseases. The Journal of clinical endocrinology and metabolism 1995; 80:2577-2585
88. Fuhrer D, Lewis MD, Alkhafaji F, et al. Biological activity of activating thyroid-stimulating hormone receptor mutants depends on the cellular context. Endocrinology 2003; 144:4018-4030
89. De Lean A, Stadel JM, Lefkowitz RJ. A ternary complex model explains the agonist-specific binding properties of the adenylate cyclase-coupled beta-adrenergic receptor. J Biol Chem 1980; 255:7108-7117
90. Kenakin T. Principles: receptor theory in pharmacology. Trends in pharmacological sciences 2004; 25:186-192
91. Rosenbaum DM, Rasmussen SG, Kobilka BK. The structure and function of G-protein-coupled receptors. Nature 2009; 459:356-363
92. Lebon G, Warne T, Tate CG. Agonist-bound structures of G protein-coupled receptors. Current opinion in structural biology 2012; 22:482-490
93. Lefkowitz RJ, Cotecchia S, Samama P, et al. Constitutive activity of receptors coupled to guanine nucleotide regulatory proteins. Trends in pharmacological sciences 1993; 14:303-307
94. Vlaeminck-Guillem V, Ho SC, Rodien P, et al. Activation of the cAMP pathway by the TSH receptor involves switching of the ectodomain from a tethered inverse agonist to an agonist. Mol Endocrinol 2002; 16:736-746
95. Kleinau G, Jaschke H, Neumann S, et al. Identification of a novel epitope in the thyroid-stimulating hormone receptor ectodomain acting as intramolecular signaling interface. J Biol Chem 2004; 279:51590-51600
96. Bruser A, Schulz A, Rothemund S, et al. The Activation Mechanism of Glycoprotein Hormone Receptors with Implications in the Cause and Therapy of Endocrine Diseases. J Biol Chem 2016; 291:508-520
97. Nakabayashi K, Matsumi H, Bhalla A, et al. Thyrostimulin, a heterodimer of two new human glycoprotein hormone subunits, activates the thyroid-stimulating hormone receptor. The Journal of clinical investigation 2002; 109:1445-1452
98. Karponis D, Ananth S. The role of thyrostimulin and its potential clinical significance. Endocrine regulations 2017; 51:117-128
99. Vassart G, Pardo L, Costagliola S. A molecular dissection of the glycoprotein hormone receptors. Trends in biochemical sciences 2004; 29:119-126
100. Schoneberg T, Kleinau G, Bruser A. What are they waiting for?-Tethered agonism in G protein-coupled receptors. Pharmacological research 2016; 108:9-15
101. Kleinau G, Worth CL, Kreuchwig A, et al. Structural-Functional Features of the Thyrotropin Receptor: A Class A G-Protein-Coupled Receptor at Work. Frontiers in endocrinology 2017; 8:86
102. Claeysen S, Govaerts C, Lefort A, et al. A conserved Asn in TM7 of the thyrotropin receptor is a common requirement for activation by both mutations and its natural agonist. FEBS letters 2002; 517:195-200
103. Ho SC, Van Sande J, Lefort A, et al. Effects of mutations involving the highly conserved S281HCC motif in the extracellular domain of the thyrotropin (TSH) receptor on TSH binding and constitutive activity. Endocrinology 2001; 142:2760-2767
104. Jaeschke H, Neumann S, Kleinau G, et al. An aromatic environment in the vicinity of serine 281 is a structural requirement for thyrotropin receptor function. Endocrinology 2006; 147:1753-1760
105. Glinoer D, Spencer CA. Serum TSH determinations in pregnancy: how, when and why? Nature reviews Endocrinology 2010; 6:526-529
106. Glinoer D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocr Rev 1997; 18:404-433
107. Grossmann M, Weintraub BD, Szkudlinski MW. Novel insights into the molecular mechanisms of human thyrotropin action: structural, physiological, and therapeutic implications for the glycoprotein hormone family. Endocr Rev 1997; 18:476-501
108. Rodien P, Bremont C, Sanson ML, et al. Familial gestational hyperthyroidism caused by a mutant thyrotropin receptor hypersensitive to human chorionic gonadotropin. The New England journal of medicine 1998; 339:1823-1826
109. Sanders J, Chirgadze DY, Sanders P, et al. Crystal structure of the TSH receptor in complex with a thyroid-stimulating autoantibody. Thyroid 2007; 17:395-410
110. Fan QR, Hendrickson WA. Structure of human follicle-stimulating hormone in complex with its receptor. Nature 2005; 433:269-277
111. Smits G, Govaerts C, Nubourgh I, et al. Lysine 183 and glutamic acid 157 of the TSH receptor: two interacting residues with a key role in determining specificity toward TSH and human CG. Mol Endocrinol 2002; 16:722-735
112. Coulon AL, Savagner F, Briet C, et al. Prolonged and Severe Gestational Thyrotoxicosis Due to Enhanced hCG Sensitivity of a Mutant Thyrotropin Receptor. The Journal of clinical endocrinology and metabolism 2016; 101:10-11
113. Stewart HJ, Jones DS, Pascall JC, et al. The contribution of recombinant DNA techniques to reproductive biology. Journal of reproduction and fertility 1988; 83:1-57
114. Szkudlinski MW, Fremont V, Ronin C, et al. Thyroid-stimulating hormone and thyroid-stimulating hormone receptor structure-function relationships. Physiological reviews 2002; 82:473-502
115. Vasseur C, Rodien P, Beau I, et al. A chorionic gonadotropin-sensitive mutation in the follicle-stimulating hormone receptor as a cause of familial gestational spontaneous ovarian hyperstimulation syndrome. The New England journal of medicine 2003; 349:753-759
116. Stein SA, Oates EL, Hall CR, et al. Identification of a point mutation in the thyrotropin receptor of the hyt/hyt hypothyroid mouse. Mol Endocrinol 1994; 8:129-138
117. Marians RC, Ng L, Blair HC, et al. Defining thyrotropin-dependent and -independent steps of thyroid hormone synthesis by using thyrotropin receptor-null mice. Proceedings of the National Academy of Sciences of the United States of America 2002; 99:15776-15781
118. Postiglione MP, Parlato R, Rodriguez-Mallon A, et al. Role of the thyroid-stimulating hormone receptor signaling in development and differentiation of the thyroid gland. Proceedings of the National Academy of Sciences of the United States of America 2002; 99:15462-15467
119. Stanbury JB, Rocmans P, Buhler UK, et al. Congenital hypothyroidism with impaired thyroid response to thyrotropin. The New England journal of medicine 1968; 279:1132-1136
120. Codaccioni JL, Carayon P, Michel-Bechet M, et al. Congenital hypothyroidism associated with thyrotropin unresponsiveness and thyroid cell membrane alterations. The Journal of clinical endocrinology and metabolism 1980; 50:932-937
121. Sunthornthepvarakui T, Gottschalk ME, Hayashi Y, et al. Brief report: resistance to thyrotropin caused by mutations in the thyrotropin-receptor gene. The New England journal of medicine 1995; 332:155-160
122. Smits G, Campillo M, Govaerts C, et al. Glycoprotein hormone receptors: determinants in leucine-rich repeats responsible for ligand specificity. The EMBO journal 2003; 22:2692-2703
123. Kleinau G, Krause G. Thyrotropin and homologous glycoprotein hormone receptors: structural and functional aspects of extracellular signaling mechanisms. Endocr Rev 2009; 30:133-151
124. Sanders P, Young S, Sanders J, et al. Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J Mol Endocrinol 2011; 46:81-99
125. Krause G, Kreuchwig A, Kleinau G. Extended and structurally supported insights into extracellular hormone binding, signal transduction and organization of the thyrotropin receptor. PloS one 2012; 7:e52920
126. Kleinau G, Neumann S, Gruters A, et al. Novel insights on thyroid-stimulating hormone receptor signal transduction. Endocr Rev 2013; 34:691-724
127. de Roux N, Misrahi M, Brauner R, et al. Four families with loss of function mutations of the thyrotropin receptor. The Journal of clinical endocrinology and metabolism 1996; 81:4229-4235
128. Abramowicz MJ, Duprez L, Parma J, et al. Familial congenital hypothyroidism due to inactivating mutation of the thyrotropin receptor causing profound hypoplasia of the thyroid gland. The Journal of clinical investigation 1997; 99:3018-3024
129. Biebermann H, Schoneberg T, Krude H, et al. Mutations of the human thyrotropin receptor gene causing thyroid hypoplasia and persistent congenital hypothyroidism. The Journal of clinical endocrinology and metabolism 1997; 82:3471-3480
130. Clifton-Bligh RJ, Gregory JW, Ludgate M, et al. Two novel mutations in the thyrotropin (TSH) receptor gene in a child with resistance to TSH. The Journal of clinical endocrinology and metabolism 1997; 82:1094-1100
131. Gagne N, Parma J, Deal C, et al. Apparent congenital athyreosis contrasting with normal plasma thyroglobulin levels and associated with inactivating mutations in the thyrotropin receptor gene: are athyreosis and ectopic thyroid distinct entities? The Journal of clinical endocrinology and metabolism 1998; 83:1771-1775
132. Tonacchera M, Agretti P, Pinchera A, et al. Congenital hypothyroidism with impaired thyroid response to thyrotropin (TSH) and absent circulating thyroglobulin: evidence for a new inactivating mutation of the TSH receptor gene. The Journal of clinical endocrinology and metabolism 2000; 85:1001-1008
133. Bretones P, Duprez L, Parma J, et al. A familial case of congenital hypothyroidism caused by a homozygous mutation of the thyrotropin receptor gene. Thyroid 2001; 11:977-980
134. Jordan N, Williams N, Gregory JW, et al. The W546X mutation of the thyrotropin receptor gene: potential major contributor to thyroid dysfunction in a Caucasian population. The Journal of clinical endocrinology and metabolism 2003; 88:1002-1005
135. Park SM, Clifton-Bligh RJ, Betts P, et al. Congenital hypothyroidism and apparent athyreosis with compound heterozygosity or compensated hypothyroidism with probable hemizygosity for inactivating mutations of the TSH receptor. Clinical endocrinology 2004; 60:220-227
136. Kanda K, Mizuno H, Sugiyama Y, et al. Clinical significance of heterozygous carriers associated with compensated hypothyroidism in R450H, a common inactivating mutation of the thyrotropin receptor gene in Japanese. Endocrine 2006; 30:383-388
137. Tsunekawa K, Onigata K, Morimura T, et al. Identification and functional analysis of novel inactivating thyrotropin receptor mutations in patients with thyrotropin resistance. Thyroid 2006; 16:471-479
138. Yuan ZF, Mao HQ, Luo YF, et al. Thyrotropin receptor and thyroid transcription factor-1 genes variant in Chinese children with congenital hypothyroidism. Endocrine journal 2008; 55:415-423
139. Sura-Trueba S, Aumas C, Carre A, et al. An inactivating mutation within the first extracellular loop of the thyrotropin receptor impedes normal posttranslational maturation of the extracellular domain. Endocrinology 2009; 150:1043-1050
140. Tenenbaum-Rakover Y, Grasberger H, Mamanasiri S, et al. Loss-of-function mutations in the thyrotropin receptor gene as a major determinant of hyperthyrotropinemia in a consanguineous community. The Journal of clinical endocrinology and metabolism 2009; 94:1706-1712
141. Alberti L, Proverbio MC, Costagliola S, et al. Germline mutations of TSH receptor gene as cause of nonautoimmune subclinical hypothyroidism. The Journal of clinical endocrinology and metabolism 2002; 87:2549-2555
142. Xie J, Pannain S, Pohlenz J, et al. Resistance to thyrotropin (TSH) in three families is not associated with mutations in the TSH receptor or TSH. The Journal of clinical endocrinology and metabolism 1997; 82:3933-3940
143. Grasberger H, Vaxillaire M, Pannain S, et al. Identification of a locus for nongoitrous congenital hypothyroidism on chromosome 15q25.3-26.1. Human genetics 2005; 118:348-355
144. Bohr UR, Behr M, Loos U. A heritable point mutation in an extracellular domain of the TSH receptor involved in the interaction with Graves' immunoglobulins. Biochimica et biophysica acta 1993; 1216:504-508
145. Bahn RS, Dutton CM, Heufelder AE, et al. A genomic point mutation in the extracellular domain of the thyrotropin receptor in patients with Graves' ophthalmopathy. The Journal of clinical endocrinology and metabolism 1994; 78:256-260
146. Gabriel EM, Bergert ER, Grant CS, et al. Germline polymorphism of codon 727 of human thyroid-stimulating hormone receptor is associated with toxic multinodular goiter. The Journal of clinical endocrinology and metabolism 1999; 84:3328-3335
147. Simanainen J, Kinch A, Westermark K, et al. Analysis of mutations in exon 1 of the human thyrotropin receptor gene: high frequency of the D36H and P52T polymorphic variants. Thyroid 1999; 9:7-11
148. Muhlberg T, Herrmann K, Joba W, et al. Lack of association of nonautoimmune hyperfunctioning thyroid disorders and a germline polymorphism of codon 727 of the human thyrotropin receptor in a European Caucasian population. The Journal of clinical endocrinology and metabolism 2000; 85:2640-2643
149. Ban Y, Greenberg DA, Concepcion ES, et al. A germline single nucleotide polymorphism at the intracellular domain of the human thyrotropin receptor does not have a major effect on the development of Graves' disease. Thyroid 2002; 12:1079-1083
150. Ho SC, Goh SS, Khoo DH. Association of Graves' disease with intragenic polymorphism of the thyrotropin receptor gene in a cohort of Singapore patients of multi-ethnic origins. Thyroid 2003; 13:523-528
151. Wellcome Trust Case Control C, Australo-Anglo-American Spondylitis C, Burton PR, et al. Association scan of 14,500 nonsynonymous SNPs in four diseases identifies autoimmunity variants. Nature genetics 2007; 39:1329-1337
152. Brand OJ, Barrett JC, Simmonds MJ, et al. Association of the thyroid stimulating hormone receptor gene (TSHR) with Graves' disease. Human molecular genetics 2009; 18:1704-1713
153. Porcu E, Medici M, Pistis G, et al. A meta-analysis of thyroid-related traits reveals novel loci and gender-specific differences in the regulation of thyroid function. PLoS genetics 2013; 9:e1003266
154. Libert F, Lefort A, Gerard C, et al. Cloning, sequencing and expression of the human thyrotropin (TSH) receptor: evidence for binding of autoantibodies. Biochemical and biophysical research communications 1989; 165:1250-1255
155. Nagayama Y, Kaufman KD, Seto P, et al. Molecular cloning, sequence and functional expression of the cDNA for the human thyrotropin receptor. Biochemical and biophysical research communications 1989; 165:1184-1190
156. Biebermann H, Schoneberg T, Schulz A, et al. A conserved tyrosine residue (Y601) in transmembrane domain 5 of the human thyrotropin receptor serves as a molecular switch to determine G-protein coupling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 1998; 12:1461-1471
157. Arseven OK, Wilkes WP, Jameson JL, et al. Substitutions of tyrosine 601 in the human thyrotropin receptor result in increase or loss of basal activation of the cyclic adenosine monophosphate pathway and disrupt coupling to Gq/11. Thyroid 2000; 10:3-10

Hashimoto’s Thyroiditis

ABSTRACT

Hashimoto's thyroiditis is characterized clinically as a commonly occurring, painless, diffuse enlargement of the thyroid gland occurring predominantly in middle-aged women. The patients are often euthyroid, but hypothyroidism may develop. The thyroid parenchyma is diffusely replaced by a lymphocytic infiltrate and fibrotic reaction; frequently, lymphoid germinal follicles are visible. Persons with Hashimoto's thyroiditis have serum antibodies reacting with TG, TPO, and against an unidentified protein present in colloid. In addition, many patients have cell mediated immunity directed against thyroid antigens, demonstrable by several techniques. The incidence is on the order of three to six cases per 10,000 population per year, and prevalence among women is at least 2%. The gland involved by thyroiditis tends to lose its ability to store iodine, produces and secretes iodoproteins that circulate in plasma, and is inefficient in making hormone. Thus, the thyroid gland is under increased TSH stimulation, fails to respond to exogenous TSH, and has a rapid turnover of thyroidal iodine.
Diagnosis is made by the finding of a diffuse, smooth, firm goiter in a young woman, with strongly positive titers of TG Ab and/or TPO Ab and a euthyroid or hypothyroid metabolic status. A patient with a small goiter and euthyroidism does not require therapy unless the TSH level is elevated. The presence of a large gland, progressive growth of the goiter, or hypothyroidism indicates the need for replacement thyroid hormone. Surgery is rarely indicated. Development of lymphoma, though very unusual, must be considered if there is growth or pain in the involved gland.

HISTORICAL REVIEW

In 1912 (Fig. 8-1) Hashimoto described four patients with a chronic disorder of the thyroid, which he termed struma lymphomatosa. The thyroid glands of these patients were characterized by diffuse lymphocytic infiltration, fibrosis, parenchymal atrophy, and an eosinophilic change in some of the acinar cells.(1) Clinical and pathologic studies of this disease have appeared frequently since Hashimoto's original description. The disease has been called Hashimoto's thyroiditis, chronic thyroiditis, lymphocytic thyroiditis, lymphadenoid goiter, and recently autoimmune thyroiditis. Classically, the disease occurs as a painless, diffuse enlargement of the thyroid gland in a young or middle-aged woman. It is often associated with hypothyroidism. The disease was thought to be uncommon for many years, and the diagnosis was usually made by the surgeon at the time of operation or by the pathologist after thyroidectomy. The increasing use of the needle biopsy and serologic tests for antibodies have led to much more frequent recognition, and there is reason to believe that it may be increasing in frequency.(2) It is now one of the most common thyroid disorders.

Figure 1. Dr. Hakaru Hashimoto

The first indication of an immunologic abnormality in this disease was an elevation of the plasma gamma globulin fraction detected by Fromm et al.(3) This finding, together with abnormalities in serum flocculation test results(4) indicated that the disease might be related to a long-continued autoimmune reaction. Rose and Witebsky(5) showed that immunization of rabbits with extracts of rabbit thyroids produced histologic changes in the thyroid glands resembling those seen in Hashimoto's thyroiditis. They also found antithyroglobulin antibodies in the blood of the animals. Subsequently, Roitt et al.(6) observed that a precipitate formed when an extract of human thyroid gland was added to serum from a patient with Hashimoto's thyroiditis. Thus, it appeared that the serum contained antibodies to a constituent of the human thyroid and that these antibodies might be responsible for the disease process. These original observations led directly to entirely new concepts of the causation of disease by autoimmunization.

PATHOLOGY

The goiter is generally symmetrical, often with a conspicuous pyramidal lobe. Grossly, the tissue involved by Hashimoto's thyroiditis is pinkish-tan to frankly yellowish and tends to have a rubbery firmness. The capsular surface is gently lobulated and non-adherent to peri-thyroid structures. Microscopically, there is a diffuse process consisting of a combination of epithelial cell destruction, lymphoid cellular infiltration, and fibrosis. The thyroid cells tend to be slightly larger and assume an acidophilic staining character; they are then called Hurthle or Askanazy cells and are packed with mitochondria. The follicular spaces shrink, and colloid is absent or sparse. Fibrosis may be completely absent or present in degrees ranging from slight to moderate; it may be severe, as observed in subacute or Riedel's thyroiditis. Foreign body giant cells and granulomas are not features of Hashimoto's thyroiditis, in contrast to subacute thyroiditis. In children, oxyphilia and fibrosis are less prominent, and hyperplasia of epithelial cells may be marked. Deposits of dense material representing IgG are found along the basement membrane on electron microscopy (Fig. 8-2).

Figure 2. Electron microscopy image of thyroid tissue from a patient with Hashimoto's thyroiditis, showing electron dense deposits of IgG and TG along the basement membrane of follicular cells.

Within the follicles may be seen clusters of macrophage-like cells. The lymphoid infiltration in the interstitial tissue is accompanied by actual follicles and germinal centers (Fig. 8-3, below). Plasma cells are prominent. Totterman has studied the characteristics of the lymphocytes in the thyroid and reports that they are made up of equal proportions of T and B cells.(7) Most infiltrating T cells have alpha/beta T cell receptors. Gamma/delta T cells are rare(8), although their proportion in intrathyroidal lymphocytes is higher than that in peripheral lymphocytes(9). CD4+CD8+ cells and CD3lo-TCRalpha/beta-lo/CD4-CD8- cells also are present in the infiltrate in the thyroid(9). Infiltrating T cells are considered to be a highly restricted population, based on the study of T cell receptor V alpha(10) and beta(11) gene expression. Heuer et al. studied cytokine mRNA expression in intrathyroidal T cells and found increased expression of IFN-gamma, IL-2 and CD25, which are Th1-related cytokines(12) in Hashimoto's thyroiditis. Thyroglobulin-binding lymphocytes were increased in percentage relative to their occurrence in blood.

Figure 3. Pathology of Hashimoto's thyroiditis. In this typical view of severe Hashimoto's thyroiditis, the normal thyroid follicles are small and greatly reduced in number, and with the hematoxylin and eosin stain are seen to be eosinophilic. There is marked fibrosis. The dominant feature is a profuse mononuclear lymphocytic infiltrate and lymphoid germinal center formation.

The quantity of parenchymal tissue left in the thyroid is variable. In some instances it is actually increased, perhaps as a compensatory hyperplastic response to inefficient iodide metabolism. Typically, the pathologic process involves the entire lobe or gland. Focal thyroiditis, which is microscopically similar, may be found in thyroid glands with diffuse hyperplasia of Graves' disease, in association with thyroid tumors, or in multinodular thyroid glands. The thymus, which is frequently enlarged in thyroiditis as it is in Graves' disease, does not present the picture of enhanced immunologic activity(13),(14). Histologic feature in painless (or silent) thyroiditis is almost similar to that of Hashimoto's thyroiditis. All specimens show chronic thyroiditis, focal or diffuse type: and lymphoid follicles were present in about half of the specimen(15). The follicular distruptions are characteristic and common histologic feature at the time of destructive thyrotoxicois but disappear during the late recovery phase of disease. Thus painless thyroiditis may be induced by the activation of autoimmune reaction within the thyroid gland in patients with Hashimoto's thyroiditis.

PATHOGENESIS

The putative causes of autoimmune thyroid disease (AITD) are reviewed in Chapter 7, and the basic concepts reviewed there apply of course to Hashimoto's thyroiditis. In Hashimoto's thyroiditis, the immunologic attack appears to be typically aggressive and destructive, rather than stimulatory, as in Graves' disease, and the difference is most likely due to the characteristics of the immune response. Hashimoto's thyroiditis is reported to occur in two varieties, an atrophic variety, perhaps associated with HLA-DR3 gene inheritance, and a goitrous form associated with HLA-DR5. The large UK Caucasian HT case control cohort study demonstrated  clear differences in association within the HLA class II region between Hashimoto's thyroiditis and Graves' disease, differences in HLA class II genotype may, in part, contribute to the different immunopathological processes and clinical presentation of these related diseases (15a).  In studies of autoimmune hypothyroidism in monozygotic twins, the concordance rate is below 1 and thus environmental factors are also etiologically important.(16) Concerning susceptibility genes for Hashimoto's thyroiditis, non-MHC class II genes have been recently investigated. A number of data accumulated, demonstrating an association between cytotoxic T cell antigen-4 (CTLA-4), which is a major negative regulator of T-cell mediated immune functions, and autoimmune diseases including Hashimoto's thyroiditis. New studies have appeared on the zinc-finger gene in AITD susceptibility region gene (ZFAT), the thyroglobulin gene, and the protein tyrosine phosphatase-22 (PTPN22) gene. Genome-wide association studies (GWAS) detected other genes including FCRL3, FOXE1 and IL2RA. (16a) Many of the genes associated with AITD are also associated with other autoimmune diseases, which highlights a key role for disrupted T cell central tolerance, antigen monitoring and peripheral immune tolerance in autoimmune onset. Association of polymorphisms in miroRNA  genes (miR499A and miR125A) with autoimmune thyroid diseases were reported (16b).

Regarding environmental factors, high iodine intake, selenium deficiency, pollutants such as tobacco smoke, infectious diseases such as chronic hepatitis C, and certain drugs are implicated in the development of autoimmune thyroiditis (16.1: Duntas LH. Environmental factors and autoimmune thyroiditis. Nat Clin Pract Endocrinol Metab. 2008 Jul 8. [Epub ahead of print]). Long-term iodine exposure leads to increased iodination of thyroglobulin, which increases its antigenicity and initiates the autoimmune process in genetically susceptible individuals. Selenium deficiency decreases the activity of selenoproteins, including glutathione peroxidases, which can lead to raised concentrations of hydrogen peroxide and thus promote inflammation and disease. Such environmental pollutants as smoke, polychlorinated biphenyls, solvents and metals have been implicated in the autoimmune process and inflammation. Environmental factors have not yet, however, been sufficiently investigated to clarify their roles in pathogenesis, and there is a need to assess their effects on development of the autoimmune process and the mechanisms of their interactions with susceptibility genes.

High titers of antibody against thyroglobulin (TG) and thyroid peroxidase (TPO) are present in most patients with Hashimoto's thyroiditis(17), and TPO antibodies are complement fixing and may be cytotoxic. However, the evidence for cytotoxicity is scant, especially since normal transplacental antibody passage of anti-TPO Ab to the human fetus does not usually induce thyroid damage.

Thus it is speculated that cytotoxic T cells, or killer (K) or natural killer (NK) cells, or regulatory T (Treg) or suppressor T cells, may play an important role. A few reports do show T cell line or clone cytotoxicity toward isologous thyroid epithelial cells, and experimental thyroiditis can be transferred by lymphocytes. T cells from patients with Hashimoto's disease proliferate when exposed to TG and TPO. These responses are known to be directed to specific sequences in the TPO molecule, including epitopes at aa 110-129, 210-230, 420-439, and 842-861(18). T cells from mice immunized to TPO react strongly to TPO sequence 540-559, and when immunized with this peptide, develop hypothyroidism and thyroiditis. This peptide may be a central factor in immunity to TPO(18.1). Muixí  et al. identified natural HLA-DR-associated peptides in autoimmune organs that will allow finding peptide-specific T cells in situ (18.2). This study reports a first analysis of HLA-DR natural ligands from ex vivo Graves' disease-affected thyroid tissue. Using mass spectrometry, they identified 162 autologous peptides from HLA-DR-expressing cells, including thyroid follicular cells, with some corresponding to predominant molecules of the thyroid colloid. Most interestingly, eight of the peptides were derived from a major autoantigen, thyroglobulin. In vitro binding identified HLA-DR3 as the allele to which one of these peptides likely associates in vivo. Computer modeling and bioinformatics analysis suggested other HLA-DR alleles for binding of other thyroglobulin peptides. Increased K and NK cell function has been reported in Hashimoto's thyroiditis (19). Dysfunction of regulatory (or suppressor) CD4+ T cell populations may lead to the development of various organ-specific autoimmune diseases including Hashimoto’s thyroiditis (19.1). Despite the lack of understanding of the primary cause(s), it is certain that thyroid autoimmunity drives the lymphocyte collection in the thyroid and is responsible for thyroid epithelial cell damage. Progressive thyroid cell damage can change the apparent clinical picture from goitrous hypothyroidism to that of primary hypothyroidism, or "atrophic" thyroiditis. Primary hypothyroidism is considered to be the end stage of Hashimoto's thyroiditis. In the TSHR-immunized murine model of Graves’ disease, Treg depletion (particularly CD25) induced thyroid lymphocytic infiltrates with transient or permanent hypothyroidism (19.2). Lymphocytic infiltration was associated with intermolecular spreading of the TSHR antibody response to other self thyroid antigens, murine thyroid peroxidase and thyroglobulin. These data suggest a role for Treg in the natural progression of hyperthyroid Graves' disease to Hashimoto's thyroiditis and hypothyroidism in humans.

An alternative cause of "atrophic" hypothyroidism is the development of thyroid stimulation blocking antibodies (TSBAb), which, as the name implies, prevent TSH binding to TSH-R, but do not stimulate thyroid cells and produce hypothyroidism. It has been proposed that TSBAb bind to epitopes near the carboxyl end of the TSH-R extracellular domain, in contrast to thyroid stimulating antibodies (TSAb), which bind to epitopes near aa 40 at the amino terminus(20). This syndrome occurs in neonates, children and adults. The prevalence of TSBAb in adult hypothyroid patients has been reported to be 10%(21). However, in contrast to the usual progressive and irreversible thyroid damage occurring in the usual setting, these blocking antibodies tend to follow the course of TSAb--that is, they decrease or disappear over time, and the patient may become euthyroid again(22). A change from a predominant TSAb response to a predominant TSBAb response can cause patients to have sequential episodes of hyper- and hypothyroid function(23). HLA antigens of hypothyroid patients with TSBAb were found to be different from patients with idiopathic myxedema or Hashimoto's thyroiditis, and rather similar to patients with Graves' disease(24).

In patients with autoimmune hypothyroidism, thyroid dysfunction might be induced by cytokine-mediated apoptosis of thyroid epithelial cells and infiltrating T lymphocytes may not directly be involved in thyrocyte cell death during Hashimoto' s thyroiditis. Fragmented DNA, a characteristic feature of apoptosis, was frequently found in the thyroid follicular cells in Hashimoto's thyroiditis(25). The ligand for Fas(Fas L)was shown to be constitutively expressed on thyrocytes and lL-1alpha, abundantly produced in the thyroid gland of Hashimoto's thyroiditis, induced Fas expression on thyrocytes. Thus Fas-FasL interaction on thyrocytes may induce apoptosis and thyroid cell destruction(26). In the thyroid follicle cells of Hashimoto's thyroiditis, Fas and FasL are strongly stained and immunostaining of Bcl-2 is weak, suggesting that cytokines cause up-regulation of apoptosis(27). Increased serum TSH may inhibit Fas-mediated apoptosis of thyrocytes(28). In contrast TSBAb block the inhibitory action of TSH toward Fas-mediated apoptosis and thus induce thyroid atrophy. On the other hand, transgenic expression of Fas L on thyroid follicular cells actually prevents autoimmune thyroiditis, possibly through inhibition of lymphocyte infiltation(29). Other death-receptor ligands might participate in  and TNF-related apoptosis-includingathyrocyte killing, including TNF- ligand(TRAIL)(30) . In relation to the Fas-Fas L system, Dong et al. reported that mutations of Fas, which induce loss of function, were found in thyroid lymphocytes in 38.1% of patients with Hashimoto's thyroiditis(31). These mutations are found in 65.4% of patients with malignant lymphoma(32), which usually develops from Hashimoto's thyroiditis. These changes are possibly important for progression of Hashimoto's thyroiditis.

Apparent de-novo development of antibodies, augmentation of pre-existing thyroid autoimmunity, goiter, and hypothyroidism, are induced in some cancer patients, when given courses of IL2, IL2a plus lymphokine activated K cells and/or IFN-gamma. It is thought that the phenomenon may reflect activation of lymphocytes by the lymphokine and lymphokine and cell-mediated attack on thyroid tissue(33). Activated lymphocytes release TNFalpha and IFNgamma, which can injure or suppress TEC function. IFNgamma may also augment thyrocyte HLA-DR expression, which could make the thyrocyte able to present self-antigens. Interferon alpha therapy for chronic active type C hepatitis also augments pre-existing thyroid autoimmunity and can induce autoimmune hypothyroidism. A humanised anti-CD52 monoclonal antibody, Campath-1H may permit the generation of antibody-mediated thyroid autoimmunity (33a,b). Campath-1H depletes lymphocytes and monocytes, and may cause the immune response to change from the Th1 phenotype.

T helper type 17 (Th17) lymphocytes, which produce a proinflammatory cytokine IL-17, have recently been shown to play a major role in numerous autoimmune diseases that had previously been thought to be Th1-dominant diseases, such as Hashimoto’s thyroiditis. It is reported that there is an increased differentiation of Th17 lymphocytes and an enhanced synthesis of Th17 cytokines in Hashimoto's disease (33c). In a mouse model of Hashimoto's thyroiditis, iodine-induced autoimmune thyroiditis in nonobese diabetic-H2(h4) mice, both Th1 and Th17 cells are found to be critical T(eff) subsets for the pathogenesis of spontaneous autoimmune thyroiditis (33d). Imbalance of Th17/Treg is reported in different subtypes of autoimmune thyroid diseases. Increased Th17 lymphocytes may play a more important role in the pathogenesis of HT and GO while decreased Treg may be involved in Graves’ disease (33d.1). In contrast, a significant decrease in the ratios of CD4 + IL17+/CD4 + CD25 + CD127 - (p < 0.0001) and CD4 + IL17+/CD4 + CD25 + CD127 - FoxP3 + (p < 0.0001) T cells was obsereved in Hashimoto’s thyroiditis in comparison to healthy children  (33d.2).

The IgG4-related disease (IgG4-RD) is a new disease entity first proposed in relation to autoimmune pancreatitis (AIP) by Hamano et al. in 2001 (33e). A high prevalence of hypothyroidism has been reported in patients with AIP (33f). In 2009, it was reported that on the basis of the immunohistochemistry of IgG4, HT can be divided into two groups, which were proposed as IgG4 thyroiditis (IgG4-positive plasma cell-rich group) and non-IgG4 thyroiditis (IgG4-positive plasma cellpoor group) (33g). The IgG4 thyroiditis group shows indistinguishable histological features and may have a close relationship with IgG4-RD in other organs. In 2010, it was demonstrated that IgG4 thyroiditis is clinically associated with a lower female-to-male ratio, more rapid progress, subclinical hypothyroidism, diffuse low echogenicity, and a higher level of circulating thyroid autoantibodies than non-IgG4 thyroiditis (33h). Riedel thyroiditis (RT) is another candidate for IgG4-RD. It is a rare form of chronic thyroiditis, characterized by inflammatory proliferative fibrosis which involves the thyroid parenchyma and surrounding tissue structures. In 2010, Dahlgren et al. reported that IgG4-RD was the underlying condition in a part of the cases of RT (33i). When IgG4-RD occurs in a systemic pattern, the thyroid involvement may present as RT rather than HT (33j).

Iodine consumption influences the incidence of Hashimoto's thyroiditis and hypothyroidism (see below: “Iodide Metabolism and Effects” in this chapter). Smoking has also been identified as a risk factor for hypothyroidism, but the reason for the association is unknown (34).

An increase in the prevalence of thyroid autoantibodies (ATAs) was reported 6-8 yr after the Chernobyl accident in radiation-exposed children and adolescents (34a). TPOAb prevalence in adolescents exposed to radioactive fallout was still increased in Belarus 13-15 yr after the Chernobyl accident (34b). This increase was less evident than previously reported and was not accompanied by thyroid dysfunction. These data suggest that radioactive fallout elicited a transient autoimmune reaction, without triggering full-blown thyroid autoimmune disease. Longer observation periods are needed to exclude later effects.

Celiac disease was positively associated with hypothyroidism (Hazard Ratio = 4.4; 95% Confidence Interval = 3.4-5.6; p < 0.001), thyroiditis (3.6; 1.9-6.7; p < 0.001) and hyperthyroidism (2.9; 2.0-4.2; p < 0.001) (34c). The highest risk estimates were found in children (hypothyroidism 6.0; 3.4-10.6, thyroiditis 4.7; 2.1-10.5 and hyperthyroidism 4.8; 2.5-9.4). In post-hoc analyses, where the reference population was restricted to inpatients, the adjusted HR for hypothyroidism was 3.4 (2.7-4.4; p < 0.001), thyroiditis 3.3 (1.5-7.7; p < 0.001) and hyperthyroidism 3.1 (2.0-4.8; p < 0.001).This indicates shared etiology and that these individuals are more susceptible to autoimmune disease.

Hashimoto thyroiditis is often associated with type 1 diabetes and other autoimmune disorders such as coeliac disease, type 2 and type 3 polyglandular autoimmune disorders (APS). Type 2 APS is defined by the occurrence of Addison's disease with thyroid autoimmune disease and/or Type 1 diabetes mellitus. Type 3 APS is thyroid autoimmune diseases associated with other autoimmune diseases (excluding Addison's disease and/or hypoparathyroidism). Clinically overt disorders are considered only the tip of the autoimmune iceberg, since latent forms are much more frequent (34d). Hashimoto thyroiditis is also often associated in lymphocytic hypophysitis (34e).

There is a report that microRNAs (miRNAs) miR-146a1, miR-155_2, and miR-200a1 are altered in AITD. In the thyroid tissue of the GD group, miR-146a1 was significantly decreased in comparison to the control group (mean relative expression 5.17 vs. 8.37, respectively, p = 0.019). In the HT group, miR-155_2 was significantly decreased in comparison to the control group (8.30 vs. 11.20, respectively, p = 0.001), and miR-200a1 was significantly increased (12.02 vs. 8.01, p = 0.016) (34f). The expression levels of miRNAs in plasma and peripheral blood mononuclear cells showed wide individual variation, and the these levels may be associated with the pathogenesis of autoimmune thyroid diseases (34g). Accumulating data suggest that miRNAs crucially control innate and adaptive immune responses, and implicate some miRNAs as having an important role in the pathophysiology of immunity and autoimmunity. (34h) For example, miR-155_2 was previously shown to possess important functions in the mammalian immune system. (34i) MicroRNA-142-5p may contribute to Hashimoto's thyroiditis by targeting CLDN1.(34j)

 INCIDENCE AND DISTRIBUTION

The incidence of Hashimoto's thyroiditis seen in practice is unknown but is roughly equal to that of Graves' disease (on the order of 0.3 - 1.5 cases per 1,000 population per year.)(35-37) The disease is 15 - 20 times as frequent in women as in men. It occurs especially during the decades from 30 to 50, but may be seen in any age group, including children. It is certain that it exists with a much higher frequency than is diagnosed clinically, and its frequency seems to be increasing. Family studies always bring to light a number of relatives with moderate enlargement of the thyroid gland suggestive of Hashimoto's thyroiditis. Many of these persons have TG and TPO antibodies, and most are entirely asymptomatic. Inoue et al. found 3% of Japanese children aged 6 - 18 to have thyroiditis(38). In most instances, biopsy revealed focal rather than diffuse thyroiditis.

In addition to overt thyroiditis, roughly 10% of most populations have positive TG and TPO antibody test results(35-37) in the apparent absence of thyroid disease by physical examination. In a classic study of an entire community, Tunbridge et al.(37) found that 1.9 - 2.7% of women had present or past thyrotoxicosis, 1.9% had overt hypothyroidism, 7.5% had elevated TSH levels, 10.3% had test results positive for TPO (microsomal antigen) Ab measured by hemagglutination assay (MCHA), and about 15.0% had goiter. Men had 10 to 4-fold lower incidence of thyroid abnormalities. In a study of children whose parents had history of thyroid disease, Carey et al.(39) found a 24% prevalence of thyroid "abnormalities", including a prevalence of 6.9% abnormal thyroids, and 9.3% with positive TG Ab measured by hemagglutination assay (TGHA) and 7.8% positive MCHA assays. Gordin et al.(35) found that 8% of adult Finns had positive TGHA results, and 26% had positive MCHA results. TSH levels were elevated in 30% of these persons. On the basis of positive antibody titers and elevated TSH levels, 2 - 5% were believed to have asymptomatic thyroiditis. These test results correlate with focal collection of lymphocytes on histologic examination of the thyroid glands(40), are frequently associated with elevated levels of TSH(41), and probably represent one end of a spectrum of thyroid damage. Women with both positive antibody test results and raised TSH levels become hypothyroid at the rate of 5%/year(42). A reasonable approximation of the prevalence of positive antibody tests in women is greater than 10%, and of clinical disease is at least 2%. Men have one-tenth this prevalence. A number of small datasets have suggested a potential role for skewed X chromosome activation (XCI), away from the expected 50:50 parent of origin ratio, as an explanation for the strong female preponderance seen in the common autoimmune thyroid diseases (AITD), Graves’ disease (GD) and Hashimoto’s thyroiditis (HT). (42a) A possible role for fetal cell microchimerism in triggering an autoimmune process has been repeatedly proposed, based on the evidence that autoimmune diseases have a higher prevalence in females, with peak incidence in women of childbearing age. Fetal microchimeric cells have been found to be significantly more represented within the thyroid gland of women with Hashimoto's thyroiditis and Graves' disease compared to those without thyroid autoimmunity, suggesting a pathogenic role. (42b, c, d)

 

COURSE OF THE DISEASE (Table 8-1)

Hashimoto's thyroiditis begins as a gradual enlargement of the thyroid gland and gradual development of hypothyroidism. It is often discovered by the patient, who finds a fullness of the neck or a new lump while self-examining because of a vague discomfort in the neck. Perhaps most often, it is found by the physician during the course of an examination for some other complaint.

Table 1. Presentations of Hashimoto's Thyroiditis

1.     Euthyroidism and goiter

2.     Subclinical hypothyroidism and goiter

3.     Primary thyroid failure

4.     Hypothyroidism

5.     Adolescent goiter

6.     Painless thyroiditis or silent thyroiditis

7.     Postpartum painless thyrotoxicosis

8.     Alternating hypo- and hyperthyroidism

In some instances the thyroid gland may enlarge rapidly; rarely, it is associated with dyspnea or dysphagia from pressure on structures in the neck, or with mild pain and tenderness. Rarely, pain is persistent and unresponsive to medical treatment and requires medical therapy or surgery. The goiter of Hashimoto's thyroiditis may remain unchanged for decades(37), but usually it gradually increases in size. Occasionally the course is marked by symptoms of mild thyrotoxicosis, especially during the early phase of the disease. Symptoms and signs of mild hypothyroidism may be present in 20% of patients when first seen(41), or commonly develop over a period of several years. Progression from subclinical hypothyroidism (normal FT4 but elevated TSH) to overt hypo-thyroidism occurs in a certain fraction (perhaps 3-5%) each year. Eventually thyroid atrophy and myxedema may occur(43). This assertion is based on the clinical observation that patients with Hashimoto's thyroiditis often develop myxedema, and the knowledge that patients with myxedema due to atrophy of the thyroid have a high incidence of TG Ab in their serum. The disease frequently produces goitrous myxedema in young women, and we have occasionally observed a goitrous and hypothyroid patient who went on to develop thyroid atrophy.Occasionally, patients with Hashimoto痴 thyroiditis have persistent pain which is unresponsive to nonsteroidal anti-inflammatory drugs, replacement with thyroid hormone, and recurs after therapy with steroids. Kon and DeGroot recently reported seven patients who finally came to subtotal or near-total thyroidectomy, some of whom received subsequent radioactive iodide thyroid ablation, with final relief of symptoms (Kon, YC; DeGroot, LJ. Painful Hashimoto痴 thyroiditis as an indication for thyroidectomy: clinical characteristics and outcome in seven patients. J Clin Endocrinol Metab 88 2667-2672 2003).

Generally the progression from euthyroidism to hypothyroidism has been considered an irreversible process due to thyroid cell damage and loss of thyroidal iodine stores (Fig. 8-4). However, it is now clear that up to one-fourth of patients who are hypothyroid may spontaneously return to normal function over the course of several years. This sequence may reflect the initial effect of high titers of thyroid stimulation blocking antibodies which fall with time and allow thyroid function to return(23).

Figure 4. Fluorescent thyroid scan in thyroiditis. The normal thyroid scan (left) allows identification of a thyroid with normal stable (127I) stores throughout both lobes. A marked reduction in 127I content is apparent throughout the entire gland involved with Hashimoto's thyroiditis (right).

Within the past few years, several unusual syndromes believed to be associated with or part of the clinical spectrum of Hashimoto's thyroiditis have been described. Occasional patients develop amyloid deposits in the thyroid (44). Shaw et al.(45) described five patients with a relapsing steroid-responsive encephalopathy including episodes like stroke and seizures, high CSF protein, abnormal EEG, and normal CAT scans (see Hashimoto's encephalopathy below). Khardon et al.(46) described a steroid responsive lymphocytic interstitial pneumonitis in four patients. It remains uncertain how these illnesses relate to lymphocytic thyroiditis, which has until now been largely identified as an organ specific disease.

At 5 years of follow-up of the natural course of euthyroid Hashimoto's thyroiditis in Italian children, more than 50% of the patients remained or became euthyroid (46-1). The presence of goiter and elevated TGab at presentation, together with progressive increase in both TPOab and TSH, may be predictive factors for the future development of hypothyroidism.

Hashimoto's thyroiditis and hypothyroidism are associated with Addison's disease, diabetes mellitus, hypogonadism, hypopara-thyroidism, and pernicious anemia. Such combinations are described as the polyglandular failure syndrome. Two forms of polyglandular autoimmunity have been recognized(47). In the Type I syndrome patients have hypoparathyroidism, muco-cutaneous candidiasis, Addison's disease, and occasionally hypothyroidism. Type II, more frequent, often includes familial associations of diabetes mellitus, hypothyroidism, hypoadrenalism, and occasionally gonadal or pituitary failure. In these syndromes, antibodies reacting with the affected end organs are characteristically present. Vitiligo, hives, and alopecia are associated with thyroiditis. There is also a clear association with primary and secondary Sjogren's syndrome(48). Some patients appear to start with Hashimoto's thyroiditis, and progress with time to the picture of Riedel's thyroiditis including the frequently-associated retroperitoneal fibrosis(49).

Musculoskeletal symptoms, including chest pain, fibrositis, and rheumatoid arthritis, occur in one-quarter of patients(50), and of course, any of the musculoskeletal symptoms of hypothyroidism may likewise occur.

It has been suggested that thyroiditis predisposes to vascular disease and coronary occlusion. Abnormally elevated titers of thyroid autoantibodies and the morphologic changes of thyroiditis are said to occur with an increased frequency among patients with coronary artery disease. Mild hypothyroidism(51) associated with asymptomatic atrophic thyroiditis could predispose patients to heart disease. Others have failed to find increased TG Ab in-patients with coronary artery disease(52) or increased coronary disease in association with thyroiditis.

Although chronic inflammation, leading to neoplastic transformation, is a well-established clinical phenomenon, the link between Hashimoto’s thyroiditis and thyroid cancer remains controversial (52a, b). Larson et al. reported that patients with Hashimoto’s thyroiditis were three times more likely to have thyroid cancer, suggesting a strong link between chronic inflammation and cancer development (52-1). PI3K/Akt expression was increased in both Hashimoto’s thyroiditis and well-differentiated thyroid cancer, suggesting a possible molecular mechanism for thyroid carcinogenesis. Thyroid cancer may be associated with less aggressive disease and better outcome in patients with coexisting Hashimoto’s thyroiditis. (52b, c, d)

In children, retarded growth, retarded bone age, decreased hydroxyproline excretion, and elevated cholesterol levels may be seen (Fig. 8-5).

Figure 5. Identical male twins with Hashimoto's thyroiditis were photographed at age 12. At age 8, they had the same height and appearance. During the intervening 4 years, small goiters developed and the growth of the twin on the right almost stopped. Biopsy indicated Hashimoto's thyroiditis in each twin's thyroid.

Hashimoto's Thyroiditis in Identical Twin Boys*

D.L. was seen at age 12 for failure to grow over the past 4 years. The patient had an identical twin, whose development up to age 8 had been entirely normal. Pubertal changes had developed at age 11. No goiter had been noted.

On physical examination, he was a short, cooperative, pubertal boy of normal intelligence, 129 cm in height and 35 kg in weight. The thyroid gland was smooth and firm, and of normal size. The skin was dry, cool, and mottled. Reflex relaxation was delayed. Estimated T4 levels were < 4 ug/dl, and the 24-hour RAIU was 4%. Thyroid scan showed a normal thyroid gland. Bone age was 8 years. The potassium thiocyanate discharge test result was negative. Thyroid biopsy showed a moderately diffuse lymphocytic infiltrate with lymphoid germinal centers and a diffuse, dense fibrous reaction.

R.L. was seen simultaneously with D.L. and was an active, healthy-appearing boy with early pubertal changes. His height was 149 cm, and his weight was 39.7 kg. The pulse was 104. The skin was normal. The thyroid gland was enlarged to about three times the normal size and was not nodular. PBI levels were 6.4 and 7.2 ug/dl, and the 24- hour RAIU was 21%. Bone age was 11 years. A potassium thiocyanate discharge test caused no decrease in neck radioactivity. Biopsy showed diffuse lymphocytic infiltration, lymphoid follicles and germinal centers, atrophy of thyroid follicles, oxyphilic cytoplasm, and dense fibrosis.

Similar fingerprints, similar lip and ear shapes, and identity of 15 blood factors indicated that they were identical twins. There was no family history of thyroid disease.

Iodide kinetic studies showed rapid turnover of thyroid iodide and production of excess quantities of plasma butanol-insoluble iodine. Hemagglutination test results for TG Ab were negative, but an immunofluorescence assay showed a strongly positive reaction against a cytoplasmic antigen. Bioassay of the serum for thyroid-stimulating activity gave a TSH-type response.* These patients were studied in cooperation with Dr. William H. Milburn, to whom we are greatly indebted.

When goiter is induced by iodine administration, lymphocytic thyroiditis is frequently found and thyroid autoantibodies are often present(53).

Remission of Hashimoto's thyroiditis, with loss of goiter, hypothyroidism, and serum thyroid autoantibodies, has been reported during pregnancy, with relapse after delivery(54). Antibody levels usually fall during pregnancy(55). These phenomena may reflect the immunosuppressive effects of pregnancy. After delivery thyroid autoantibody levels rise, and after 2-6 months there may be sudden development (? return) of goiter and hypothyroidism (56). Concerning management of thyroid dysfunction during pregnancy and postpartum, an Endocrine Society Clinical Practice Guideline was developed (56a, Chapter 14). Management of thyroid diseases during pregnancy requires special considerations because pregnancy induces major changes in thyroid function, and maternal thyroid disease can have adverse effects on the pregnancy and the fetus. Care requires coordination among several healthcare professionals. Avoiding maternal (and fetal) hypothyroidism is of major importance because of potential damage to fetal neural development, an increased incidence of miscarriage, and preterm delivery. Maternal hyperthyroidism and its treatment may be accompanied by coincident problems in fetal thyroid function. Autoimmune thyroid disease is associated with both increased rates of miscarriage, for which the appropriate medical response is uncertain at this time, and postpartum thyroiditis. Fine-needle aspiration cytology should be performed for dominant thyroid nodules discovered in pregnancy. Radioactive isotopes must be avoided during pregnancy and lactation. Universal screening of pregnant women for thyroid disease is not yet supported by adequate studies, but case finding targeted to specific groups of patients who are at increased risk is strongly supported. One report recommended screening all pregnant women for autoimmune thyroid disease in the first trimester in terms of cost-effectiveness (56b).

Of course maternal antibodies cross the placenta, and as in Graves' disease, may affect the fetus and neonate. TPO and TG Ab typically appear to have no adverse effect. Some evidence suggests cytotoxic antibodies, which are thought to be different from TPO Ab or TG Ab, could cause fetal hypothyroidism(57). However, TSBAb can rarely produce neonatal hypothyroidism, which is self-limiting over 4-6 weeks as the maternal IgG is metabolized. Women with positive TPO antibody before assisted reproduction have a significantly increased risk for miscarriage, with an odds ratio of 3.77 (Poppe, K; Glinoer, D; Tournaye, H; Devroey, P; van Steirteghem, A; Kaufman, L; Velkeniers, B. Assisted reproduction and thyroid autoimmunity: an unfortunate combination? J Clin Endocrinol Metab 88 4149-4152 2003).

Y.L.C., 24-Year-Old Woman, Postpartum, Not-So-Transient Hypothyroidism

The patient had menarche at age 16 and had regular periods. She married at age 24 and was not able to conceive. After receiving danazol therapy for 7 months for treatment of extensive endometriosis, she became pregnant and delivered after 36 weeks' gestation. During the course of this pregnancy, her thyroid gland was noted to be normal; no thyroid function tests were done. After delivery, she nursed the infant for 1 week. She then stopped nursing, but galactorrhea and amenorrhea continued for the next 5 months. After the fourth month, she was noted to have an enlarged thyroid gland; the FT4I was found to be 3.4 (normal, 6.0 - 10.5) and TSH level 27 uU/ml. There were symptoms of mild hypothyroidism, with some lowering of the voice and increase in fatigue. A sister had an overactive thyroid and mild exophthalmos.

Her thyroid was estimated to weigh about 40 g, with a smooth surface and an enlarged lobe. Skin was dry, and there was some delay in the reflex relaxation. TGAb were present at a titer of 1/160 and TPOAb at 1/20480. Serum T3 level was 123 ng/dl, and the RAIU was 16% at 4 hours and 32% at 24 hours. The thyroid scan was within normal limits. Prolactin (PRL) level was elevated at 43 ng/ml. Sella turcica X-ray films and a CT scan of the head were normal.

It was hypothesized that the patient had postpartum hypothyroidism due to transient exacerbation of thyroiditis and that this condition might resolve spontaneously. Whether the hyperprolactinemia, amenorrhea, and galactorrhea were secondary to the hypothyroidism or were independent problems was at first unclear. The patient was treated expectantly, since she appeared to be in no distress and there was no evidence of pituitary tumor. One month after the initial observations, the TSH level had fallen to 13.5 uU/ml and the T3 level remained at 126 ng/dl. Eight weeks later, the FT4I had risen to 5.8, the T3 level was 113 ng/dl, TSH 9.1 uU/ml, and the PRL remained at 66 ng/ml. Later, all test results became normal.

Painless (silent) and Postpartum Thyroiditis

In the last decade several syndromes involving clinically significant, but self-limited, exacerbations of AITD have been delineated(54)-(59). Silent (painless) thyroiditis is a syndrome that has a clinical course of thyroid dysfunction similar to subacute thyroiditis but with no anterior neck pain and no tenderness of the thyroid. Initially, patients have a thyrotoxic phase, later passing through euthyroidism to hypothyroidism and, finally, return to euthyroidism. Postpartum thyroiditis occurs within 6 months after delivery and runs an identical clinical course(57). Postpartum thyroiditis is now considered to be identical to silent thyroiditis, and this term is used for patients who developed silent thyroiditis in the postpartum period(57). After delivery, other forms of autoimmune thyroid dysfunction also occur, including Graves' disease, transient hypothyroidism without preceding destructive thyrotoxicosis, and persistent hypothyroidism (Fig. 8-6). In recent years, the term painless thyroiditis also has been used frequently, and the same disorder has been described using different names, such as thyrotoxicosis with painless thyroiditis(60), occult subacute thyroiditis(61), hyperthyroiditis(64), lymphocytic thyroiditis with spontaneously resolving hyperthyroidism(62), painless thyroiditis and transient hyperthyroidism without goiter(63), and transient hyperthyroidism with lymphocytic thyroiditis(65). The thyrotoxicosis is induced by leakage of intrathyroidal hormones into the circulation caused by damage to thyroid epithelial cells from inflammation. Thus the thyroid radioactive iodine uptake (RAIU) is low(59). Therefore, the early phase of thyrotoxicosis in silent thyroiditis, postpartum thyroiditis, and subacute thyroiditis can be grouped together as destruction-induced thyrotoxicosis or simply as destructive thyrotoxicosis(66). When the measurement of radioactive iodine uptake is difficult, the measurement of anti-TSH receptor antibody and/or thyroid blood flow by ultrasonography may be useful to differentiate between destruction-induced thyrotoxicosis and Graves' thyrotoxicosis. The quantitative measurement by power Doppler ultrasonography was more effective than that of anti-TSH receptor antibody for differential diagnosis of these two types of thyrotoxicosis and may omit the radioactive iodine uptake test (66-1).

Figure 6. 

Much evidence, including histopathological and immunological studies, indicates that this disorder is an autoimmune thyroid disease(68). It is believed to be due to autoimmune induced damage to the thyroid causing excess hormone release, and for this reason is not responsive to antithyroid drugs, KI or KCLO4, but does, if treatment is necessary, respond to prednisone(67). During the clinical course of subclinical or very mild autoimmune thyroiditis, aggravating factors cause exacerbation of the destructive process. All women with subclinical autoimmune thyroiditis(40) and antithyroid microsomal antibodies of more than 1:5120 before pregnancy develop postpartum thyroiditis(57). A significant percentage of patients with silent thyroiditis have personal or family histories of autoimmune thyroid disease. Most patients have a complete remission, but some develop persistent hypothyroidism(70). Some patients have had alternating episodes of typical "high-uptake" thyrotoxicosis and episodes of "transient" low-uptake thyrotoxicosis(69). Recurrence of disease is common in silent thyroiditis but very rare in subacute thyroiditis. Considering all these data, it is assumed that silent thyroiditis is caused by an exacerbation of autoimmune thyroiditis induced by aggravating factors. Thyroiditis frequently recurs, and seasonal allergic rhinitis is reported to be an initiation factor(71). Physically vigorous massage on the neck also was reported to be a contributing factor for silent thyroiditis(72). The prevalence of silent thyroiditis, including postpartum disease, is around 5 per cent of all types of thyrotoxicosis. Spontaneous silent thyroiditis is three times more frequent than postpartum thyroiditis.

An immune rebound mechanism has been established for the induction of postpartum thyroiditis(57). Postpartum thyroid destruction is associated with an increase in NK cell counts and activity(57). Cessation of steroid therapy has initiated silent thyroiditis in a patient with autoimmune thyroiditis and rheumatoid arthritis(73), presumably because this also allows immune rebound. In patients with Cushing's syndrome who have associated subclinical autoimmune thyroiditis, silent thyroiditis has occurred after unilateral adrenalectomy(74). Typically, painless thyroiditis or destructive thyrotoxicosis occurs at 2 to 4 months postpartum. The prevalence of postpartum thyroiditis ranges from 3 to 8 per cent of all pregnancies(57).POSSIBLE PREVENTION OF PPT-In a randomized prospective controlled study, 77 TPO+ pregnant women received 200 ug selenomethionine daily starting at the 12th week of pregnancy, and 74 TPO+ women received a placebo. The treated group had significantly lower TPO antibody levels at the end of pregnancy and during the post-partum while on treatment. The incidence of PPT was reduced from 48.6 to 28.6% in the treated group, and the incidence of permanent hypothyroidism was equivalently reduced. Thyroid hormone levels did not differ.( Negro R, Greco G, Mangieri T, Pezzarossa A, Dazzi D, Hassan H. The influence of selenium supplementation on postpartum thyroid status in pregnant women with thyroid peroxidase autoantibodies. J Clin Endocrinol Metab. 2007 Apr;92(4):1263-8) .

Hashimoto's encephalopathy

Hashimoto's encephalopathy or encephalitis is a very rare complication of Hashimoto's thyroiditis. Neurological complications are sometimes associated with thyroid dysfunction but patients with this encephalopathy are usually euthyroid. It is treatable, steroid-responsive, progressive or relapsing encephalopathy associated with elevation of thyroid specific autoantibodies (75). This condition was first described in 1966 (76) and may present as a subacute or acute encephalopathy with seizures and stroke-like episodes, often in association with myoclonus and tremor (77). It is associated with abnormal EEG and high CSF proteins without pleocytosis. Some patients suffer from a significant residual disability(78). Antibody to α-enolase has been identified in some patients (79) but this antibody is also frequently found in other autoimmune diseases. Sawka et al. reported that this condition is not caused by thyroid dysfunction or antithyroid antibodies but represents an association of an uncommon autoimmune encephalopathy with a common autoimmune thyroid disease (80). Identification of antibodies to brain specific antigens may disclose the real pathogenesis of this condition. Recently, autoantibodies against the amino (NH2)-terminal of α-enolase (referred to as NAE) were reported to be highly specific in sera from a limited number of HE patients (68-83% with HE; 11%, 2 of 17 with HT without any neuropsychiatric features; none of controls [50 individuals] including those with other neurological or immunological conditions involving encephalopathy [25 individuals]) (80.1, 80.2). Steroid reversible cerebral hypometabolism was recently documented by PET scanning in this condition. (80.3) There is a report that Hashimoto’s encephalopathy associated with elevated intrathecal and serum IgG4 levels. (80.3a) Additional case studies, including histological investigations as well as measurements of IgG4, are needed to elucidate the pathological role of IgG4 in Hashimoto’s encephalopathy.

Hashimoto's ophthalopathy

Thyroid-associated orbitopathy (TAO) usually occurs in Graves’s disease with hyperthyroidism, and sometimes in euthyroid and hypothyroid patients. Since most euthyroid and hypothyroid patients with orbitopathy are thyrotropin receptor antibody (TRAb)-positive, they are diagnosed as having euthyroid Graves’ disease or hypothyroid Graves’ disease. When euthyroid and hypothyroid patients with orbitopathy are TRAb-negative but associated with Hashimoto’s thyroiditis, “Hashimoto's ophthalopathy” may be considered (80.4, 80.5). Because patients with Hashimoto’s thyroiditis test negative for TRAb, other autoantibodies against an eye muscle antigen, such as calsequestrin, flavoprotein, or G2s are postulated (80.6).

IODIDE METABOLISM AND EFFECTS

Many patients with Hashimoto's thyroiditis do not respond to injected TSH with the expected increase in RAIU or release of hormone from the gland(81). These findings probably mean that the gland is partially destroyed by the autoimmune attack and is unable to augment iodine metabolism further. Further, the thyroid gland of the patient with Hashimoto's disease does not organify normally(82) (Fig. 8-4). Administration of 400 mg potassium perchlorate 1 hour after giving a tracer iodide releases 20 - 60% of the glandular radioactivity. Also, a fraction of the iodinated compounds in the serum of patients with Hashimoto's thyroiditis is not soluble in butanol, as are the thyroid hormones, but is an abnormal peptide-linked iodinated component. This low-weight iodoprotein is probably serum albumin that has been iodinated in the thyroid gland. A similar iodoprotein is also found in several other kinds of thyroid disease, including carcinoma, Graves' disease, and one form of goitrous cretinism. It may be formed as part of the hyperplastic response. TG is also detectable in their serum.

Iodide is actively transported from blood to thyrocytes and recently the sodium / iodide symporter (NIS) has been cloned. Antibodies against NIS were found in autoimmune thyroid disease(83). This antibody has an inhibitory activity on iodide transport and may modulate the thyroid function in Hashimoto's thyroiditis. More recent studies reported rather low prevalence (less than 10%) of anti-NIS antibodies in Hashimoto's disease and clinical relevance is still unknown(84),(85).

In animal experiment iodine depletion prevents the development of autoimmune thyroiditis(86). It is suggested that mild iodine deficiency partly protect against autoimmune thyroid disease(87), although it is controversial(88). In a region where iodine-containing food (such as seaweed) is common, as in Japan, excessive dietary iodine intake (1000 micro g/day or more) may cause transient hypothyoidism in patients with subclinical autoimmune thyroiditis. This condition is easily reversible with a reduction in iodine intake(89). Iodine is important not only for thyroid hormone synthesis but also for induction and modulation of thyroid autoimmunity. In general, iodine deficiency attenuates, which iodine excess accelerates autoimmune thyroiditis in autoimmune prone individuals(90). In animal experiment, it is revealed that enhanced iodination of thyroglobulin facilitates the selective processing and presentation of a cryptic phatogenic peptide in vivo or in vitro. Moreover, it is suggested that iodine excess stimulates thymus development and effects function of various immune cells(91).

DIAGNOSIS

Diagnosis involves two considerations -- the differential diagnosis of the thyroid lesion and the assessment determination of the metabolic status of the patient.

A diffuse, firm goiter with pyramidal lobe enlargement, and without signs of thyrotoxicosis, should suggest the diagnosis of Hashimoto's thyroiditis. Most often the gland is bosselated or "nubbey." It is usually symmetrical, although much variation in symmetry (as well as consistency) can occur. The trachea is rarely deviated or compressed. The association of goiter with hypothyroidism is almost diagnostic of this condition, but is also seen in certain syndromes due to defective hormone synthesis or hormone response, as described in Chapter 9. Pain and tenderness are unusual but may be present. A rapid onset is also unusual, but the goiter may rarely grow from normal to several times the normal size in a few weeks. Most commonly the gland is two to four times the normal size. Satellite lymph nodes may be present, especially the Delphian node above the isthmus. Multinodular goiter occurs in significant incidence in adult women; thus the co-occurrence of multinodular goiter and Hashimoto's thyroiditis is not rare, and may provide the finding of a grossly nodular gland in a patient who is mildly hypothyroid and has positive antibody tests.

The T4 concentration and the FT4 range from low to high but are most typically in the normal or low range(92). The RAIU (rarely required) is variable and ranges from below normal to elevated values, depending on such factors as TSH levels, the efficiency of use of iodide by the thyroid, and the nature of the components being released into the circulation. Gammaglobulin levels may be elevated, although usually they are normal(93). This alteration evidently reflects the presence of high concentrations of circulating antibodies to TG, for an antibody concentration as high as 5.2 mg/ml has been reported.

T4 and FTI are normal or low(92). Serum TSH reflects the patient's metabolic status. However, some patients are clinically euthyroid, with normal FTI and T3 levels, but have mildly elevated TSH. Whether this "subclinical hypothyroidism" represents partial or complete compensation is a matter of debate. TPOAb, and less frequently TGAb are present in serum. High levels are diagnostic of autoimmune thyroid disease. TGAb are positive in about 80% of patients, and if both TGAb and TPOAb are measured, 97% are positive. Young patients tend to have lower and occasionally negative levels. In this age group, even low titers signify the presence of thyroid autoimmunity.

FNA can be a useful diagnostic procedure but is infrequently required, except in patients that seem to have- or have- a discreet nodule in the gland. FNA typically reveals lymphocytes, macrophages, scant colloid, and a few epithelial cells which may show Hurthle cell change. In this context Hurthle cells do not represent a discrete adenoma. However if only abundant Hurthle cells dominate the specimen, and there are few or no lymphocytes or macrophages, the biopsy must be interpreted as a possible Hurthle cell tumor. Biopsy results are less frequently diagnostic in children(95).

Thyroid isotope scan is not usually necessary, but can be helpful. The image is characteristically that of a diffuse or mottled uptake in an enlarged gland, in striking contrast to the focal "cold" and "hot" areas of multinodular goiter. Focal loss of isotope accumulation may occur in severely diseased portions of the thyroid.

Table 2. Guideline for the diagnosis of Hashimoto's thyroiditis (Chronic thyroiditis)

* Some clinicians don't use the term Hashimoto's thyroiditis if patients have no goiter, although association of positive antibodies and lymphocytic infiltration in the thyroid gland was proved by histological examination.

1.     Clinical findings Diffuse swelling of the thyroid gland without any other cause (such as Graves' disease)

2.     Laboratory findings

a.     Positive for anti-thyroid microsomal antibody or anti-thyroid peroxidase(TPO) antibody

b.     Positive for anti-thyroglobulin antibody

c.      Lymphocytic infiltration in the thyroid gland confirmed with cytological examination

1.     A patient shall be said to have Hashimoto's thyroiditis if he/she has satisfied clinical criterion and any one laboratory criterion.Notes

a.     A patients shall be suspected to have Hashimoto's thyroiditis, if he/she has primary hypothyroidism without any other cause to induce hypothyroidism.

b.     A patient shall be suspected to have Hashimoto's thyroiditis, if he/she has anti-thyroid microsomal antibody and/or anti-thyroglobulin antibody without thyroid dysfunction nor goiter formation.*

c.      If a patient with thyroid neoplasm has anti-thyroid antibody by chance, he or she should be considered to have Hashimoto's thyroiditis.

d.     A patient is possible to have Hashimoto's thyroiditis if hypoechoic and/or inhomogeneous pattern is observed in thyroid ultrasonography.

Ultrasound may display an enlarged gland with normal texture, a characteristic picture with very low echogenicity, or a suggestion of multiple ill-defined nodules. Diagnostic guidelines made by The Japan Thyroid Association are shown in Table 8-2. The flow chart of diagnosis is shown in Figure 8-7.The incidental finding of diffusely increased (18)F-FDG uptake in the thyroid gland is mostly associated with chronic lymphocytic (Hashimoto's) thyroiditis and does not seem to be affected by thyroid hormone therapy (95.1).

DIFFERENTIAL DIAGNOSIS

Hashimoto's thyroiditis is to be distinguished from nontoxic nodular goiter or Graves' disease. The presence of gross nodularity is strong evidence against Hashimoto's thyroiditis, but differentiation on this basis is not infallible. In multinodular goiter, thyroid function test results are usually normal, and the patient is only rarely clinically hypothyroid. Thyroid autoantibodies tend to be absent or titers are low, and the scan result is typical. FNA can resolve the question but is usually unnecessary. In fact, the two conditions quite commonly occur together in adult women. Whether this is by chance, or due to the effect of thyroid growth stimulating antibodies (or other causes) is unknown.

Moderately and diffusely enlarged thyroid glands in teenagers are usually the result of thyroiditis, but some may be true adolescent goiters; that is, the enlargement may result from moderate hyperplasia of the thyroid gland in response to a temporarily increased demand for hormone. This condition is more often diagnosed than proved. Thyroid function test results should be normal. Antibody assays may resolve the issue. The diagnosis can be settled with certainty only by a biopsy disclosing normal or hyperplastic thyroid tissue and absence of findings of thyroiditis. The possibility of colloid goiter may be entertained in the differential diagnosis. Colloid goiter is a definite pathologic entity, as described in Chapter 17. Presumably it is the resting phase after a period of thyroid hyperplasia.

Tumor must also be considered in the differential diagnosis, especially if there is rapid growth of the gland or persistent pain. The diffuse nature of autoimmune thyroiditis, the characteristic hypothyroidism and involvement of the pyramidal lobe are usually sufficient for differentiation. FNA is indicated if there is uncertainty. However, it must be remembered that lymphoma or a small-cell carcinoma of the thyroid can be and has been mistaken for Hashimoto's thyroiditis. Clusters of nodes at the upper poles strongly suggesting papillary cancer may disappear when thyroid hormone replacement therapy is given. However, we have seen a sufficient number of patients with both thyroiditis and tumor to know that one diagnosis in no way excludes the other. Thyroid lymphoma must always be considered if there is continued (especially asymmetric) enlargement of a Hashimoto's gland, or if pain, tenderness, hoarseness, or nodes develop. Thyroiditis is a risk factor for thyroid lymphoma, although the incidence is very low. Thyroid lymphoma develops in most cases in glands which harbor thyroiditis. Distinguishing thyroid lymphoma from Hashimoto's thyroiditis is sometimes quite difficult Reverse transcription-polymerase chain reaction (RT-PCR) detecting the monoclonality of immunoglobulin heavy chain mRNA is useful for differentiation between the two(99). This condition and its management are discussed in Chapter 18.

Occasionally the picture of Hashimoto's thyroiditis blends rather imperceptibly into that of thyrotoxicosis, and some patients have symptoms of mild thyrotoxicosis, but then develop typical Hashimoto's thyroiditis. In fact, it is best to think of Graves' disease and Hashimoto's thyroiditis as two very closely related syndromes produced by thyroid autoimmunity. Categorization depends on associated eye findings and the metabolic level, but the pathogenesis, histologic picture, and function may overlap.

Likewise, we have seen patients who appear to have a mixture of Hashimoto's thyroiditis and subacute thyroiditis, with goiter, positive thyroid autoantibodies, normal or low FT4, and biopsies which have suggested Hashimoto's on one occasion and included giant cells on another. A form of painful chronic thyroiditis with amyloid infiltration has also been described, and is probably etiologically distinct from Hashimoto's thyroiditis(100).

THERAPY

Many patients need no treatment, for frequently the disease is asymptomatic and the goiter is small. This approach is justified by the study of Vickery and Hamlin(101), who found, on both clinical and pathologic grounds, that the disease may remain static and the clinical condition unchanged over many years.

If the goiter is a problem because of local pressure symptoms, or is unsightly, thyroid hormone therapy is indicated. Thyroid hormone often causes a gratifying reduction in the size of the goiter after several months of treatment(100). We have been especially impressed with this result in young people. It seems likely that in older patients there may be more fibrosis and therefore less tendency for the thyroid to shrink. In young patients the response often occurs within 2 - 4 weeks, but in older ones the thyroid decreases in size more gradually. Aksoy et al (100a) report that "prophylactic" thyroid hormone treatment is associated after 15 months with a decrease in thyroid size and in thyroid antibody levels. Thyroid hormone in a full replacement dose is, of course, indicated if hypothyroidism is present. Therapy is probably indicated if the TSH level is elevated and the FT4 is low normal, since the onset of hypothyroidism is predictable in such patients. There is no evidence that thyroid replacement actually halts the ongoing process of thyroiditis, but in some patients receiving treatment, antibody levels gradually fall over many years(102).

Figure 7. Diagnosis of Hashimoto’s thyroiditis (chronic thyroiditis)

The dosage of thyroxine should normally be that required to bring the serum TSH level to the low normal range, such as .3 - 1 uU/ml. This is typically achieved with 1 ug L-T4/lb body weight/day, ranges from 75 - 125 ug/day in women, and 125 - 200 ug/day in men. It is sensible to initiate therapy with a partial dose, since in some instances the thyroid gland may be nonsuppressible even though functioning at a level below normal. Once thyroxine treatment is initiated, it is required indefinitely in most patients. However, it has been found that up to 20% of initially hypothyroid individuals will later recover and have normal thyroid function if challenged by replacement hormone withdrawal. This may represent subsidence of cytotoxic antibodies, modulation of TSBAb, or some other mechanism(22). These individuals can be identified by administration of TRH, which will induce an increase in serum T4 and T3 if the thyroid has recovered(103). Replacement T4 therapy should be taken several hours before or after medications such as cholesterol binding resins, carafate, and FSO4, which can reduce absorption(104). (See Chapter 9) Autoimmune disease is usually takes an ongoing process and Hashimoto's thyroiditis develops into hypothyroidism. Recent trial of proplylactic treatment with T4 (1.0 ~ 2.0µg/Kg/day) for one year in euthyroid patients with Hashimoto's thyroiditis showed decrease of anti-TPO antibodies and thyroid B-lymphocytes(105), suggesting prophylactic T4 therapy might be useful to stop progression of disease. The long-term clinical benefit should be established in the future.Whether or not subclinical hypothyroidism should be treated is still under debate (see Chapter 9.10 SUBCLINICAL HYPOTHYROIDISM). Cardiac dysfunction may be associated with subclinical hypothyroidism, even when serum TSH is still in the normal range. These abnormalities are reversible with l-T4 replacement therapy (22-1).

In some instances the acute onset of the disease, in association with pain, has prompted therapy with glucocorticoids. This treatment alleviates the symptoms and improves the associated biochemical abnormalities, and in some studies has been shown to increase plasma T3 and T4 levels by suppression of the autoimmune process(106). Blizzard and co-workers(107) have given steroids over several months to children in an attempt to suppress antibody production and possibly to achieve a permanent remission. The adrenocortical hormones dramatically depress clinical activity of the disease and antibody titers, but all return to pre-therapy levels when treatment is withdrawn. We cannot recommend steroid therapy for this condition because of the undesirable side effects of the drug. Chloroquine has been reported in one study to reduce antibody titers(108). Because of toxicity, its use is not advised. X-ray therapy also results in a decrease in goiter size, and frequently in myxedema, but should not be used because of the possible induction of thyroid carcinoma.

SELENIUM- In a randomized prospective controlled study, 77 TPO+ pregnant women received 200 ug selenomethionine daily starting at the 12th week of pregnancy, and 74 TPO+ women received a placebo. The treated group had significantly lower TPO antibody levels at the end of pregnancy and during the post-partum while on treatment. The incidence of PPT was reduced from 48.6 to 28.6% in the treated group, and the incidence of permanent hypothyroidism was equivalently reduced. Thyroid hormone levels did not differ. This one report is certainly most interesting, but needs confirmation before this treatment can be suggested for general application (108.1). Confirming earlier studies, in Hashimoto’s patients, 200 mug Se in the form of l-selenomethionine orally for 6 months caused a significant decrease of 21% in serum anti-TPO levels. Cessation caused an increase in the anti-TPO concentrations.(108.2). A slightly opposing study, however, was reported no immunological benefit of selenium in patients with moderate disease activity (in terms of TPOAb and cytokine production patterns) may not (equally) benefit as patients with high disease activity (108.3). Selenium responsiveness may be different among patients with Hashimoto’s thyroiditis. A systematic review and meta-Analysis revealed that  selenium supplementation reduced serum TPOAb levels after 3, 6, and 12 months in an LT4-treated Hashimoto’s population, and after three months in an untreated population (108.4). However, no effect of selenium supplementation on thyroid stimulating hormone, health-related quality of life or thyroid ultrasound was found in levothyroxine substitution-untreated individuals, and sporadic evaluation of  clinically  relevant  outcomes  in  levothyroxine substitution-treated patients (108.5). Future well-powered RCTs, evaluating e.g. disease progression or health-related quality of life, are warranted before determining the relevance of selenium supplementation in autoimmune thyroiditis. Further, combined treatment with Myo-inositol and selenium was reported that the beneficial effects obtained by selenomethionine treatment on patients affected by subclinical hypothyroidism were further improved by cotreatment with Myo-Inositol (108.6). Myo-Inositol s an isomer of a C6 sugar alcohol an plays an important role in several cellular processes. In particular, it has been demonstrated that Myo-Inositol is the precursor for the synthesis of phosphoinositides, which are part of the phosphatidylinositol (PtdIns) signal transduction pathway. In one study. the administration of myo- inositol plus selenium has been reported to be effective in decreasing TSH, TPOAb, and TgAb levels, as well as enhancing thyroid hormones and personal wellbeing, therefore restoring euthyroidism in patients diagnosed with Hashimoto’s thyroiditis (108.7).

Anatabine- Anatabine, an alkaloid found in Solanaceae plants including tobacco, has been reported to ameliorate a mouse model of Hashimoto's thyroiditis. (108.8). In a double-blind, randomized, placebo-controlled multi-site study for three months, anatabine treated patients had a significant reduction in absolute serum TgAb levels from baseline by study end relative to those on placebo (p=0.027) (108.9). Further studies are warranted to dissect longer-term effects and possible actions of anatabine on the course of Hashimoto's thyroiditis.

Surgery has been used as a method of therapy. This treatment, of course, removes the goiter but usually results in hypothyroidism. We believe that it is not indicated unless significant pain, cosmetic, or pressure symptoms remain after a fair trial of thyroid therapy, and probably steroid therapy, but is appropriate in some cases. Among patients with postpartum thyroid dysfunction, the most common type is destructive thyrotoxicosis and simple symptomatic treatment, using beta-adrenergic--antagonists, is usually sufficient(109). In the case of postpartum hypothyroidism, replacement with a submaximal dose of T3 is useful to relieve symptoms more quickly and to predict spontaneous recovery which is detected by an increase of T4.

Some patients do not fit easily into the usual diagnostic categories; accordingly, choosing an appropriate course of therapy is more difficult. Frequently, it is impossible to differentiate Hashimoto's thyroiditis from multinodular goiter short of performing an open biopsy. In these cases, if there is no suggestion of carcinoma, it is logical to treat the patient with hormone replacement and to observe closely. A reduction in the goiter justifies continuation of the therapy, even in the absence of a diagnosis.

In some patients, especially teenagers, the examination discloses peri-thyroidal lymph nodes or an apparent discrete nodule, in addition to the diffusely enlarged thyroid of Hashimoto's thyroiditis. Such nodules should be evaluated by FNA, ultrasound and possibly scintiscan. Thyroid hormone treatment may cause regression of the nodes or nodule. If after full evaluation uncertainty persists, if nodes remain present, or if a nodule grows, surgical exploration is indicated.

Treatment of children and adolescents with 1.3ug/kg/day thyroxine for 24 months was shown in a recent study to cause significant reduction in thyroid size in patients with Autoimmune thyroiditis, but not affect antibody levels, or significantly alter TSH or freeT4. (110)

Occasionally, symptoms of serositis or arthritis suggest the coincident occurrence of another autoimmune disorder. We have given thyroid hormone to decrease thyroid activity and possibly reduce a tendency to antibody formation, and have treated the generalized disorder independently as indicated.

 

SUMMARY

Hashimoto's thyroiditis is characterized clinically as a commonly occurring, painless, diffuse enlargement of the thyroid gland occurring predominantly in middle-aged women. The patients are often euthyroid, but hypothyroidism may develop. The thyroid parenchyma is diffusely replaced by a lymphocytic infiltrate and fibrotic reaction; frequently, lymphoid germinal follicles are visible. Attention has been focused on this process because of the demonstration of autoimmune phenomena in most patients. Persons with Hashimoto's thyroiditis have serum antibodies reacting with TG, TPO, and against an unidentified protein present in colloid. In addition, many patients have cell mediated immunity directed against thyroid antigens, demonstrable by several techniques. Cell mediated immunity is also a feature of experimental thyroiditis induced in animals by injection of thyroid antigen with adjuvants.

All theories also emphasize a basic abnormality in the immune surveillance system, which in some way allows autoimmunity to develop against thyroid antigens, and as well against other tissues, including stomach, adrenal, and ovaries, in many patients with thyroiditis.

We suggest that Hashimoto's thyroiditis, primary myxedema, and Graves' disease are different expressions of a basically similar autoimmune process, and that the clinical appearance reflects the spectrum of the immune response in the particular patient. This response may include cytotoxic antibodies, stimulatory antibodies, blocking antibodies, or cell mediated immunity. Thyrotoxicosis is viewed as an expression of the effect of circulating thyroid stimulatory antibodies. Hashimoto's thyroiditis is predominantly the clinical expression of cell mediated immunity leading to destruction of thyroid cells, which in its severest form produces thyroid failure and idiopathic myxedema.

The clinical disease is more frequent than Graves' Disease when mild cases are included. The incidence is on the order of three to six cases per 10,000 population per year, and prevalence among women is at least 2%.

The gland involved by thyroiditis tends to lose its ability to store iodine, produces and secretes iodoproteins that circulate in plasma, and is inefficient in making hormone. Thus, the thyroid gland is under increased TSH stimulation, fails to respond to exogenous TSH, and has a rapid turnover of thyroidal iodine.

Diagnosis is made by the finding of a diffuse, smooth, firm goiter in a young woman, with strongly positive titers of TG Ab and/or TPO Ab and a euthyroid or hypothyroid metabolic status. A patient with a small goiter and euthyroidism does not require therapy unless the TSH level is elevated. The presence of a large gland, progressive growth of the goiter, or hypothyroidism indicates the need for replacement thyroid hormone. Surgery is rarely indicated. Development of lymphoma, though very unusual, must be considered if there is growth or pain in the involved gland.

References

  1. Hashimoto H. Zur Kenntniss der lymphomatosen Veranderung der Schilddruse (struma lymphomatosa), Arch Klin Chir 97:219, 1912.
  2. McConahey WM, Keating FR Jr, Beahrs OH, Woolner LB. On the increasing occurrence of Hashimoto's thyroiditis. J Clin Endocrinol Metab 22:542, 1962.
  3. Fromm GA, Lascano EF, Bur GE, Escalenta D. Tiroiditis cronica inespecifica. Rev Assoc Med Arg 67:162, 1953.
  4. Luxton RW, Cooke RT. Hashimoto's struma lymphomatosa: Diagnostic value and significance of serum-flocculation reactions. Lancet 2:105, 1956.
  5. Rose NR, Witebsky E. Studies on organ specificity. V. Changes in thyroid glands of rabbits following active immunization with rabbit thyroid extracts. J Immunol 76:417, 1956.
  6. Roitt IM, Doniach D, Campbell PN, Hudson RV. Auto- antibodies in Hashimoto's thyroiditis (lymphadenoid goiter). Lancet 2:820, 1956.
  7. Totterman TH, Maenpaa J, Gordin A, Makinen T, Andersson AC. Blood and thyroid-infiltrating lymphocyte subclasses in juvenile autoimmune thyroiditis. Clin exp Immunol 30:193, 1977.
  8. Paolieri F, Pronzato C, Battifora M, Fiorino N, Canonica GW, Bagnasco M. Infiltrating gamma/delta T-cell receptor-positive lymphocytes in Hashimoto's thyroiditis, Graves' disease and papillary thyroid cancer. J Endocrinol Invest. 18 : 295-8, 1995.
  9. Iwatani Y, Hidaka Y, Matsuzuka F, Kuma K, Amino N. Intrathyroidal lymphocyte subsets, including unusual CD4+ CD8+ cells and CD3loTCR alpha beta lo/-CD4-CD8- cells, in autoimmune thyroid disease. Clin Exp Immunol. 93: 430-6, 1993.
  10. Davies TF, Martin A, Concepcion ES, Graves P, Cohen L, Ben Nun A. Evidence of limited variability of antigen receptors on intrathyroidal T cells in autoimmune thyroid disease. N Engl J Med. 325: 238-44, 1991.
  11. Davies TF, Concepcion ES, Ben Nun A, Graves PN, Tarjan,G. T-cell receptor V gene use in autoimmune thyroid disease: direct assessment by thyroid aspiration. J Clin Endocrinol Metab. 76: 660-6, 1993.
  12. Heuer M, Aust G, Ode-Hakim S, Scherbaum WA. Different cytokine mRNA profiles in Graves' disease, Hashimoto's thyroiditis, and nonautoimmune thyroid disorders determined by quantitative reverse transcriptase polymerase chain reaction (RT-PCR). Thyroid. 6: 97-106, 1996.
  13. Gunn A, Michie W, Irvine WJ. The thymus in thyroid disease. Lancet 2:776, 1964.
  14. Michie W, Beck JS, Mahaffy RG, Honein EF, Fowler GB. Quantitative radiological and histological studies of the thymus in thyroid disease. Lancet 1:691, 1967.
  15. Mizukami Y, Michigishi T, Hashimoto T, Tonami N, Hisada K, Matsubara F, Takazakura E. Silent thyroiditis: a histologic and immunohistochemical study. Hum Pathol 19:423-431, 1988.

15a. Zeitlin AA, Heward JM, Newby PR, Carr-Smith JD, Franklyn JA, Gough SC, Simmonds MJ. Analysis of HLA class II genes in Hashimoto's thyroiditis reveals differences compared to Graves' disease. Genes Immun. 2008 Jun;9(4):358-63

  1. Brix TH, Kyvik KO, Hegedus L: A population-based study of chronic autoimmune hypothyroidism in Danish twins. J Clin Endocr Metab 85:536-539, 2000

16a. Simmonds MJ. GWAS in autoimmune thyroid disease: redefining our understanding of pathogenesis. Nat Rev Endocrinol. 2013 May;9(5):277-87. doi: 10.1038/nrendo.2013.56. Epub 2013 Mar 26.

16b. Cai T, Li J, An X, Yan N, Li D, Jiang Y, Wang W, Shi L, Qin Q, Song R, Wang G, Jiang W, Zhang JA. Polymorphisms in MIR499A and MIR125A gene are associated with autoimmune thyroid diseases. Mol Cell Endocrinol. 2017 Jan 15;440:106-115

16.1 Duntas LH. Environmental factors and autoimmune thyroiditis. Nat Clin Pract Endocrinol Metab. 2008 Jul 8. [Epub ahead of print]

  1. Mori T, Kriss JP. Measurements by competitive binding radioassay of serum antimicrosomal and anti-thyroglobulin antibodies in Graves' disease and other thyroid disorders. J Clin Endocrinol Metab 33:688, 1971.
  2. Kawakami Y, Fisfalen M-E, DeGroot LJ. Proliferative responses of peripheral blood mononuclear cells from patients with autoimmune thyroid disease to synthetic peptide epitopes of human thyroid peroxidase. Autoimmunity, 13:17-26, 1992.

18.1 Ng HP, Kung AW Induction of autoimmune thyroiditis and hypothyroidism by immunization of immunoactive T cell epitope of thyroid peroxidase. Endocrinology. 2006 Jun;147(6):3085-92

18.2 Muixí L, Carrascal M, Alvarez I, Daura X, Martí M, Armengol MP, Pinilla C, Abian J, Pujol-Borrell R, Jaraquemada D. Thyroglobulin peptides associate in vivo to HLA-DR in autoimmune thyroid glands. J Immunol. 2008 Jul 1;181(1):795-807

  1. Hidaka Y, Amino N, Iwatani Y, Kaneda T, Nasu M, Mitsuda N, Tanizawa O, Miyai K Increase in peripheral natural killer cell activity in patients with autoimmune thyroid disease. Autoimmunity 11:239, 1992.

19.1 Itoh M, Takahashi T, Sakaguchi N, Kuniyasu Y, Shimizu J, Otsuka F, Sakaguchi S 1999 Thymus and Autoimmunity: Production of CD25+CD4+ naturally anergic and suppressive T Cells as a key function of the thymus in maintaining immunologic self-tolerance. J Immunol 162: 5317-5326

19.2. McLachlan SM, Nagayama Y, Pichurin PN, Mizutori Y, Chen CR, Misharin A, Aliesky HA, Rapoport B. The link between Graves' disease and Hashimoto's thyroiditis: a role for regulatory T cells. Endocrinology. 2007 Dec;148(12):5724-33

  1. Kosugi S, Ban T, Akamizu T, Kohn LD. Identification of separate determinants on the thyrotropin receptor reactive with Graves' thyroid-stimulating antibodies and with thyroid-stimulating blocking antibodies in idiopathic myxedema: These determinants have no homologous sequence on gonadotropin receptors. Molecul Endocrinol 6:168-180, 1992.
  2. Tamaki H, Amino N, Kimura M, Hidaka Y, Takeoka K, Miyai K. Low prevalence of thyrotropin receptor antibody in primary hypothyroidism in Japan. J Clin Endocrinol Metab 71:1382, 1990.
  3. Okamura K, Sato K, Yoshinari M, Ikenoue H, Kuroda T, Nakagawa M, Tsuji H, Washio M, Fujishima M. Recovery of the thyroid function in patients with atrophic hypothyroidism and blocking type TSH binding inhibitor immunoglobulin. Acta Endocrinol (Copenh) 122:107-114, 1990.

22-1. Mariotti S, Zoncu S, Pigliaru F, Putzu C, Cambuli VM, Vargiu S, Deidda M, Mercuro G. Cardiac effects of l-thyroxine administration in borderline hypothyroidism. Int J Cardiol. 2007 May 9; [Epub ahead of print]

  1. Takasu N, Yamada T, Takasu M, Komiya I, Nagasawa Y, Asawa T, Shinoda T, Aizawa T, Koizumi Y. Disappearance of thyrotropin-blocking antibodies and spontaneous recovery from hypothyroidism in autoimmune thyroiditis. N Engl J Med 326:513-518, 1992.
  2. Mori T, Akamizu T, Kosugi S, Sugawa H, Inoue D, Okuda J, Ueda Y. Recent progress in TSH receptor studies with a new concept of "Autoimmune TSH receptor disease". Endocr J 41:1-11, 1994.
  3. Kotani T, Aratake Y, Hirai K, Fukazawa Y, Sato H, Ohtaki S. Apoptosis in thyroid tissue from patients with Hashimoto's thyroiditis. Autoimmunity 20:231, 1995.
  4. Giordano C, Stassi G, Maria R, Todaro M, Richiusa P, Papoff G, Ruberti G, Bagnasco M, Testi R, Galluzzo A. Potential involvement of Fas and its ligand in the pathogenesis of Hashimoto's thyroidiitis. Science 275:960, 1997.
  5. Mitsiiades N, Poulaki V, Kotoula V, Mastorakos G, Balafouta S, Koutras DA, Tsokos M.Fas/Fas ligand up-regulation and BCL-2 down-regulation may be significant in thepathogenesis of Hashimoto' thyroiditis. J Clin Endo Metab 83:2199,1998.
  6. Kawakami A, Eguchi K, Matsuoka N, Tsuboi M, Kawabe Y, Ishikawa N, Ito K, Nagataki S.Thyroid-stimulating hormone inhiibits Fas antigen-mediated apoptosis ofhuman thyrocytes in vitro. Endocrinology 137:3163, 1996.
  7. Batteux F, Lores P, Bucchini D, Chiocchia G. Transgenic expression of Fas ligand on thyroid follicular cells prevents autoimmune thyroiditis. J Immunol 164:1681-1688, 2000.
  8. Stassi G, DeMaria R. Autoimmune thyroid disease: new models of cell death in autoimmunity. Nat Rev Immunol 2:195-204, 2002.
  9. Dong Z, Takakuwa T, Takayama H, Luo WJ, Takano T, Amino N, Matsuzuka F, Aozasa K. Fas and Fas ligand gene mutations in Hashimoto's thyroiditis. Lab Invest 82: 1611-1616, 2002.
  10. Takakuwa T, Dong Z, Takayama H, Matsuzuka F, Nagata S, Aozasa K. Frequent mutations of Fas gene
  11. Atkins MB, Mier JW, Parkinson DR, Gould JA, Berkman EM, Kaplan MM. Hypothyroidism after treatment with interleukin- 2 and lymphokine-activated killer cells. N Engl J Med 318:1557-1563, 1988.

33a Coles AJ, Wing M, Smith S, Coraddu F, Greer S, Taylor C, Weetman A, Hale G, Chatterjee VK, Waldmann H, Compston A (1999) Pulsed monoclonal antibody treatment and autoimmune thyroid disease in multiple sclerosis. Lancet 354:1691–1695

33b Hirst CL, Pace A, Pickersgill TP, Jones R, McLean BN, Zajicek JP, Scolding NJ, Robertson NP. Campath 1-H treatment in patients with aggressive relapsing remitting multiple sclerosis. J Neurol. 2008 Feb;255(2):231-8

33c Figueroa-Vega N, Alfonso-Pérez M, Benedicto I, Sánchez-Madrid F, González-Amaro R, Marazuela M. Increased circulating pro-inflammatory cytokines and Th17 lymphocytes in Hashimoto's thyroiditis. J Clin Endocrinol Metab. 2010 Feb;95(2):953-62

33d Horie I, Abiru N, Nagayama Y, Kuriya G, Saitoh O, Ichikawa T, Iwakura Y, Eguchi K. T helper type 17 immune response plays an indispensable role for development of iodine-induced autoimmune thyroiditis in nonobese diabetic-H2h4 mice. Endocrinology. 2009 Nov;150(11):5135-42

33d.1. 1. Li C, Yuan J, Zhu YF, Yang XJ, Wang Q, Xu J, He ST, Zhang JA. Imbalance of Th17/Treg in Different Subtypes of Autoimmune Thyroid Diseases. Cell Physiol Biochem. 2016;40(1-2):245-252.

33d.1.2. 2.          Bossowski A, Moniuszko M, Idźkowska E, Grubczak K, Singh P, Bossowska A, Diana T, Kahaly GJ. Decreased proportions of CD4 + IL17+/CD4 + CD25 + CD127- and CD4 + IL17+/CD4 + CD25 + CD127 - FoxP3+ T cells in children with autoimmune thyroid diseases (.). Autoimmunity. 2016 Aug;49(5):320-8.

33e. Hamano H, Kawa S, Horiuchi A, Unno H, Furuya N, Akamatsu T, Fukushima M, Nikaido T, Nakayama K, Usuda N, Kiyosawa K. High serum IgG4 concentrations in patients with sclerosing pancreatitis. N Engl J Med. 2001 Mar 8;344(10):732-8.

33f. Komatsu K, Hamano H, Ochi Y, Takayama M, Muraki T, Yoshizawa K, Sakurai A, Ota M, Kawa S. High prevalence of hypothyroidism in patients with autoimmune pancreatitis. Dig Dis Sci. 2005 Jun;50(6):1052-7

33g. Li Y, Bai Y, Liu Z, Ozaki T, Taniguchi E, Mori I, Nagayama K, Nakamura H, Kakudo K. Immunohistochemistry of IgG4 can help subclassify Hashimoto's autoimmune thyroiditis. Pathol Int. 2009 Sep;59(9):636-41.

33h. Li Y, Nishihara E, Hirokawa M, Taniguchi E, Miyauchi A, Kakudo K. Distinct clinical, serological, and sonographic characteristics of hashimoto's thyroiditis based with and without IgG4-positive plasma cells. J Clin Endocrinol Metab. 2010 Mar;95(3):1309-17

33i. Dahlgren M, Khosroshahi A, Nielsen GP, Deshpande V, Stone JH. Riedel's thyroiditis and multifocal fibrosclerosis are part of the IgG4-related systemic disease spectrum. Arthritis Care Res (Hoboken) 2010 62: 1312-1318.

33j. Kakudo K, Li Y, Taniguchi E, Mori I, Ozaki T, Nishihara E, Matsuzuka F, Miyauchi A. IgG4-related disease of the thyroid glands [Review]. Endocr J. 2011 Dec 2. [Epub ahead of print]

  1. Nystrom E, Bengtsson C, Lapidus L, Petersen K, Lindstedt G. Smoking--A risk factor for hypothyroidism. J Endocrinol Invest 16:129-131, 1993.

34a. Pacini F, Vorontsova T, Molinaro E, Kuchinskaya E, Agate L, Shavrova E, Astachova L, Chiovato L, Pinchera A. Prevalence of thyroid autoantibodies in children and adolescents from Belarus exposed to the Chernobyl radioactive fallout. Lancet. 1998 Sep 5;352(9130):763-6.

34b. Agate L, Mariotti S, Elisei R, Mossa P, Pacini F, Molinaro E, Grasso L, Masserini L, Mokhort T, Vorontsova T, Arynchyn A, Tronko MD, Tsyb A, Feldt-Rasmussen U, Juul A, Pinchera A. Thyroid Autoantibodies and Thyroid Function in Subjects Exposed to Chernobyl Fallout during Childhood: Evidence for a Transient Radiation-Induced Elevation of Serum Thyroid Antibodies without an Increase in Thyroid Autoimmune Disease. J Clin Endocrinol Metab. 2008 Jul;93(7):2729-36

34c Elfström P, Montgomery SM, Kämpe O, Ekbom A, Ludvigsson JF. Risk of Thyroid disease in individuals with Celiac disease. J Clin Endocrinol Metab. 2008 Jul 8. [Epub ahead of print]

34d Betterle C, Lazzarotto F, Presotto F. Autoimmune polyglandular syndrome Type 2: the tip of an iceberg? Clin Exp Immunol. 2004 Aug;137(2):225-33

34e Molitch ME, Gillam MP. Lymphocytic hypophysitis. Horm Res. 2007;68 Suppl 5:145-50

34f. Christian Bernecker, Luisa Lenz, Martin S. Ostapczuk, Sven Schinner, Holger Willenberg, Margret Ehlers, Stefan Vordenbäumen, Joachim Feldkamp, Matthias Schott. MicroRNAs miR-146a1, miR-155_2, and miR-200a1 Are Regulated in Autoimmune Thyroid Diseases. Thyroid. December 2012, 22(12): 1294-1295.

34g. Otsu H, Watanabe M, Inoue N, Masutani R, Iwatani Y. Intraindividual variation of microRNA expression levels in plasma and peripheral blood mononuclear cells and the associations of these levels with the pathogenesis of autoimmune thyroid diseases. Clin Chem Lab Med. 2017 May 1;55(5):626-635.

34h. Xiao C, Rajewsky K 2009 MicroRNA control in the immune system: basic principles. Cell 136: 26–36.

34i. Thai T-H, Calado DP, Casola S, Ansel KM, Xiao C, Xue Y, Murphy A, Frendewey D, Valenzuela D, Kutok JL, Schmidt-Supprian M, Rajewsky N, Yancopoulos G, Rao A, Rajewsky K 2007 Regulation of the germinal center response by microRNA-155. Science 316: 604–606

34j. Zhu J, Zhang Y, Zhang W, Zhang W, Fan L, Wang L, Liu Y, Liu S, Guo Y, Wang Y, Yi J, Yan Q, Wang Z, Huang G. MicroRNA-142-5p contributes to Hashimoto's thyroiditis by targeting CLDN1. J Transl Med. 2016 Jun 8;14(1):166.

  1. Gordin A, Maatela J, Miettinen A, Helenius T, Lamberg B-A. Serum thyrotrophin and circulating thyroglobulin and thyroid microsomal antibodies in a Finnish population. Acta Endocrinol 90:33, 1979.
  2. Ling SM, Kaplan SA, Weitzman JJ, Reed GB, Costin G, Landing BH. Euthyroid goiters in children. Correlation of needle biopsy with other clinical and laboratory findings in chronic lymphocytic thyroiditis and simple goiter. Pediatrics 44:695, 1969.
  3. Tunbridge WMG, Evered DC, Hall R, et al. The spectrum of thyroid disease in a community. The Whickham Survey. Clin Endocrinol 7:481, 1977.
  4. Inoue M, Taketani N, Sato T, Nakajima H. High incidence of chronic lymphocytic thyroiditis in apparently healthy school children: Epidemiological and clinical study. Endocrinol Jpn 22:483, 1975.
  5. Carey C, Skosey C, Pinnamaneni KM, Barsano CP, DeGroot LJ. Thyroid abnormalities in children of parents who have Graves' disease. Possible pre-Graves' disease. Metabolism 29:369, 1980.
  6. Yoshida H, Amino N, Yagawa K, Uemura K, Satoh M, Miyai K, Kumahara Y. Association of serum antithyroid antibodies with lymphocytic infiltration of the thyroid gland : studies of seventy autopsied cases. J Clin Endocrinol Metab 46:859, 1978.
  7. Gordin A, Saarinen P, Pelkonen A, Lamberg B-A. Serum thyroglobulin and the response to thyrotropin releasing hormone in symptomless autoimmune thyroiditis and in borderline and overt hypothyroidism. Acta Endocrinol 75:274, 1974.
  8. Tunbridge WMG, Brewis M, French JM, Appleton D, Bird T, Clark F, Evered DC, Evans JG, Hall R, Smith P, Stephenson J, Young E. Natural history of autoimmune thyroiditis. Br Med J 282:258, 1981.

42a. Simmonds MJ, Kavvoura FK, Brand OJ, Newby PR, Jackson LE, Hargreaves CE, Franklyn JA, Gough SC. Skewed X chromosome inactivation and female preponderance in autoimmune thyroid disease: an association study and meta-analysis. J Clin Endocrinol Metab. 2014 Jan;99(1):E127-31

42b. Klintschar, M, , Schwaiger, P, , Mannweiler, S, , Regauer, S, , Kleiber, M, 2001 Evidence of fetal microchimerism in Hashimoto's thyroiditis. J Clin Endocrinol Metab 86:2494–2498

42c. Klintschar, M, , Immel, UD, , Kehlen, A, , Schwaiger, P, , Mustafa, T, , Mannweiler, S, , Regauer, S, , Kleiber, M, , Hoang-Vu, C, 2006 Fetal microchimerism in Hashimoto's thyroiditis: a quantitative approach. Eur J Endocrinol 154:237–241

42d. Renné, C, , Ramos Lopez, E, , Steimle-Grauer, SA, , Ziolkowski, P, , Pani, MA, , Luther, C, , Holzer, K, , Encke, A, , Wahl, RA, , Bechstein, WO, , Usadel, KH, , Hansmann, ML, , Badenhoop, K, 2004 Thyroid fetal male microchimerism in mothers with thyroid disorders: presence of Y-chromosomal immunofluorescence in thyroid infiltrating lymphocytes is more prevalent in Hashimoto's thyroiditis and Graves' disease than in follicular adenomas. J Clin Endocrinol Metab 89:5810–5814

  1. Buchanan WW, Harden RM. Primary hypothyroidism and Hashimoto's thyroiditis. Arch Intern Med 115:411, 1965.
  2. Moriuchi A, Yokoyama S, Kashima K, Andoh T, Nakayama I, Noguchi S. Localized primary anyloid tumor of the thyroid developing in the course of Hashimoto's thyroiditis. Acta Pathologica Japonica 42:210-216, 1992.
  3. Shaw PJ, Walls TJ, Newman PK, Cleland PG, Cartlidge NE. Hashimoto's encephalopathy: A steroid-responsive disorder associated with high antithyroid antibody titers -- report of five cases. Neurology 41:228-233, 1991.
  4. Khardori R, Eagleton LE, Soler NG, McConnachie PR. Lymphocytic interstitial pneumonitis in autoimmune thyroid disease. Amer J Med 90:649-652, 1991.

46-1. Radetti G, Gottardi E, Bona G, Corrias A, Salardi S, Loche S; Study Group for Thyroid Diseases of the Italian Society for Pediatric Endocrinology and Diabetes (SIEDP/ISPED).The natural history of euthyroid Hashimoto's thyroiditis in children. J Pediatr. 149:827-32, 2006.

  1. Eisenbart GS, Wilson PW, Ward F, Buckley C, Lebovitz H. The polyglandular failure syndrome. Disease inheritance, HLA type, and immune function. Ann Intern Med 91:528, 1979.
  2. Loviselli A, Mathieu A, Pala R, Mariotti S, Cau S, Marongiu C, Mazzoleni AP, Maggio P, Martino E. Development of thyroid disease in patients with primary and secondary Sjogren's syndrome. J Endocrinol Invest 11:653, 1988.
  3. Best TB, Munro RE, Burwell S, Volpe R. Riedel's thyroiditis associated with Hashimoto's thyroiditis, hypoparathyroidism, and retroperitoneal fibrosis. J Endocrinol Invest 14:767- 772, 1991.
  4. Becker KL, Ferguson RH, McConahey WM. The connective-tissue diseases and symptoms associated with Hashimoto's thyroiditis. N Engl J Med 268:277, 1963.
  5. Bastenie PA, Vanhaelst L, Golstein J, Smets P, Keys MJ, Karvonen MJ, Punsar S. Asymptomatic autoimmune thyroiditis and coronary heart-disease. Lancet 1:155, 1977.
  6. Heinonen OP, Aho K, Pyorala K, Gordin A, Punsar S, Puro K. Symptomless autoimmune thyroiditis in coronary heart disease. Lancet 1:785, 1972.

52a. Jankovic B, Le KT, Hershman JM. Clinical Review: Hashimoto's thyroiditis and papillary thyroid carcinoma: is there a correlation? J Clin Endocrinol Metab. 2013 Feb;98(2):474-82.

52b. Lee JH, Kim Y, Choi JW, Kim YS. The association between papillary thyroid carcinoma and histologically proven Hashimoto's thyroiditis: a meta-analysis. Eur J Endocrinol. 2013 Feb 15;168(3):343-9.

52c. Dvorkin S, Robenshtok E, Hirsch D, Strenov Y, Shimon I, Benbassat CA. Differentiated thyroid cancer is associated with less aggressive disease and better outcome in patients with coexisting Hashimotos thyroiditis. J Clin Endocrinol Metab. 2013 Jun;98(6):2409-14

52d. Marotta V, Guerra A, Zatelli MC, Uberti ED, Di Stasi V, Faggiano A, Colao A, Vitale M. BRAF mutation positive papillary thyroid carcinoma is less advanced when Hashimoto's thyroiditis lymphocytic infiltration is present. Clin Endocrinol (Oxf). 2013 Nov;79(5):733-8

52-1. Larson SD, Jackson LN, Riall TS, Uchida T, Thomas RP, Qiu S, Evers BM. Increased incidence of well-differentiated thyroid cancer associated with Hashimoto thyroiditis and the role of the PI3k/Akt pathway. J Am Coll Surg. 2007 May; 204 (5):764-73

  1. Hall R, Turner-Warwick M, Doniach D. Autoantibodies in iodide goiter and asthma. Clin Exp Immunol 1:285, 1966.
  2. Amino N, Miyai K, Onishi T, Hashimoto T, Arai K, Ishibashi K, Kumahara Y. Transient hypothyroidism after delivery in autoimmune thyroiditis. J Clin Endocrinol Metab 42:296, 1976.
  3. Amino N, Kuro R, Tanizawa O, Tanaka F, Hayashi C, Kotani K, Kawashima M, Miyai K, Kumahara Y. Changes of serum antithyroid antibodies during and after pregnancy in autoimmune thyroid diseases. Clin Exp Immunol 31:30, 1978.
  4. Amino N, Miyai K, Kuro R, Tanizawa O, Azukizawa M, Takai S, Tanaka F, Nishi K, Kawashima M, Kumahara Y. Transient postpartum hypothyroidism. Fourteen cases with autoimmune thyroiditis. Ann Intern Med 87:155, 1977.

56a. Abalovich M, Amino N, Barbour LA, Cobin RH, De Groot LJ, Glinoer D, Mandel SJ, Stagnaro-Green A. Management of thyroid dysfunction during pregnancy and postpartum: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2007 92; Aug (8 Suppl):S1-47.

56b. Dosiou C, Sanders GD, Araki SS, Crapo LM. Screening pregnant women for autoimmune thyroid disease: a cost-effectiveness analysis. Eur J Endocrinol. 2008 Jun;158(6):841-51.

  1. Amino N, Tada H, Hidaka Y. Postpartum autoimmune thyroid syndrome: a model of aggravation of autoimmune disease. Thyroid 9: 705-713, 1999.
  2. Roti E, Emerson CH. Clinical Review 29. Postpartum thyroiditis. J Clin Endocrinol Metab 74:3-5, 1992.
  3. Gluck FB, Nusynowitz ML, Plymate S. Chronic lymphocytic thyroiditis, thyrotoxicosis, and low radioactive iodine uptake: Reports of four cases. N Engl J Med 292:624, 1975.
  4. Woolf PD, Daly R: Thyrotoxicosis with painless thyroiditis. Am J Med 60: 73-79, 1976.
  5. Hamburger JI: Occult subacute thyroiditis: Diagnostic challenge. Mich Med 70: 1125- , 1976.
  6. Nikolai TF, Brosseau J, Kettrick MA, Roberts R, Beltaos E. Lymphocytic thyroiditis with spontaneously resolving hyperthyroidism (silent thyroiditis). Arch Intern Med 140:478, 1980.
  7. Dorfman SG, Copperman MT, Nelson RL, Depuy H, Peake RL, Young RL. Painless thyroiditis and transient hyperthyroidism without goiter. Ann Intern Med 86:24, 1977.
  8. Woolf PD. Transient painless thyroiditis with hyperthyroidism. A variant of lymphocytic thyroiditis. Endocrine Reviews 1:411, 1980.
  9. Gorman CA, Duick DS, Woolner LB, Wahner HW: Transient hyperthyroidism in patients with lymphocytic thyroiditis. Mayo Clin Proc 53: 359-365, 1978.
  10. Amino N, Yabu Y, Miyai K, Fujie T, Azukizawa M, Onishi T, Kumahara Y: Differentiation of thyrotoxicosis induced by thyroid destruction from Graves' disease. Lancet 2: 344-346, 1978.

66-1. Ota H, Amino N, Morita S, Kobayashi K, Kubota S, Fukata S, Kamiyama N, Miyauchi A.Quantitative measurement of thyroid blood flow for differentiation of painless thyroiditis from Graves' disease. Clin Endocrinol (Oxf). 67:41-5, 2007

  1. Nikolai TF, Coombs GJ, McKenzie AK, Miller RW, Weir Jr, GJ. Treatment of lymphocytic thyroiditis with spontaneously resolving hyperthyroidism (silent thyroiditis). Arch Intern Med 142:2281-2283, 1982.
  2. Inada M, Nishikawa M, Naito K, Ishii H, Tanaka K, Imura H. Reversible changes of the histological abnormalities of the thyroid in patients with painless thyroiditis. J Clin Endocrinol Metab 52:431, 1981.
  3. Taylor HC, Sheeler LR. Recurrence and heterogeneity in painless thyrotoxic lymphocytic thyroiditis. Report of five cases. J Amer Med Assn 248:1085-1088, 1982.
  4. Amino N, Tada H, Hidaka Y. Autoimmune thyroid disease and pregnancy. J Endocrinol Invest 19:59,1996.
  5. Yamamoto M, Shibuya N, Chen LC, Ogata E: Seasonal recurrence of transient hypothyroidism in a patient with autoimmune thyroiditis. Endocr J 35: 135-142, 1988.
  6. Tachi J, Amino N, Miyai K: Massage therapy on neck: a contributing factor for destructive thyrotoxicosis? .Thyroidology 2: 25-27, 1990.
  7. Maruyama H, Kato M, Mizuno O, Kataoka K, Matusi S: Transient thyrotoxicosis occurred after cessation of steroid therapy in a patient with autoimmune thyroiditis and rheumatoid arthritis. Endocr J 29: 583-588, 1982.
  8. Takasu N, Komiya I, Nagasawa Y, et al.: Exacerbation of autoimmune thyroid dysfunction after unilateral adrenalectomy in patients with Cushing's syndrome due to an adrenocortical adenoma. N Engl J Med 322: 1708-1712, 1990.
  9. Peschen-rosin R, Schabet m, Dichgans J. Manifestation of Hashimoto's encephalopathy years before onset of thyroid disease. Eur Neurol 41: 79-84, 1999.
  10. Brain L, Jellinek EH, Ball K. Hashimoto's disease and encephalopathy. Lancet 2:512-514, 1966.
  11. Pozo-Rosich P, Villoslada P, Canton A, Simo R, Rovira A, Montalban X. Reversible white matter alterations in encephalopathy associated with autoimmune thyroid disease. J Neurol 249:1063-1065,2002.
  12. Canton A, de Fabregas O, Tintore M, Mesa J, Codina A, Simo R. Encephalopathy associated to autoimmune thyroid disease: a more appropriate term for an underestimated condition? J Neurol Sci 176:65-69,2000.
  13. Ochi H, Horiuchi I, Araki n, Toda T, Araki T, Sato K, Murai H, Osoegawa M, Yamada T, Okamura K, Ogino T, Mizumoto K, Yamashita H, Saya H, Kira J. Proteomic analysis of human brain identifies ソ-enolase as a nobel autoantigen in Hashimoto's encephalopathy. FEBS Lett 528:197-202, 2002.
  14. Sawka AM, Fatourechi V, Boeve BF, Mokri B. Rarity of encephalopathy associated with autoimmune thyroiditis: A case series from Mayo Clinic from 1950 to 1996. Thyroid 12:393-398, 2002.

80.1 Fujii A, Yoneda M, Ito T, Yamamura O, Satomi S, Higa H, Kimura A, Suzuki M, Yamashita M, Yuasa T, Suzuki H, Kuriyama M. Autoantibodies against the amino terminal of a-enolase are a useful diagnostic marker of Hashimoto's encephalopathy. J Neuroimmunol. 162:130-6, 2005.

80.2 Yoneda M, Fujii A, Ito A, Yokoyama H, Nakagawa H, Kuriyama M. High prevalence of serum autoantibodies against the amino terminal of a-enolase in Hashimoto's encephalopathy. J Neuroimmunol. 185:195-200, 2007

80.3 Seo SW, Lee BI, Lee JD, Park SA, Kim KS, Kim SH, Yun MJ. Thyrotoxic autoimmune encephalopathy: a repeat positron emission tomography study.J Neurol Neurosurg Psychiatry. 2003 Apr;74(4):504-6

80.3a. Hosoi Y, Kono S, Terada T, Konishi T, Miyajima H. J Neurol. Hashimoto's encephalopathy associated with an elevated intrathecal IgG4 level. 2013 Apr;260(4):1174-6. doi: 10.1007/s00415-013-6878-2. Epub 2013 Mar 8.

80.4 Tateno F, Sakakibara R, Kishi M, Ogawa E. Hashimoto's ophthalmopathy. Am J Med Sci. 2011 Jul;342(1):83-5

80.5 Yoshihara A, Yoshimura Noh J, Nakachi A, Ohye H, Sato S, Sekiya K, Kosuga Y, Suzuki M, Matsumoto M, Kunii Y, Watanabe N, Mukasa K, Inoue Y, Ito K, Ito K. Severe thyroid-associated orbitopathy in Hashimoto's thyroiditis. Report of 2 cases. Endocr J. 2011;58(5):343-8.

  1. Skillern PG, Crile G Jr, McCullaugh EP, Hazard JB, Lewis LA, Brown H. Struma lymphomatosa: Primary thyroid failure with compensatory thyroid enlargement. J Clin Endocrinol Metab 16:35, 1956.
  2. Paris J, McConahey WM, Tausie WN, Woolner LB, Bahn RC. The effect of iodides on Hashimoto's thyroiditis. J Clin Endocrinol Metab 21:1037, 1961.
  3. Endo T, Kaneshige M, Nakazato M, Kohgai T, Saito T, Onaya T. Autoantibody against thyroid iodide transporter in the sera from patients with Hashimoto's thyroiditis possesses iodide transport inhiibitory activity. Bioch Bioph Res Com 228:199, 1996
  4. Chin HS, Chin DK, Morgenthaler NG, Vassart G, Costagliola S. Rarity of anti- Na+/I-symporter (NIS) antibody with iodide uptake inhibiting activity in autoimmune thyroid diseases (AITD). J Clin Endocrinol Metab 85: 3937-3940, 2000.
  5. Seissler J, Wagner S, Schott M, Feldkamp J, Scherbaum WA, Morgenthaler NG. Low frequency of autoantibodies to the human Na+/I -symporter (NIS) in patients with autoimmune thyroid disease. J Clin Endocrinol Metab 85: 4630-4634, 2000.
  6. Brown TR, Zhao G, Palmer KC, Sundick RS: Thyroid injury, autoantigen availability, and the initiation of autoimmune thyroiditis. Autoimmunity 27:1-12, 1998.
  7. Laurberg P, Pedersen KM, Hreidarsson A, Sigfusson N, Iversen E, Knudsen PR: Iodine intake and the pattern of thyroid disorders: A comparative epidemiological study of thyroid abnormalities in the elderly in Iceland and in Jutland, Denmark. J Clin Endocr Metab 83:765-769, 1998.
  8. Nagata K, Takasu N, Akamine H, Ohshiro C, Komiya I, Murakami K, Suzawa A, Nomura T: Urinary iodine and thyroid antibodies in Okinawa, Yamgata, Hyogo, and Nagano, Japan: The differences in iodine intake do not affect thyroid antibody positivity. Endoc J 45:797-803, 1998.
  9. Tajiri J, Higashi K, Morita M, Umeda T, Sato T: Studies of hypothyroidism in patients with high iodine intake. J Clin Endocr Metab 63:412-417, 1986.
  10. Ruwhof C, Drexhage HA. Iodine and thyroid autoimmune disease in animal models. Thyroid 11:427-436, 2001.
  11. Dai YD, Rao VP, Carayanniois G. Enhanced iodination of thyroglobulin facilitates processing and presentation of a cryptic pathogenic peptide. J Immunol 168:5907-5911, 2002.
  12. McConahey WM, Keating FR, Butt HR, Owen CA. Comparison of certain laboratory tests in the diagnosis of Hashimoto's thyroiditis. J Clin Endocrinol Metab 21:879, 1961.
  13. Glynne A, Thomson JA. Serum immunoglobulin levels in thyroid disease. Clin Exp Immunol 12:71, 1972.
  14. Wilkin TJ, Beck JS, Hayes PC, Potts RC, Young RJ. A passive hemagglutination (TRC) inhibitor in thyrotoxic serum. Clin Endocrinol 10:507, 1979.
  15. Monteleone JA, Davis RK, Tung KSK, Ramos CV, Peden VH. Differentiation of chronic lymphocytic thyroiditis and simple goiter in pediatrics. J Pediatr 83:381, 1973.

95-1. Karantanis D, Bogsrud TV, Wiseman GA, Mullan BP, Subramaniam RM, Nathan MA, Peller PJ, Bahn RS, Lowe VJ. Clinical significance of diffusely increased 18F-FDG uptake in the thyroid gland. J Nucl Med. 48:896-901, 2007

  1. Pedersen OM, Aardal NP, Larssen TB, Varhaug JE, Myking O, Vik-Mo H: The value of ultrasonography in predicting autoimmune thyroid disease. Thyroid 10:251-259, 2000.
  2. Hoffer PB, Gottschalk A, Refetoff S. Thyroid scanning techniques. The old and the new. Curr Probl in Radiol 2:5, 1972.
  3. Jonckheer MH, VanHaelst L, DeConinck F, Michotte Y. Atrophic autoimmune thyroiditis. Relationship between the clinical state and intrathyroidal iodine as measured in vivo in man. J Clin Endocrinol Metab 53:476, 1981.
  4. Takano T, Miyauchi A, Matsuzuka F, Yoshida H, Kuma K, Amino N: Diagnosis of thyroid malignant lymphoma by reverse transcription-polymerase chain reaction detecting the monoclonality of immunoglobulin heavy chain messenger ribonucleic acid. J Clin Endocr Metab 85:671-675, 2000.
  5. McConahey WM, Woolner LB, Black BM, Keating FR, Jr. Effect of desiccated thyroid in lymphocytic (Hashimoto's) thyroiditis. J Clin Endocrinol Metab 19:45, 1959.

100a Aksoy DY, Kerimoglu U, Okur H, Canpinar H, Karaagaoglu E, Yetgin S, Kansu E,Gedik O. Effects of prophylactic thyroid hormone replacement in euthyroid Hashimoto's thyroiditis. Endocr J. 2005 Jun;52(3):337-43.

  1. Vickery AL, Hamlin E Jr: Struma lymphomatosa (Hasimoto's thyroiditis): Observations on repeated biopsies in 16 patients. N Engl J Med 264:226, 1961.
  2. Papapetrou PD, MacSween RNM, Lazarus JH, Harden R McG. Long-term treatment of Hashimoto's thyroiditis with thyroxine. Lancet 2:7786, 1972.
  3. Takasu N, Komiya I, Asawa T, Nagasawa Y, Yamada T. Test for recovery from hypothyroidism during thyroxine therapy in Hashimoto's thyroiditis. Lancet 336:1084-1086, 1990.
  4. Campbell NRC et al. Effect of ferrous sulfate on thyroid hormone replacement in hypothyroidism. Ann Int Med 117:1010-1013, 1992.
  5. Padberg S, Heller K, Usadel KH, Schumm-Draeger PM. One-year prophylactic treatment of euthyroid hashimoto's thyroiditis patients with levothyroxine: Is there a benefit? Thyroid 11: 249-255, 2001.
  6. Yamada T, Ikejiri K, Kotani M, Kusakabe T. An increase of plasma triiodothyronine and thyroxine after administration of dexamethasone to hypothyroid patients with Hashimoto's thyroiditis. J Clin Endocrinol Metab 46:784, 1981.
  7. Blizzard RM, Hung M, Chandler RW, Aceto T Jr, Kyle M, Winship T. Hashimoto's thyroiditis. Clinical and laboratory response to prolonged cortisone therapy. N Engl J Med 267:1015, 1962.
  8. Ito S, Tamura T, Nishikawa M. Effects of desiccated thyroid, prednisolone and chloroquine on goiter and antibody titer in chronic thyroiditis. Metabolism 17:317, 1968.

108.1. Negro R, Greco G, Mangieri T, Pezzarossa A, Dazzi D, Hassan H.The influence of selenium supplementation on postpartum thyroid status in pregnant women with thyroid peroxidase autoantibodies. J Clin Endocrinol Metab. 92:1263-8, 2007.

108.2: Mazokopakis EE, Papadakis JA, Papadomanolaki MG, Batistakis AG, Giannakopoulos TG, Protopapadakis EE, Ganotakis ES. Effects of 12 Months Treatment with l-Selenomethionine on Serum Anti-TPO Levels in Patients with Hashimoto's Thyroiditis.Thyroid. 2007 Aug;17(7):609-12

108.3 Karanikas G, Schuetz M, Kontur S, Duan H, Kommata S, Schoen R, Antoni A, Kletter K, Dudczak R, Willheim M. No immunological benefit of selenium in consecutive patients with autoimmune thyroiditis. Thyroid. 2008 Jan;18(1):7-12

108.4 Wichman J, Winther KH, Bonnema SJ, Hegedüs L. Selenium Supplementation Significantly Reduces Thyroid Autoantibody Levels in Patients with Chronic Autoimmune Thyroiditis: A Systematic Review and Meta-Analysis. Thyroid. 2016 Dec;26(12):1681-1692

108.5 Winther KH, Wichman JE, Bonnema SJ, Hegedüs L. Insufficient documentation for clinical efficacy of selenium supplementation in chronic autoimmune thyroiditis, based on a systematic review and meta-analysis. Endocrine. 2017 Feb;55(2):376-385

108.6 Nordio M, Pajalich R. Combined treatment with Myo-inositol and selenium ensures euthyroidism in subclinical hypothyroidism patients with autoimmune thyroiditis. J Thyroid Res. 2013;2013:424163. doi: 10.1155/2013/424163. Epub 2013 Oct

108.7 Nordio M, Basciani S. Treatment with Myo-Inositol and Selenium Ensures Euthyroidism in Patients with Autoimmune Thyroiditis. Int J Endocrinol. 2017;2017:2549491. doi: 10.1155/2017/2549491. Epub 2017 Feb 15.

108.8 Caturegli P, De Remigis A, Ferlito M, Landek-Salgado MA, Iwama S, Tzou SC, Ladenson PW. Anatabine ameliorates experimental autoimmune thyroiditis. Endocrinology. 2012 Sep;153(9):4580-7

108.9 Schmeltz LR, Blevins TC, Aronoff SL, Ozer K, Leffert JD, Goldberg MA, Horowitz BS, Bertenshaw RH, Troya P, Cohen AE, Lanier RK, Wright C 4th. Anatabine supplementation decreases thyroglobulin antibodies in patients with chronic lymphocytic autoimmune (Hashimoto's) thyroiditis: A randomized controlled clinical trial. J Clin Endocrinol Metab. 2014 Jan;99(1):E137-42

 

  1. Amino N, Tada H, Hidaka Y. The spectrum of postpartum thyroid dysfunction: diagnosis, management, and long-term prognosis. Endoc Prac 2:406, 1996.
  2. Karges B, Muche R, Knerr I, Ertelt W, Wiesel T, Hub R, Neu A, Klinghammer A, Aufschild J, Rapp A, Schirbel A, Boehm BO, Debatin KM, Heinze E, Karges W.Levothyroxine in euthyroid autoimmune thyroiditis and type 1 diabetes: a randomized, controlled trial.J Clin Endocrinol Metab. 2007 May;92(5):1647-52.

 

Disorders of the Thyroid Gland in Infancy, Childhood and Adolescence

This chapter is, in part, based on the previous version written by Prof. Rosalind Brown.

ABSTRACT

Thyroid disorders in infancy, childhood and adolescence represent common and usually treatable endocrine disorders. Thyroid hormones are essential for normal development and growth of many target tissues, including the brain and the skeleton. Thyroid hormone action on critical genes for neurodevelopment is limited to specific time window, and even a short period of deficiency of TH can cause irreversible brain damage. During the first trimester of pregnancy fetal brain development is totally dependent on maternal thyroid function. Congenital hypothyroidism is one of the most preventable causes of mental retardation, but early diagnosis is needed in order to prevent irreversible SNC damage. Today more than 70% of the babies worldwide are born in areas without an organized screening program. New insights about genetic causes, screening strategies and treatment of congenital hypothyroidism are reported. Hyperthyroidism in newborns is usually a transient consequence of transplacental passage of TSH receptor stimulating antibodies. Hypothyroidism can be detected in infants born to hyperthyroid mothers, due to transplacental passage of TSH receptor antibodies or hypothalamic-pituitary suppression. In childhood and adolescence autoimmune thyroid disease (AITD) as chronic lymphocytic thyroiditis and Graves’ disease account for the main cause of hypothyroidism and hyperthyroidism, respectively. Incidence of AITD increase from infancy to adolescence. Other autoimmune disorders are frequently associated. An increased risk of thyroid nodules and cancer is suggested. Differentiated thyroid cancer and medullary thyroid carcinoma in childhood and adolescence require specific expertise. Follow up programs are advised for high risk patients as long term survivors of childhood cancer. For complete coverage of this and related areas of Endocrinology, please visit our free online textbook, WWW.ENDOTEXT.ORG.

INTRODUCTION

Thyroid hormone is essential for the growth and maturation of many target tissues, including the brain and skeleton. As a result, abnormalities of thyroid gland function in infancy and childhood result not only in the metabolic consequences of thyroid dysfunction seen in adult patients, but in unique effects on the growth and /or maturation of these thyroid hormone-dependent tissues as well. In most instances, there are critical windows of time for thyroid hormone-dependent development and so the specific clinical consequence of thyroid dysfunction depends on the age of the infant or child. For example, newborn infants with congenital hypothyroidism frequently have hyperbilirubinemia, and delayed skeletal maturation, reflecting immaturity of liver and bone, respectively, and they are at risk of permanent mental retardation if thyroid hormone therapy is delayed or inadequate; their size at birth, however, is normal. In contrast, hypothyroidism that develops after the age of three years (when most thyroid hormone-dependent brain development is complete) is characterized predominantly by a deceleration in linear growth and skeletal maturation but there is no permanent effect on cognitive development. In general, infants with severe defects in thyroid gland development or inborn errors of thyroid hormonogenesis present in infancy whereas babies with less severe defects or acquired abnormalities, particularly autoimmune thyroid disease, present later in childhood and adolescence. In the newborn infant, thyroid function is influenced not only by the neonate ’ s own thyroid gland but by the transplacental passage from the mother of factors that affect the fetal thyroid gland.

In the last several decades, there have been exciting advances in our understanding of fetal and neonatal thyroid physiology, and screening for congenital hypothyroidism has enabled the virtual eradication of the devastating effects of mental retardation due to sporadic congenital hypothyroidism in most developed countries of the world. In addition, advances in molecular biology have led to new insights regarding the early events in thyroid gland embryogenesis and mechanisms of thyroid action in the brain. At the same time, the molecular basis for many of the inborn errors of thyroid hormonogenesis and thyroid hormone action is being unraveled. However, new questions and new challenges arise. In particular, the survival of increasingly small and premature fetuses has resulted in a growing number of neonates with abnormalities in thyroid function and a continuing controversy as to which of these infants require therapy. This chapter will focus on current concepts regarding the ontogenesis of thyroid function in the fetus and will review the major disorders of thyroid gland function in infants and children.

ONTOGENESIS OF THYROID FUNCTION IN THE FETUS AND INFANT

The ontogeny of mature thyroid function involves the organogenesis and maturation of the hypothalamus, pituitary, and thyroid glands as well as the maturation of thyroid hormone metabolism and thyroid hormone action. The placenta also plays a key role in the transfer of hormones and factors other than T4 that impact on thyroid function. In the first half of pregnancy, maternal T4 provides an important source of hormone for the developing fetus. Much of our knowledge derives from work in animal models, particularly sheep and rat. In interpreting these data, it is important to remember potential limitations in these models because of differences both in the structure of the placenta and timing of maturation. For example, the rat thyroid gland is much less mature at birth than its human counterpart and significant maturation of the thyroid gland and of the hypothalamic-pituitary-thyroid axis in this species occurs in the first 2 or 3 weeks after birth in the absence of placental or maternal influence, as compared with the third trimester in human infants.

Thyroid Gland Embryogenesis

Thyroid gland development is extensively reviewed in an earlier chapter and is shown diagrammatically in Figure 1. In brief, the thyroid gland is derived from the fusion of a medial outpouching from the floor of the primitive pharynx, the precursor of the thyroxine (T4)-producing follicular cells, and bilateral evaginations of the fourth pharyngeal pouch, which gives rise to the parafollicular, or calcitonin (C) secreting cells. Commitment towards a thyroid-specific phenotype as well as the growth and descent of the thyroid anlage into the neck results from the coordinate action of a number of transcription factors, including thyroid transcription factor 1 (TTF1, now called NKX2 (1), TTF2 (now called FOXE1) and PAX8 (1,2). Because these transcription factors are also expressed in a limited number of other cell types, it appears to be the specific combination of transcription factors and possibly non-DNA binding cofactors acting coordinately that determine the phenotype of the cell.

Other transcription factors and growth factors that play a role in early thyroid gland organogenesis include HHEX1, HOXA3 (3) and members of the fibroblast growth factor family, i.e., FGF10, but the initial inductive signal is unknown. A role of the neighboring heart primordium in the specification of the thyroid anlage has been postulated. Studies of cadherin expression suggest that the caudal translocation of the thyroid anlage may also arise indirectly, as a result of the growth and expansion of adjacent tissues, including the major blood vessels (4). In late organogenesis, the sonic hedgehog (SHH) gene and its downstream target TBX1 appear to play an important role in the symmetric bilobation of the thyroid (5); SHH also suppresses the ectopic expression of thyroid follicular cells (6).

During caudal migration the pharyngeal region of the thyroid anlage contracts to form a narrow stalk, known as the thyroglossal duct, which subsequently atrophies. Usually no lumen is left in the tract of its descent but, occasionally, an ectopic thyroid and/or persistent thyroglossal duct or cyst form if thyroid descent is abnormal.

Figure 15- 1. Approximate timing of thyroid gland maturation in the human fetus.

In the human, embryogenesis is largely complete by 10 to 12 weeks gestation. At this stage, tiny follicle precursors can be seen, iodine binding can be identified and thyroglobulin (Tg) detected in follicular spaces (7,8) . Thyroid hormones are detectable in fetal serum by gestational age 11 to 12 weeks with both thyroxine (T4) and triiodothyronine (T3) being measurable. However, as discussed in further detail below, it is likely that a fraction of the hormones detectable at this early stage is contributed by the mother through transplacental transfer. Thyroid hormones continue to increase gradually over the entire period of gestation as does serum thyroxine-binding globulin (TBG) (9,10) . TBG is present at levels of 100 nmol/L (5 mg/L) at gestational age 12 weeks and progressively increases up to the time of birth, reaching concentrations of 500 nmol/L (25 mg/L). The serum TBG concentrations are higher in the infant then in adult humans as a consequence of placental estrogen effects on the fetal liver. In addition to the increase in total T4 there is also a progressive increase of the free T4 concentration indicating a maturation of the hypothalamic- pituitary- thyroid axis. The increased total T4 / thyrotropin (TSH) and free T4 /TSH ratios also indicate an increased ability of the thyroid gland to respond to TSH due to upregulation of the TSH receptor (11). Whereas the TBG and total T4 levels rise throughout gestation, the concentrations of free T4, and TSH rise until 31 to 34 weeks, declining thereafter to term (12).

Tg can be identified in the fetal thyroid as early as the 5th week, and is certainly present in follicular spaces by 10 to11 weeks, but maturation of Tg secretion takes much longer and it is not known when circulating Tg first appears in the fetal serum (not shown). By the time of gestational age 27 to 28 weeks, however, Tg levels average approximately 100 mg/L, much higher than in the adult and they remain approximately stable until the time of birth (13,14) . Iodide concentrating capacity can be detected in the thyroid of the 10 to 11 week fetus, but maturation of the Wolff-Chaikoff effect (reduction of iodide trapping in response to excess iodide) does not appear until 36 to 40 weeks gestation. Thus the premature fetus is more sensitive than the full term neonate to the thyroid-suppressive effects of iodine exposure.

The Hypothalamic-pituitary Axis

TSH is detectable at levels of 3 to 4 mU/L at gestational age 12 weeks and increases moderately over the last two trimesters to levels of 6 to 8 mU/L (8,9).The maturation of the negative feedback control of thyroid hormone synthesis is observed by approximately mid-gestation (Figure 1), with elevated serum TSH concentrations being observed in hypothyroid infants as early as 28 weeks (8). When TSH-Releasing Hormone (TRH) is given to mothers, a rise in TSH in the fetal circulation has been noted as early as 25 weeks gestation (15). It is of interest that the fetal TSH increment after TRH is greater than is the paired-maternal response, a consequence either of enhanced TSH release or impaired TSH degradation, perhaps due to immaturity of the hepatic glycoprotein metabolic clearance system. Similarly TSH is reduced in the cord serum of infants with neonatal thyrotoxicosis due to the transplacental passage of thyroid-stimulating antibodies from mothers with Graves’ disease as early as the end of the 2nd trimester.

Serum levels of TRH are higher in the fetal circulation than in maternal blood, the result both of extrahypothalamic TRH production (placenta and pancreas) and the decreased TRH degrading-activity in fetal serum. The physiological significance of these increased levels of TRH in the fetal circulation is not known.

Maturation Of Peripheral Thyroid Hormone Metabolism

As discussed in an earlier chapter, there are three iodothyronine deiodinases involved in the activation and inactivation of thyroid hormone. All three are coordinately regulated during gestation and function to closely regulate the supply of T3 to developing tissues while at the same time protecting the fetus against the effects of excess thyroid hormone. The physiological rationale for the maintenance of reduced circulating T3 concentrations throughout fetal life is still unknown, but it has been suggested that its function may be to avoid tissue thermogenesis and potentiate the anabolic state of the rapidly growing fetus while at the same time permitting highly regulated, tissue- specific maturation in an orderly, temporal sequence.

The seleno-enzyme type 1 iodothyronine deiodinase (D1), an important activating enzyme in adult life, is low throughout gestation. In addition to catalyzing T4 to T3 conversion, D1 catalyzes the inactivation of the sulfated conjugates of T4. As a consequence, circulating T3 concentrations in the fetus are quite low whereas the serum levels of the biologically inactive isomer reverse T3 and of T3 sulfate are increased (10). Unlike D1, both the Type 2 deiodinase (D2), an activating enzyme and D3, an inactivating enzyme are present in fetal brain as early as 7 weeks ’ gestation (16) . D2 converts T4 to T3 while D3 converts T4 to reverse T3. D2 and D3 are the major isoforms present in the fetus and are especially important in defining the level of T3 in the brain and pituitary. The highest concentration of D2 is in brain, pituitary, placenta and brown adipose tissue. D3 is present in many fetal tissues, most prominently the brain, uteroplacental unit, skin, and gastrointestinal tract (17). This is consistent with the key role of D3 in protecting fetal tissues against high maternal T4 concentrations present either in the placenta or in amniotic fluid.

In the presence of hypothyroidism, D2 activity increases while D3 decreases These coordinate activities have been found to be critically important in defending the rat fetus against the effects of fetal hypothyroidism as long as maternal T4 levels are maintained at normal concentrations (18, 19). Despite the low levels of circulating T3, brain T3 levels are 60-80% those of the adult by fetal age 20-26 weeks (20). Thus, whereas the physiological interrelationships between the various deiodinases in the fetus and placenta seem designed to maintain circulating T3 concentrations at a reduced level, specific mechanisms have evolved for maintaining brain T3 concentrations so that normal development can proceed.

Role of the Placenta

Contributions of the maternal thyroid to fetal thyroid economy.

In the human infant under normal circumstances, the placenta has only limited permeability to thyroid hormone and the fetal hypothalamic-pituitary-thyroid system develops relatively independent of maternal influence. Placental thyroid hormone transporters, thyroid hormone binding proteins, iodothyronine deiodinases, sulfotransferases and sulfatases regulate the transport of maternal thyroid hormones to the fetus (20a,20b). The transport of iodine through the placenta is also important as the organ has shown to actively concentrate the anion (20c).

The human placenta expresses iodothyronine Type 2 deiodinase I (D2) (which activates T4 to T3) and Type 3 (D3) (which inactivates T4 and T3). Maternal T4 is metabolized by D3 having 200 times the activity of D2 (20b). Both D2 and D3 activity decrease with advancing gestation (20b). Thus, the relative impermeability of the human placenta to thyroid hormone is due to the presence of D3 which serves to inactivate most of the thyroid hormone presented from the maternal or fetal circulation. The iodide released in this way can then be used for fetal thyroid hormone synthesis. Iodine is actively transported from the maternal circulation to the fetus through the placenta that express placental sodium iodide transporter (NIS) (20c,20d). NIS actively concentrates Iodine. NIS protein levels are significantly correlated with gestational age during early pregnancy and increase with increased placental vascularization (20e).

Interest in the potential role of maternal T4 in the fetal thyroid economy was reawakened with the recognition that in infants with the congenital absence of thyroid peroxidase, the cord serum concentration of T4 is nonetheless between 25 and 50% of normal (21). Since these infants are completely unable to synthesize T4, the measured hormone must be maternal in origin. Similar results are obtained in retrospective studies of cord serum in infants with sporadic congenital athyreosis. This maternal T4 disappears rapidly from the newborn circulation with a half-life of approximately 3 to 4 days.

There is also evidence that maternal-fetal T4 transfer occurs in the first half of pregnancy, when fetal thyroid hormone levels are low (19,22). Low concentrations of T4, presumably of maternal origin, have been detected in human embryonic coelomic fluid as early as 6 weeks gestation and in fetal brain as early 10 weeks gestation prior to the onset of fetal thyroid function indicating its maternal origin (22a-22f). Furthermore, both D2 and D3 activity as well as thyroid hormone receptor (TR) isoforms are present in low concentrations in human fetal brain from the mid first trimester, indicating that the machinery to convert T4 to T3 and to respond to T3 is present. The mechanisms of actions of thyroid hormones in the developing brain are mainly mediated through two ligand activated thyroid hormone receptor isoforms (22b,22c). There is also an important role for the thyroid hormone transporters in one or more of these processes (22g).

Between 6-12 weeks gestation, if maternal total T4 concentration is 100%, the total T4 concentration in the coelomic fluid would be 0.07% and T4 in the amniotic cavity as little as 0.0003-0.0013% of maternal total T4 concentrations. Fetal circulating concentrations of T3 are at least 10 fold lower than T4, whereas by fetal age of 20-26 weeks T3 levels in the fetal brain are 68-80% of the adult brain (20). Unlike adults, the proportion of free unbound T4 is also higher than bound T4 in early gestation. Free T4 levels are determined by the fetal concentrations of the thyroid hormone binding proteins in the circulation and the amount of maternal T4 crossing the placenta (7-9). It seems likely that when fetal thyroid function is normal, the net flux of T4 from mother to fetus is relatively limited. However, when the fetus is hypothyroxinemic, there is significant bulk transfer of T4 to the fetal circulation. This can occur both at the level of the placental maternal capillary interface and via uptake of thyroid hormone from the amniotic fluid through the immature epidermis. T4 uptake by the fetus can also occur via fetal ingestion of amniotic fluid. While the T4 concentrations in amniotic fluid appear modest, the fraction of T4 free in amniotic fluid is approximately ten-fold higher than that of serum and thus the free T4 concentration in amniotic fluid is approximately equal to that in fetal serum at 20 weeks gestation. It has been shown on numerous occasions in both animals and humans that amniotic fluid iodothyronine concentrations reflect those in the maternal circulation (23).

Placenta is permeable to TRH (15) and to immunoglobulins G (IgG) from midgestation. At the time of delivery, cord blood TPOAb correlate with maternal TPOAb concentrations (23a). Maternal passage of TPOAb and TgAb are not associated with thyroid fetal dysfunction. On the contrary, maternal TSH receptor antibodies (both stimulating and blocking) can be dangerous for the fetus and the newborn.

Fetal and neonatal hyperthyroidism can be caused by transplacental passage of TSH receptor antibodies (TRAb), whereas hypothyroidism can be due to transpancental passage of blocking TSH receptor antibodies, from mothers with severe Graves’ disease or severe hypothyroidism due to chronic lymphocytic thyroiditis.( The placenta is also permeable to certain drugs (15). Thus, the administration to the mother of excess iodide, drugs (especially propylthiouracil or methimazole), can affect thyroid function in the fetus and the newborn.

Role of Maternal Thyroid Hormone for fetal brain development and neurocognitive development in the offspring

The essential role that thyroid hormones (TH) play on the fetal brain development starts long before the onset of fetal thyroid function (22-22a). Thus, during the first trimester of pregnancy, fetal brain development is totally dependent on maternal thyroid function. Because the action of TH on critical genes for fundamental neurobiological processes is limited to specific time window, even a short period deficiency of TH may cause permanent brain damage. TH deficiency may affect neuronal cell differentiation and migration, axonal and dendritic outgrowth, myelin formation and synaptogenesis (22b-22f). It is well known that severe Iodine deficiency during pregnancy causes inadequate thyroid hormone production and irreversible brain damage known as cretinism, still endemic in many areas of the world (23b). None of the neurological features of severe endemic cretinism (24) due to iodine deficiency are found in infants with sporadic congenital hypothyroidism whose mothers have normal thyroid function and who receive early and adequate postnatal treatment. Similarly, impaired hearing, when found is much milder and less frequent (25). This would appear to provide unequivocal evidence that the neurological damage sustained by infants with endemic cretinism can be largely prevented by maternal T4. In addition to endemic cretinism, significant developmental delay despite early and adequate postnatal therapy has also been reported in other models of combined maternal-fetal hypothyroidism, such as materno-fetal POU1F1 deficiency (26) and TSH receptor blocking antibody-induced congenital hypothyroidism (27).

In iodine sufficient areas the main cause of maternal thyroid dysfunction (hypothyroidism, subclinical hypothyroidism or hypothyroxinemia) is thyroid autoimmunity, detectable in up to 17% of women (27a). Several studies reported on the consequences of maternal thyroid dysfunction in the progeny. Studies in children born to women with non-iodine deficient hypothyroidism during pregnancy (28,29,29a,29b) as well as in children from hypothyroxinemic mothers (30,30a-30e) have been published. Different parameters and different periods of pregnancy (i.e., increased TSH, low T4, presence or absence of autoimmunitity, prevalent obstetrical or developmental outcome) were analyzed, reporting conflicting results and conclusions.

Impairment in psychomotor development in the offspring of pregnant women with thyroid dysfunction was first reported by Man et al (28). They examined 131 hypothyroxinemic untreated pregnant women and found 36% of their 7-year-old children scored in the dull normal range or below compared to 16% of children of euthyroid mothers (28). Haddow detected a seven point IQ deficit in 7 to 9 year old children whose mothers were retrospectively found to have been hypothyroid at 17 weeks gestation (29). Accordingly, Pop demonstrated that even babies born to women whose free T4 levels were in the lowest 10% of normal at 12 weeks gestation had a measurable impairment in psychomotor development at 2 years of age as compared with the rest of the population, but this effect was not observed if maternal thyroid function was normal at 32 weeks (30). At variance with the aforementioned studies, Liu and more recently, Momotami failed to demonstrate any IQ deficit in babies born to hypothyroid mothers as long as the hypothyroidism was corrected by the end of the second trimester (31a, 31b). Similar results were obtained by Downing et al in 3 children born after severe feto-maternal hypothyroidism due to TSH receptor blocking antibodies (31c). Attention deficit disorder (30f,30g) autistic symptoms in offspring (30h) and schizophrenia in later life (30K) have also been associated with maternal hypothyroxinemia. Attention deficit disorder was previously noted in offspring from mothers with thyroid autoimmunity (30i). Children from mothers with anti-thyroid peroxidase antibodies have been found to have intellectual impairment in early infancy (30j) and a reduced childhood cognitive performance at age 4 and 7 and sensineural hearing loss at both ages (30l). An interesting association study, derived from the Rotterdam cohort, (the population based prospective study from Rotterdam (Generation R) for the first time analyzed the effects of maternal thyroid function on brain morphology of the offspring. In this study 3839 mother-child pairs were included. Maternal serum samples were taken before 18 weeks of gestation (9-18w). MRI were performed in 646 children (mean age 8 years) and IQ determined at mean age 6 years. They found that both high maternal and low FT4 showed an inverted U shaped association with child IQ (-1.4-3.8 points), child grey matter volume and cortex volume (32c). Recently, in the prospective double blind randomized controlled antenatal thyroid screening study (CATS), levothyroxine treatment was started from the 13th week of gestation if serum TSH was >97.5th percentile and/or FT4 was <2.5th percentile. The outcome was the IQ in the offspring at 3 years. No significant differences in IQ values were found between 390 children of treated mothers compared to 404 children of untreated mothers (32).

The incidence of maternal hypothyroidism during pregnancy (3 per 1000 in iodine-sufficient populations (33) is almost ten times that of congenital hypothyroidism for which routine population screening is widespread. Because maternal hypothyroidism has been associated not only with potential adverse effects on fetal brain development but an increased risk of preterm delivery and of miscarriage as well (33a ), some have argued that all pregnant women should be screened for hypothyroidism, a position that has been endorsed by some but not other professional societies.

Updated guidelines for the management of thyroid disease during pregnancy have been recently released from ATA (33b).

THYROID FUNCTION IN THE NEONATE, THE INFANT, AND DURING CHILDHOOD

The Full-term Neonate

Marked changes occur in thyroid physiology at the time of birth in the full term newborn. One of the most dramatic changes is an abrupt rise in the serum TSH which occurs within 30 minutes of delivery, reaching concentrations as high as 60 to 70 mU/L (8). This causes a marked stimulation of the thyroid and an increase in the concentrations of both serum T4 and T3 (34). These consist of an approximate 50% increase in the serum T4 and an increase of three- to four-fold in the concentration of serum T3 to adult levels at 1 to 4 days of life. Serum levels of T4, free T4 and TBG remain elevated over cord levels at 7 days of postnatal life (Figure 2), decreasing thereafter. The T3 concentration rises strikingly at Day 7, and continues to rise for the first 28 days. Opposite effects are noted in the reverse T3 levels and T3 sulfate.

Studies in experimental animals suggest that the increase in TSH is a consequence of the relative hypothermia of the ambient extrauterine environment. However, while a significant portion of the marked increase in T3 from its low basal levels in cord serum can be explained by the abrupt increase in TSH, the simultaneous fall in reverse T3 and T3 sulfate are consistent with an increase in D1 activity occurring at the same time. D2 has been identified in human brown adipose tissue as well as brain and the acute increase in T3 in adipose tissue at birth is required for optimal uncoupling protein synthesis and thermogenesis (35,36).

Premature Infants

Thyroid function in the premature infant reflects, in part, the relative immaturity of the hypothalamic-pituitary-thyroid axis that is found in comparable gestational age infants in utero. Following delivery, there is a surge in T4 and TSH analogous to that observed in term infants, but the magnitude of the increase is less in premature neonates (8). In infants <31 weeks, the circulating T4 concentration may not increase and may even fall in the first 1 to 2 weeks of life (37) (Figure 2). This decrease in the T4 concentration is particularly significant in very premature infants, in whom the serum T4 may occasionally be undetectable. In most cases, the total T4 is more affected than the free T4 (38), a consequence of abnormal protein binding and/or the decreased TBG in these babies with immature liver function.

Figure 15-2. Postnatal changes in of T4, free T4, TBG, T3, rT3 and TSH according to gestational age. Values determined in babies born at gestational age of 23-27, 28-30, 31-34 and 37 weeks or more are reported. Note the increase in T4, free T4 and TBG in the full term infant in the first week of life. T3 also rises strikingly, while rT3 and TSH decline. The increase in T4 and free T4 is blunted in infants

The causes of the decrease in T4 observed postnatally in premature infants are complex. In addition to the clearance of maternal T4 from the neonatal circulation, preterm babies have decreased thyroidal iodide stores (39) (a problem of particular significance in borderline iodine-deficient areas of the world), they are frequently sicker than their more mature counterparts, are less able to regulate iodide balance, and they may be treated by drugs that affect neonatal thyroid function (particularly dopamine and steroids). In addition, since the capacity of the immature thyroid to adapt to exogenous iodide is reduced, there is an increase in sensitivity to the thyroid-suppressive effects of excess iodide found in certain skin antiseptics and drugs to which these babies are frequently exposed (see below).

Despite the reduced total T4 observed in some preterm babies, the TSH concentration is not significantly elevated in most of these infants. In some babies, transient elevations in TSH are seen, the finding of a TSH concentration >40 mU/L being more frequent the greater the degree of prematurity. Frank et al found, for example, that the prevalence of a TSH concentration >40 mU/L in very low birth weight, (<1.5 kg), i.e., very premature, infants was 8-fold higher and in low birth weight, (1.5 kg-2.5 kg) neonates 2-fold higher than the prevalence in term babies (40). Whereas in some cases, an elevated TSH concentration may reflect true primary hypothyroidism, in other instances this increase in TSH may reflect the elevated TSH observed in adults who are recovering from severe illness. Such individuals may develop transient TSH elevations that are associated with still reduced serum T4 and T3 concentrations. These have been interpreted as reflecting a “ re-awakening ” of the illness-induced suppression of the hypothalamic pituitary axis. As the infant recovers from prematurity associated illnesses such as respiratory distress syndrome (RDS), a recovery of the illness-induced suppression of the hypothalamic- pituitary- thyroid axis would also occur.

Figure 15-3. Cord blood levels of T4, free T4, TBG, T3, reverse T3 and TSH in the human infant. Note the low T3 and high reverse T3 concentrations as well as the discrepancy between the total T4 and free T4 levels in very premature babies. (Redrawn from Williams et al (10). See text for details).

Somewhat surprisingly, given the relative immaturity of the thyroid gland, serum Tg concentrations are higher in the premature than in the full term infant (41), particularly in those who are sick with respiratory distress syndrome. In view of the attenuated postnatal TSH rise in the latter babies, it is likely that impaired clearance and/or degradation of this glycoprotein from the circulation rather than increased secretion plays an important role.

Small-for-gestational-age (SGA) Infants

SGA infants have significantly higher TSH and lower total and free T4 values than do infants of normal weight (42). This can be related to the severity of the malnutrition in these infants, as well as to fetal hypoxemia and acidemia. Impaired placental perfusion and chronic starvation may also play a role. This pattern of reduced T4 and elevated TSH differs from the response to starvation in older individuals and healthy adults in whom TSH is reduced. The explanation for the relatively higher TSH in duch infants is not known.

Infants and Children

Following the acute perturbations of the neonatal period there is a slow and progressive decrease in the concentrations of T4, free T4, T3 and TSH during infancy and childhood (43). Younger children tend to have slightly higher serum concentrations of T3 and TSH, so age-specific normative values should always be consulted. The serum concentration of reverse T3 remains unchanged or increases slightly. Serum Tg levels also fall over the first year of life reaching concentrations typical of adults by about 6 months of age. Another important aspect of thyroid physiology in the infant and child is the markedly higher T4 turnover in this age group relative to that in the adult. In infants, T4 production rates are estimated to be on the order of 5 to 6 mcg/kg per day decreasing slowly over the first few years of life to about 2 to 3 mcg/kg/day at ages 3 to 9 years. This is to be contrasted with the production rate of T4 in the adult which is about 1.5 mcg/kg/day. The size of the infant thyroid gland increases quite slowly. The thyroid gland of the newborn weighs approximately 1 gram and increases about 1 gram per year until age 15 when it has achieved its adult size of about 15 to 20 g. In general, the size of the thyroid lobe is comparable to that of the terminal phalanx of the infant or child’s thumb.

THYROID DISEASE IN INFANCY

Congenital hypothyroidism

Non endemic congenital hypothyroidism is one of the commonest treatable causes of mental retardation. The association between goitrous hypothyroidism and mental retardation was first noted more than 400 years ago by Paracelsus in 1527, and Thomas Curling first described sporadic nongoitrous hypothyroidism in 1850. However, despite the demonstration by Murray in 1891 that thyroid extract could ameliorate many of the features of untreated cretinism, it was not until the 1970’ that the importance of early treatment in diminishing the neuro-psychological abnormalities of congenital hypothyroidism was demonstrated convincingly (45). The development by Dussault et al of a sensitive and specific radioimmunoassay for the measurement of T4 in dried whole blood eluated from filter paper (and later tests for T4 and TSH using 1/8 ″ discs) provided the technical means to screen all newborns for congenital hypothyroidism prior to the development of clinical manifestations (46). Thus, as summarized by Delange, congenital hypothyroidism includes all the characteristics of a disease for which screening is justified: 1) it is common (4-5 times more common than phenylketonuria for which screening programs were initially developed); 2) to prevent mental retardation, the diagnosis must be made early, preferably within the first few days of life; 3) at that age, clinical recognition is difficult if not impossible; 4) sensitive, specific screening tests and 5) simple, cheap effective treatment are available; and 6) the benefit-cost ratio is highly favorable (approximately 10/1, a ratio that does not include the loss of tax income that would result from impaired intellectual capacity in the untreated, but non-institutionalized, person) (47). Since the development of the first pilot screening program for the detection of congenital hypothyroidism in Quebec in 1972, newborn screening programs have been introduced throughout the industrialized nations and are under development in many other parts of the world. It has been estimated that as of 1999, some 150 million infants had been screened for congenital hypothyroidism worldwide with 42,000 cases detected (46). Although there continues to be some disagreement as to whether minor neuro-intellectual sequelae remain in the most severely affected infants, accumulating evidence suggests that a normal outcome is possible even in the latter group of babies as long as treatment is started sufficiently early and is adequate (48-50). Certainly, the main objective of screening, the eradication of mental retardation, has been achieved.

National screening programs are well organized in many developed countries. However, it must be emphasized that approximately 71% of babies worldwide are not born in an area with an established national screening program for CH. The economic burden of disability owing to congenital hypothyroidism is still a significant public health challenge (50a).

The prevalence of CH was approximately 1:7000 to 1:10000 in the prescreening era and decreased to1;3000 to 1.4000 in the 1970s and 1980s when the screening programs were applied. Rates ranging from 1:1400 to 1:2800 have been recently reported by screening programs in USA, Canada, Italy, Greece, and New Zealand (50b).

Lower TSH cut off values used in the screening programs and changes in birth population partially explained the higher incidence reported. Lower cutoff values for TSH have been adopted in many countries over the years, leading to the identification of milder forms of CH essentially with eutopic thyroid gland (thyroid in situ). Ford and LaFranchi in 2014 (50a) found that lowering the TSH cutoff value from greater than 20-25 uU/mL to greater than 6-10 approximately doubled the incidence of CH. A study from Italy reported that 21.6% of babies with permanent CH had TSH value at screening less than 15 uU/mL (applied between 2000 and 2006, cutoff TSH value ranged from 15 to 7uU/mL in different regions). The frequency of thyroid dysgenesis in this group was 19.6% and TSH levels at confirmation ranged from 9.9 to 708 uU/mL .It is important to remember that in this study TSH value at screening does not discriminate between transient and permanent forms of CH (50c).

Harris and Pass reported that CH incidence increased from 1:3373 in 1978 to 1:1415 in 2005 (50d). Changes in the demographics of the birth population in New York partially explained the increased incidence of CH. They found a 23% increase with a birth weight < 1500 gr., 50% increase of twin/multiple births, 41% increase in mothers >30 years of age (50d). Also changes in percentage of races or ethnicity of newborns play a role, as shown in the State of California. In this study, the incidence of CH in Asian Indian is reported to be 1:1200 and in Hispanic 1:1600, versus 1:11000 in Non Hispanic Black (50e). A further study from the Italian Study Group, based on data from the Italian National Registry from 1987 to 2008 showed an increased incidence of both permanent and transient CH, in more recent years (50f). The authors investigated trends in the incidence of CH between the period 1987-1998, and 1999-2008. They found an increasing of 38% (from 1:3200 to 1:2320) of the incidence of permanent CH and of 54% (from1:3000 to 1:1940) including the transient forms in the period 1999-2008. The most important factor was the lowering of cutoff TSH values (from greater than 20 to 7/15 uU/ml since 1999. Moreover an increment of 58% of preterm babies with permanent CH was also reported in the second period. Permanent CH due to thyroid dysgenesis had a slight increase, being the great majority of cases presented with normal/hyperplastic thyroid.

A national study from France, including 6622 cases of CH identified from 1982 to 2012 showed that the incidence rate CH due to eutopic glands increased by 4.4 fold in this period, regardless of the screening method adopted. Interestingly, also severe eutopic forms of CH increased by 2.1%. The incidence of dysgenesis did not change (50g).

Screening Strategies

Screening for primary CH worldwide should be performed on the basis of national resources. The aim of neonatal screening is the earliest identification of any form of congenital hypothyroidism, but particularly those patients with severe hypothyroidism in whom disability is greatest if not treated. The identification of Central Congenital Hypothyroidism (CCH) by screening programs is under debate. Two screening strategies for the detection of congenital hypothyroidism have evolved. In the primary T4/backup TSH method, still favored in much of North America and the Netherlands, T4 is measured initially while TSH is checked on the same blood spot in those specimens in which the T4 concentration is low. In the primary TSH approach, favored in most parts of Europe and Japan, blood TSH is measured initially.

A primary T4/backup TSH program will detect overt primary hypothyroidism, secondary or tertiary hypothyroidism, babies with a low serum T4 level but delayed rise in the TSH concentration, TBG deficiency and hypothyroxinemia; this approach may, however, miss subclinical hypothyroidism. A primary TSH strategy, on the other hand, will detect both overt and subclinical hypothyroidism, but will miss secondary or tertiary hypothyroidism, a delayed TSH rise, TBG deficiency and hypothyroxinemia. There are fewer false positives with a primary TSH strategy. Both programs will miss the rare infant whose T4 level on initial screening is normal but who later develops low T4 and elevated TSH concentrations. This pattern has been termed “atypical” congenital hypothyroidism or “delayed TSH” and is observed most commonly in premature babies with transient hypothyroidism or infants with less severe forms of permanent disease.

In a few regions, a second routine specimen is collected from all births at 2-4 weeks of age (51). Results from the Northwest Regional Screening program, coordinated in Oregon, (USA), that applied this method, have recently been published (51a). In 2014 the European Society for Pediatric Endocrinology, (ESPE) on behalf of all the scientific societies of pediatric endocrinologists worldwide (ESPE,PES, SLEP, JSPE, APEG, APPES, ISPAE) published updated guidelines about screening, diagnosis, and management of congenital hypothyroidism (51b, 51c).

According to the ESPE guidelines, the most sensitive test for detecting primary CH is the determination of TSH concentration that detects primary CH more effectively than primary T4 screening (51b,51c). Primary T4 screening with confirmatory TSH testing can detect some cases of CCH, but some cases of mild CH can be missed, depending on the cutoff T4 value used.

When available, screening strategies for the identification of CCH are: a) a combination of primary T4 and primary TSH screening, b) a combination of primary T4 screening with secondary TSH testing followed by T4 binding protein determination (TBG). The last one is employed by the Netherlands where, in addition to a primary T4/backup TSH approach, TBG is assessed in those filter paper specimens with the lowest 5% of T4 values (52). The T4/TBG ratio is used as an indirect reflection of the free T4, which is difficult to be measured directly in dried blood spots. This approach has been reported to result in improved sensitivity and specificity in detecting milder cases of primary congenital hypothyroidism that might otherwise be missed. An additional reported advantage was the identification of >90% of infants with central hypothyroidism compared with only 22% with primary T4 screening and none with a primary TSH approach. Since on subsequent testing > 80% of the babies with central hypothyroidism had multiple pituitary hormone deficiencies, a disorder associated with high morbidity and mortality for which effective treatment exists (53,53a), and in view of an apparent frequency (1 in 16,000) similar to that of phenylketonuria (1 in 18,000), the authors have argued that the goals of newborn thyroid screening should be extended to include the detection of babies with central hypothyroidism.

Recently a primary FT4 and TSH strategy was applied in Kanagawa Prefecture in Japan. A different method to determine FT4, based on enzyme-immunometric assays in filter paper blood eluates was used. They found a CCH prevalence of 1:31000 infants (53b,53c).

Measurement of T4 and/or TSH is performed on an eluate of dried whole blood (DBS) collected on filter paper by skin puncture on day 1-4 of life. Primary CH screening has been shown to be effective for the testing of cord blood or the blood collected on filter paper after the age of 24 hours. Blood is applied directly to the filter paper and after drying the card is sent to the laboratory. The best time to collect blood for TSH screening is 48 to 72 hours of age. The practice of early discharge from the hospital of otherwise healthy full term infants has resulted in a greater proportion of babies being tested before this time. For example, it has been estimated that in North America 25% or more of newborns are now discharged within 24 hours of delivery and 40% in the second 24 hours of life (54). Because of the neonatal TSH surge and the dynamic changes in serum T4 and T3 concentrations that occur within the first few days of life, early discharge increases the number of false positive results. It is important that in the screening laboratory the results of TSH are interpreted in relation to time of sampling. Ethnicity seems to play a role in determining mean TSH values at birth (54a).

Physicians caring for infants need to appreciate that there is always the possibility for human error in failing to identify affected infants, whichever screening program is utilized. This can occur due to poor communication, lack of receipt of requested specimens, or the failure to test an infant who is transferred between hospitals during the neonatal period (55). Therefore if the diagnosis of hypothyroidism is suspected clinically, the infant should always be tested (Figure 5).

Similarly, as is obvious from the discussion earlier in the chapter, adult normative values, provided by many general hospital laboratories, differ from those in the newborn period and should never be employed. Normal values according to both gestational and postnatal age for cord blood T4, free T4, TBG, T3, reverse T3, and TSH up to 28 days of life (10) are shown in Figure 2. Normal serum levels of Tg in premature and full-term infants (13,14) and normal serum levels of free T4 and TSH in the first week of life (56) have also been published, though it should be noted that precise values may vary somewhat, depending on the specific assays used.

Figure 15-4. Three month old male infant who was diagnosed clinically when he presented with a history of poor feeding at 3 months of age. The child was born in Puerto Rico prior to the development of newborn screening. Note the dull face, periorbital edema and enlarged tongue.

Screening in special categories of neonates at risk of CH

Special categories of neonates with CH can be missed at screening performed at usual time, particularly preterm babies and neonates with serious illnesses and multiple births. Drugs used in neonatal intensive care (i.e., dopamine, glucocorticoids that suppresses TSH), immaturity of hypothalamic-pituitary thyroid axis, decreased hepatic production of thyroid binding globulin, reduced transfer of maternal T4, reduced intake of iodine or excess iodine exposure, fetal blood mixing in multiple births can affect the first sample, and in many center a second specimen is required to rule out CH. (See section thyroid function in infants for more details).

Preterm babies have a higher incidence of a unique form of hypothyroidism, characterized by a delayed elevation of TSH. These babies can later develop low T4 and elevated TSH concentrations. This pattern has been termed “atypical” congenital hypothyroidism or “delayed TSH”. Preterm babies with a birth weight of less than 1500 gr. have an incidence of congenital hypothyroidism of 1:300. Survival of even extremely premature babies (<28 weeks of gestation) is around 90% in developed countries, and the incidence of prematurity is around 11.5 % in US and 11.8 % worldwide. So, an increasing subpopulation of preterm babies and high risk newborns deserves a special sight about screening and follow up of CH.

In these categories a second specimen 2-6 weeks from the first (ESPE guidelines suggested at about 15 days, or after 15 days from the first) may be indicated: preterm neonates with a gestational age of less than 37 weeks, Low Birth Weight and Very Low Birth Weight neonates and ill and preterm neonates admitted to neonatal intensive care unit, specimen collection within the first 24 hours of life, and multiple births, particularly in the case of same sex twins. The interpretation of the screening results should consider the results of a multiple sampling strategy, the age of sampling and the maturity (GA/birth weight) of the neonate.

Two recent papers (56a,56b) showed that a second screen (using a lower TSH cutoff) is able to detect the delayed elevation of TSH that occurs in these babies. Vigone et al (56a) revaluated the children with a diagnosis of CH detected at second screen and treated with L-thyroxine after 2 years of age and found 24% of cases with permanent congenital hypothyroidism, 52% with transient hypothyroidism and 24% with persistent hypertropinemia. Neither screening nor confirmatory TSH levels were able to predict the thyroid function after 2 years of age in these children.

Timing of normalization of thyroid hormones is critical for brain development (56c) and treatment should be started immediately if DBS TSH concentration is 40 mUI/l or more, after baseline TSH and FT4 serum determination, because this value strongly suggests decompensated hypothyroidism (56d). If TSH is < 40 mUI/l the clinician may postpone treatment, pending the serum results, for 1-2 days. ESPE guidelines (51b,51c) suggest treatment should be started if venous TSH concentration is persistently >20 mUI/l, even if serum FT4 is normal. Overtreatment can be dangerous for neurocognitive outcome and should be avoided, individualizing the dosage.

It is still a matter of debate if treatment can be beneficial in otherwise healthy babies with venous TSH concentration between 6-20 mUI/l and FT4 concentration within the normal limits for age. In these cases, diagnostic imaging is recommended to try to establish a definitive diagnosis. If TSH concentration remains high for more than 3 or 4 weeks, it is possible (in discussion with the family) either starting LT4 supplementation immediately and retesting, off treatment, at a later stage, or retesting two weeks later without treatment. Waiting for larger studies that are able to answer to this question, and given the irreversibility of a possible harm to the child, treating during early childhood and revaluating the thyroid function after myelination of the central nervous system is completed (by 36 to 40 months of age) can be a prudent behavior (56e). LT4 treatment must be started immediately if FT4 or TT4 levels are low, given the known adverse effect of untreated decompensated CH on neurodevelopment and somatic growth.

CH is defined on the basis of serum FT4 levels as severe when FT4 is <5 pmol/l, moderate when FT4 is 5 to 10 pmol/l and mild when FT4 is 10 to 15 pmol/l respectively. Determination of serum thyroglobulin (Tg) is useful, if below the detection threshold, to suggest athyreosis or a complete thyroglobulin synthesis defect. Measurement of Tg is most helpful when a defect in Tg synthesis or secretion is being considered. In the latter condition the serum Tg concentration is low or undetectable despite the presence of a normal or enlarged, eutopic thyroid gland. Serum Tg concentration also reflects the amount of thyroid tissue present and the degree of stimulation. For example, Tg is undetectable in most patients with thyroid agenesis, intermediate in babies with an ectopic thyroid gland and may be elevated in patients with abnormalities of thyroid hormonogenesis not involving Tg synthesis and secretion. Considerable overlap exists, and so, the Tg value needs to be considered in association with the findings on imaging. In patients with inactivating mutations of the TSH receptor discordance between findings on thyroid imaging and the serum Tg concentration has been described in some but not all studies (56f).

Clinical findings are usually difficult to appreciate in the newborn period except in the unusual situation of combined maternal-fetal hypothyroidism. Many of the classic features (large tongue, hoarse cry, facial puffiness, umbilical hernia, hypotonia, mottling, cold hands and feet and lethargy), when present, are subtle and develop only with the passage of time. In addition to the aforementioned findings, nonspecific signs that should suggest the diagnosis of neonatal hypothyroidism include: prolonged, unconjugated hyperbilirubinemia, gestation longer than 42 weeks, feeding difficulties, delayed passage of stools, hypothermia or respiratory distress in an infant weighing over 2.5 kg ( 57). A large anterior fontanelle and/or a posterior fontanelle > 0.5 cm is frequently present in affected infants but may not be appreciated. In general, the extent of the clinical findings depends on the cause, severity and duration of the hypothyroidism. Babies in whom severe feto-maternal hypothyroidism was present in utero tend to be the most symptomatic at birth. Similarly, babies with athyreosis or a complete block in thyroid hormonogenesis tend to have more signs and symptoms at birth than infants with an ectopic thyroid, the most common cause of congenital hypothyroidism. Unlike acquired hypothyroidism, babies with congenital hypothyroidism are of normal size. However, if diagnosis is delayed, subsequent linear growth is impaired. The finding of palpable thyroid tissue suggests that the hypothyroidism is due to an abnormality in thyroid hormonogenesis or in thyroid hormone action.

Bone maturation reflects the duration and the severity of hypothyroidism. Signs of delayed epiphyseal maturation on knee x-rays, persistence of the posterior fontanelle, a large anterior fontanelle, and a wide sagittal suture all reflect delayed bone maturation. The absence of one or both knee epiphyses has been shown to be related to T4 concentration at diagnosis and to IQ outcome, and is thus a reliable index of intrauterine hypothyroidism.

Imaging Techiniques in CH

Imaging studies are helpful to determine the specific etiology of CH. Both scintigraphy and ultrasound (US) should be considered in neonates with high TSH concentrations. Ideally, the association of US and scintigraphy gives the best information in a child with primary hypothyroidism. Scintigraphy shows the presence/absence (athyreosis), position (ectopic gland, in any point from the foramen caecum at the base of the tongue to the anterior mediastinum) and rough anatomic structure of the thyroid gland.

US, in experienced hands, is a valid tool in defining size and morphology of a eutopic thyroid gland, however, US alone is less effective in detecting ectopic glands. Color Doppler US improves the effectiveness of US (57a).

It is important to remember that an attempt to obtain an imaging of the thyroid in a newborn should never delay the initiation of treatment. Scintigraphy should be carried out within 7 days of starting LT4 treatment. Scintigraphy may be carried out with either 10-20 MBq of technetium-99m (99mTc) or 1-2 MBq of iodine-123 (I123). Tc is more widely available, less expensive, and quicker to use than I 123. Scintigraphy with I123, if available, is usually preferred because of the greater sensitivity and because, I123, unlike of technetium-99 is organified. Therefore, imaging with this isotope allows quantitative uptake measurements and tests for both iodine transport defects and abnormalities in thyroid oxidation. An enrichment of the tracer within the salivary gland can lead to misinterpretation, especially on lateral views, but this can be avoided by allowing the infant to feed before scintigraphy, thus empting the salivary glands and keeping the child calm under the camera. The perchlorate discharge test is considered indicative for a organification defect when a discharge of > 10% of I123 administred dose occurs in a thyroid in normal position (when perchlorate is given at 2 hours).

Excess iodine intake through exposure (i.e from antiseptic preparation), maternal TSH receptor blocking antibodies, inactivating mutation in the TSH receptor and in the sodium/iodide symporter (NIS), and TSH suppression from LT4 treatment can give interfere with the I123 uptake, showing no uptake in the presence of a thyroid in situ (apparent athyreosis).

Thyroid ultrasonography is performed with a high frequency linear array transducer (10-15 MHz) and allows a resolution of 0.7 to 1mm. Thyroid tissue is more echogenic than muscle and less echogenic than fat. In the case of absence of the thyroid fat tissue can be misdiagnosed as dysplastic thyroid gland in situ. Distinguish between thyroid hypoplasia and dysplastic non thyroidal tissue in a newborn requires an enormous experience, and reevaluation at later age can result in a different diagnosis (57a).

Combining scintigraphy and thyroid ultrasound improve diagnostic accuracy, and helps to address further investigations, including molecular genetic studies. Infants found to have a normal sized gland in situ in the absence of a clear diagnosis should undergo further reassessment of the thyroid axis and imaging at a later age.

Therapy

Replacement therapy with L-thyroxine (L-T4) should be begun as soon as the diagnosis of congenital hypothyroidism is confirmed. In babies whose initial results on newborn screening are suggestive of severe hypothyroidism therapy should be begun immediately without waiting for the results of the confirmatory serum. Severe hypothyroidism is defined by T4 <5 mcg/dL (64 nmol/L) and/or TSH >40 mU, or. accordingly with ESPE guidelines(51g,51k), CH is defined on the basis of serum FT4 levels as severe when FT4 is <5 pmol/l, moderate when FT4 is 5 to 10 pmol/l and mild when FT4 is 10 to 15 pmol/l. As noted above, treatment need not be delayed in anticipation of performing thyroid imaging studies as long as the latter are done within 5-7 days of initiating treatment (before suppression of the serum TSH). Parents should be counseled regarding the causes of congenital hypothyroidism, the importance of compliance and the excellent prognosis in most babies if therapy is initiated sufficiently early and is adequate and educational materials should be provided (58). An initial dosage of 10-15 mcg/kg/day of L-T4 is generally recommended to normalize the T4 as soon as possible. The highest dose is indicated in infants with severe disease, and the lower in those with a mild to moderate form. L-T4 Tablets can be crushed and given via a small spoon, with suspension, if necessary in a few milliliters of water or breast milk or formula or juice, but care should be taken that all of the medicine has been swallowed. Thyroid hormone should not be given with substances that interfere with its absorption, such as iron, calcium, soy, or fiber. Drugs such as antacids (aluminium hydroxide) or infantile colic drops (simethicone) can interfere with L-thyroxine absorption. Many babies will swallow the pills whole or will chew the tablets with their gums even before they have teeth. Reliable liquid preparations are not available commercially in the US, although they have been used successfully in Europe. L-T4 can also be administred in liquid form, but only if pharmaceutically produced and licensed L-T4 solutions are available. A brand name rather a generic formulation of L-T4 is recommended because they are not bioequivalent (58a).

The aims of therapy are to normalize the T4 as soon as possible, to avoid hyperthyroidism where possible, and to promote normal growth and development. When an initial dosage of 10-15 mcg/kg is used, the T4 will normalize in most infants within 1 week and the TSH will normalize within 1 month, Subsequent adjustments in the dosage of medication are made according to the results of thyroid function tests and the clinical picture. Often small increments or decrements of L-thyroxine (12.5 mcg) are needed. This can be accomplished by 1/2 tablet changes, by giving an alternating dosage on subsequent days, or by giving an extra tablet once a week.

As stated in ESPE guidelines: “ L-T4 alone is recommended as the medication of choice and should be started as soon as possible, no later than two weeks of life or immediately after confirmatory test results in infants identified in a second routine screening test. L-T4 should be given orally. If intravenous administration is necessary, the dose should be no more than 80% of the oral dose”. Serum or plasma FT4 (or TT4) and TSH concentration should be determined at least 4 hours after the last L-T4 administration. TSH should be maintained in the age-specific reference range and FT4 in the upper half of the age- specific reference range. “The first follow up examination is indicated after 1-2 weeks after the start of LT4 treatment and then every 2 weeks until TSH levels are completely normalized and then every 1- 3 months until 12 months of age. Between the age of one and three years, children should undergo frequent clinical and laboratory evaluations (every 2 to 4 months).” Thereafter, evaluations should be carried out every 3 to 12 months until growth is completed. “More frequent evaluations should be carried out if compliance is questioned or abnormal values are obtained. Any reduction of L-T4 dose should not be based on a single increase of FT4 concentration during treatment. “Measurements should be performed after 4-6 weeks any change in the dosage or in the L-T4 formulation”.

Re-evaluation and Trial Off Therapy

In hypothyroid babies in whom an organic basis was not established at birth and in whom transient disease is suspected, a trial off replacement therapy can be initiated after the age of 3 years when most thyroxine-dependent brain maturation has occurred, as shown by magnetic risonance imaging studies (56e). Re-evaluation is recommended if the treatment was started in a sick child (i.e. preterm), if thyroid antibodies were detectable, if no diagnostic assessment was completed, and in children who have required no increase in L-T4 dosage since infancy. Re-evaluation is recommended also in the case of a eutopic gland with or without goiter, if not enzyme defects have been detected, if any other cause of transient hypothyroidism is suspected.

Re-evaluation is not necessary if venous TSH concentration has risen during the first year of life, due to either LT4 underdosage or poor compliance. To perform a precise diagnosis LT4 treatment is suspended for 4-6 weeks, and biochemical testing and thyroid imaging are carried out. To establish the presence of primary hypothyroidism, without defining the cause, L-T4 dose may be decreased by 20-30% for 2 to 3 weeks. If TSH serum levels rise to > 10 mU/L during this period, the hypothyroidism can be confirmed.

Prognosis

Although all are agreed that the mental retardation associated with untreated congenital hypothyroidism has been largely eradicated by newborn screening, controversy persists as to whether subtle cognitive and behavioral deficits remain, particularly in the most severely affected infants (59-64). Both the initial treatment dose and early onset of treatment (before 2 weeks) are important. Time to normalization of circulating thyroid hormone levels, the initial free T4 concentration, maternal IQ, socioeconomic and ethnic status have also been related to outcome (59,62,63,64). The long term problems for these babies appear to be in the areas of memory, language, fine motor, attention and visual spatial. Inattentiveness can occur both in patients who are overtreated and those in whom treatment was initiated late or was inadequate. In addition to adequate dosage, assurance of compliance and careful long-term monitoring are essential for an optimal developmental outcome. More details about long term follow up are reported in ESPE guidelines (51g,51K). Progressive hearing loss in CH should be recognized and corrected, because strongly influenced the outcome). Recently, extensive reports on long term outcome of congenital hypothyroidism in young adults have been published (64a,64b). In the French cohort of 1202 CH young adults, hearing impairment was found at a mean age of 23.4 years in 9.5% versus 2.5% of general population, and the risk of developing hearing impairment was three times higher in these patients than in general population (64c). Also interesting data about pregnancy outcomes in young women with CH came out from the French cohort (64d).

CAUSES OF PERMANENT CONGENITAL HYPOTHYROIDISM

Permanent congenital thyroidal (primary) hypothyroidism can be the consequence of a disorder in thyroid development and/or migration (thyroid dysgenesis), or due to defects at every step in thyroid hormone synthesis (thyroid dyshormonogenesis). Although congenital hypothyroidism (CH) is in the great majority of cases a sporadic disease, the recent guidelines (51g,51k) for CH recommend genetic counseling in targeted cases. Positive family history for CH, association with cardiac or kidney malformation, midline malformation deafness, neurological sigs (i.e., choreoathetosis, hypotonia, any sign of Albright hereditary osteodystrophy, lung disorders, suggest genetic counseling, in order to assess the risk of recurrence and to provide further information about a possible genetic etiology of CH. Recently a targeted next-generation (NGS) panel, covering all exons of the major CH genes, has been proposed as a useful tool to identify the genetic etiology of CH (64e). Lowering TSH cut off value at screening increases the diagnosis of CH with eutopic thyroid. A targeted next-generation (NGS) panel has been applied to patients with CH and thyroid in situ (64f).

 

Thyroid Dysgenesis

 

Unlike in iodine-deficient areas of the world where endemic cretinism continues to be a major health hazard, the majority (85 to 90%) of cases of permanent congenital hypothyroidism in North America, Western Europe and Japan are due to an abnormality of thyroid gland development (thyroid dysgenesis). Thyroid dysgenesis may result in the complete absence of thyroid tissue (agenesis, 20-30%) owing to a defect in survival of the thyroid follicular cells precursors) or it may be partial (hypoplasia); the latter often is accompanied by a failure to descend into the neck (ectopy) mostly located in a sublingual position as a result of a premature arrest of its migratory process. Lowering of cut off TSH values for newborn screening increases the percentage of CH with thyroid in situ. Females are affected twice as often as males. In the United States, thyroid dysgenesis, is less frequent among African Americans and more common among Hispanics and Asians. Babies with congenital hypothyroidism have an increased incidence of cardiac anomalies, particularly atrial and ventricular septal defects (65). An increased prevalence of renal and urinary tract anomalies has also been reported recently (66). Most cases of thyroid dysgenesis are sporadic. Familial cases represent 2%. Discordance between monozigotic twins is inexplained (67). Although both genetic and environmental factors have been implicated in its etiology, in most cases the cause is unknown (67a).

The occasional familial occurrence, the higher prevalence of thyroid dysgenesis in babies of certain ethnic groups and in female versus male infants as well as the increased incidence in babies with Down syndrome (68) all suggest that genetic factors might play a role in some patients. Thyroid transcription factors would appear to be obvious candidate genes in view of their important role in thyroid organogenesis and in thyroid-specific gene expression. To date, however, abnormalities in these genes have been found in only a small proportion of affected patients, usually in association with other developmental abnormalities (68a).

Thyroid transcription factors (TTF) such as NKX2-1 (or formerly TTF1/TITF1), FOXE1 (Forkhread Box E1, formerly TTF2/TITF2), PAX8 (Paired box gene 8), and NKX2-5, are expressed during early phases of thyroid organogenesis (budding and migration), instead thyroid stimulating hormone receptor gene (TSHR) is expressed during the later phases of thyroid development. All these genes are involved in normal thyroid development and in thyroid dysgenesis. Alternately, epigenetic modifications, early somatic mutations or stochastic developmental events may play a role. Five monogenic forms due to mutations in TSHR, NXK2-1, PAX8, FOXE-1. NXK2-5 have been reported. Monogenic forms represent less than 10% in TD (68a).


TABLE 1. GENETIC CAUSES OF CONGENITAL HYPOTHYROIDISM

 

1.1 PRIMARY HYPOTHYROIDISM Gene locus Inheritance
Monogenic forms of thyroid dysgenesis    
·       Thyroid stimulating hormone receptor (TSHR)   AR
·       NK2 1 (NK2-1, TTF1) brain-lung thyroid syndrome 14q13 AD
·       Paired box gene 8 (PAX8) 2q11.2 AD
·       Forkhead boxE1 (FOXE1, TTF2) (Bambforth-Lazarus syndrome) 9q22 AR
·       NK2 homeobox 5 (NKX2-5)    
New candidates gene    
·       Nertrin 1 (NTN-1)    
·       JAG1 20p.12.2  
Inborn errors of thyroid hormonogenesis    
·       Sodium/Iodide symporter (SLC5A5,NIS 19p13.2 AR
·       Thyroid peroxidase (TPO) 2p25 AR
·       Pendred syndrome (SLC26A4,PDS) 7q31 AR
·       Thyroglobulin (TG) 8q24 AR
·       Iodothyrosine deiodinase (IYD,DEHAL1) 6q24-25 AR
·       Dual oxidase 2 (DUOX2) 15q15.3 AR/AD
·       Dual oxidase maturation factor 2 (DUOXA2)   AR/AD
B1.2 CENTRAL HYPOTHYROIDISM    
Isolated TSH deficiency    
·       TRHR 14q31 AR
·       TSHB 1p13 AR
Isolated TSH deficiency or combined pituitary hormone deficiency    
Immunoglobulin superfamily member1 (IGSF1) gene defects Xq26.1 X-Linked
Combined pituitary hormone deficiency    
·       POU1F1 3p11 AR,AD
·       PROP1 5q AR
·       HESX1 3p21.2-21.2 AR/AD
·       LHX3 9q.34 AR
·       LHX4 1q25 AD
·       SOX3   X-linked
·       OTX2   AD

 

 

Monogenic Forms of Thyroid Dysgenesis

 

Thyroid stimulating hormone receptor resistance (TSHR gene #OMIM 603372)

Described in 1968, is mostly caused by biallelic inactivating mutations in the TSH receptor gene (TSHR). TSH affects follicular thyroid cell proliferation and many cellular processes, including thyroidal iodine uptake, thyroglobulin iodination, and reuptake of iodinated thyroglobulin. Phenotype varies from mild hyperthyrotropinemia with normal thyroid gland to severe CH with thyroid hypoplasia and absence of tracer uptake at scintigraphy (apparent athyreosis).

Inactivating TSHR mutations are the most frequent cause of monogenic TD and non syndromic CH, with prevalence in CH cohorts around 4 % (68b). Clinically a classic and a non-classic TSH resistance form are described, based on different TSHR mutations (68c). Both Gs and Gq proteins are involved Heterozygous non polymorphic TSHR mutations were found in a high frequency (11.8-29%) in children and adolescents with isolated non-autoimmune hyperthyrotropinemia (68d).

NKX2-1 (OMIM 600635)

NKX2-1 (previously TITF-1, TTF-1) gene encodes for a transcription factor of the NK family. It is involved in early development of brain, thyroid and lung. In thyrocytes, NKX2-1 activates the transcription of TG, TPO, TSHR and PDS genes. In the lung is important for the branching of the lobar bronchi and regulates the expression of surfactant proteins in pneumocytes. In the brain, NKX2 is expressed in basal ganglia and forebrain and it is involved in the specification and migration of neurons. Haploinsufficiency of NKX2-1 is responsible for the brain-lung-thyroid (BLT) syndrome (OMIM 610978) characterized by CH, infant respiratory distress syndrome and benign hereditary chorea. NKX2-1 defects occur either as a sporadic cases or as familial cases inherited in an autosomal-dominant manner. The clinical presentation ranges from the complete BLT syndrome (50%) to incomplete forms with brain and thyroid disease (30%) or only benign hereditary chorea (13%), the mildest expression of the syndrome. TD ranges from hypoplasia (about 35%) to normal morphology (>50% of patients) (68e). Recently, a case of BLT syndrome has been reported with thyroid ectopy (68f).

The severity of symptoms varies widely, even in families with the same disease causing mutation. In a detailed study (68g) lung disease, if present at birth, manifests as a surfactant deficiency syndrome and can be fatal. Asthma, recurrent pneumonia in childhood, spontaneous pneumothorax, and interstitial lung disease has also been reported. Neurologic forms present with muscular hypotonia in early infancy and psychomotor delay, which progresses to benign hereditary chorea between 1 and 5 years. Additional non classical features including hypodontia o oligodontia, microcephaly, growth retardation, genitourinary abnormalities, skeletal disorders, and congenital heart defects have been reported in patients with large deletions on chromosome 14, including the NKX2-1 gene and surrounding genes. Interestingly, a more extended phenotype associating hypothalamic symptoms, frequent recurrence of fever without infection, dysrhytmic sleep, and abnormal height in patients with point NKX2-1 mutations was described (68g). So far, 116 NKK2-1 genetic anomalies have been reported worldwide (68h).

PAX8 (OMIM218700)

Paired box gene 8 (PAX8) codes for a TTF of the paired homeodomain transcription factors family. PAX8 is expressed during thyroid organogenesis in the median anlage and in the kidney development. In synergy with NKX2-1, PAX 8 influences the expression of TPO, TG and NIS in thyroid follicular cells. The prevalence of PAX8 mutations in CH patients is about 1%, ranging from 0.3 to 3.4% (68b,68i).Thyroid hypoplasia is the more common phenotype, but athyreosis to normal morphology have also been reported. Thyroid function varies from severe hypothyroidism to mild hypertropinemia, and different phenotypes can be found in the same family. The association with kidney malformations is possible, but remains a facultative sign in CH patients with PAX8 mutations. So far, 29 mutations have been reported (68h).

FOXE1 (OMIM#602617)

The Forkhead Box 1 E1 (FOXE1) gene encodes for a transcription factor of the forkhead/winged-helix transcription factor family. Foxe1 is expressed in the thyroid primordium, in the pharyngeal endoderm derivates such as the palate and the esophagus and in the hair follicoles (68j). Foxe1 interacts with TG and TPO promoters and with regulatory regions of DUOX2 and NIS genes (68k).

The Bamforth-Lazarus syndrome is caused by FOXE1 mutations. It is characterized by CH (usually athyreosis), cleft palate and spinky hair. Bifid epiglottis and choanal atresia can be present. So far, six mutation with loss of function (68h) and 1 mutation with gain of function have been reported in patients with Bamforh-Lazarus syndrome, showing the effect of FOXE1 gene dosage in this disorder (68m).

 

NKX2-5 (OMIM #600584)

Because an increased prevalence of heart congenital malformations have been reported in CH, genes involved in heart organogenesis as NKX2-5 have been suggested as a cause of CH. NKX2-5, that encodes for a transcription factor with a major role in heart development has been investigated in a cohort of 241 patients with thyroid dysgenesis. Heterozygous missense mutations had been reported in this study in 4 patients with ectopy and athyreosis, and all mutations were transmitted from one of the parents but only 1 patient had minor cardiac phenotype (68n).

A major pathogenetic role of NKX2-5 mutations in thyroid dysgenesis has been questioned: given the absence of TD in carriers of NKX2-5 mutations, and the high number of TD patients without mutations. Better defining the role of NKX2-5 in thyroid organogenesis need further studies (68o).

 

New Candidates Genes

 

NTN-1

A new gene Netrin-1 (NTN-1), has been recently identified in a patient with thyroid ectopy and ventricular sept defect, and considered as a possible link between thyroid and heart defects (68p).

JAG1 (20p12.2 OMIM 6019220)

A role for the Notch pathway in thyroid morphogenesis has recently been demonstrated in zebrafish (68q). JAG1 is a gene encoding one single pass transmembrane ligand of the notch receptors. Heterozygous variations of JAG1 are the cause of Alagille syndrome type 1, an autosomal dominant disorder characterized by paucity of intrahepatic bile ducts, cardiac malformations as pulmonary artery stenosis, coarctaction of aorta, atrio-ventricular septal defects and Fallot tetralogy. Many other organs as eye, skeleton, kidney, nervous system can be involved, with a characteristic facial phenotype. A study investigating the role of JAG1 loss of function variations in the pathogenesis of congenital thyroid defects in Alagille syndrome and in patients with congenital hypothyroidism supported the role of this gene as a predisposing factor in congenital hypothyroidism (68r). The authors reported, in a series of 21 patients affected with Alagille syndrome non autommune hypothyroidism in 6 patients (28%), two of them with thyroid hypoplasia. Analyzing 100 patients with congenital hypothyroidism for JAG1 variants they found JAG1 variants in 4. Interestingly, 2 of them had cardiac malformations.

 

Inborn Errors of Thyroid Hormonogenesis

 

Inborn errors of thyroid hormonogenesis (thyroid dyshormonogenesis) are responsible for most of the remaining cases (15%) of neonatal thyroidal hypothyroidism. Unlike thyroid dysgenesis, mostly a sporadic condition, these inborn errors of thyroid hormonogenesis are commonly associated with an autosomal recessive form of inheritance, consistent with a single gene abnormality. DUOX2 mutations can be transmitted in autosomal dominant way. Thyroid dysormonogenesis is caused by genetic defects in proteins involved in all steps of thyroid hormone synthesis (68s) often associated with goiter formation. Goiter can be present in utero or at birth.

.A number of different defects have been characterized based on radioiodine uptake and perchlorate test and include:
1) Iodide transport defect (ITD)
(SLC5A5, Sodium/Iodide Symporter NIS), that shows failure to concentrate iodide, with low or absent radioiodine uptake, also in salivary glands and gastric mucosa;


2) Iodide organification defect (IOD)
with normal radioiodine uptake and altered perchlorate discharge test. In these patients, less than 90% of the iodide is organified and remains stored in the follicles. Total IOD is defined as >90% of the given dose back to the blood. Partial IOD is defined as 10-90% of radioiodine washout after perchlorate application. Total IOD is due to Thyroid peroxidase mutations (TPO) and Dual Oxidase 2 (DUOX2), partial IOD is due to DUOX2, Dual Oxidase Maturation Factor 2 mutations (DUOX2A), SLC26A4, pendrin and TPO defects.


3
) Forms with normal radioiodine uptake and a normal perchlorate test:
Thyroglobulin TG mutations, iodide recycling defects IYD, Iodothyrosine Deiodinase mutations (DEHAL1).
4) Iodide Transport Defect (OMIM 274400)

ITD is rather a rare form and is due a mutation of the Sodium/Iodide Symporter (NIS). The NIS is expressed at the basolateral membrane of the thyrocite and it is responsible for the active iodide uptake through the membrane into the thyrocite (69). This form of hypothyroidism is characterized by goiter and absence of radioiodine uptake. In contrast with athyreosis, uptake is lacking also in salivary glands and in the stomach (white scintigraphy).

The severity of hypothyroidism depends on the residual function of the mutated NIS protein, ranging with severe to mild forms, often detected in infancy or childhood.


Pendred Syndrome (OMIM274600)

Pendred syndrome is defined by the association of familial profound deafness with multinodular goiter. It is caused by biallelic mutation in the pendrin gene (70-71). Pendred syndrome is the only form of thyroid dyshormonogenesis associated with a malformation. The inner ear presents a characteristic malformation of the cochlea.

Congenital hypothyroidism is present in only 30% of cases, goiter occurs often in childhood. Thyroid phenotype is variable. Perchlorate test shows a partial organification defect. Pendred syndrome is the most frequent etiology of familial deafness. SLC264A mutations (mostly in the heterozygous state) have been also described in isolated enlargement of the vestibular aqueduct, with no thyroid disease (71a). More than 150 mutations have been described. Specific mutation cluster in Asia (H723R), and Europe (L236P, T416P, IVS8, 1-GA) (71b).

Thyroid peroxidase mutations (OMIM #274500)

Thyroid peroxidase (TPO) is a heme peroxidase that regulates two rate-limiting step of thyroid hormones synthesis, first the organification of iodide to iodinated thyrosyl residuates such as MIT and DIT, and then the coupling of MIT and DIT to T3 and T4. TPO action needs hydrogen peroxide as the final electron acceptor. Mutations are mostly in the heme-binding domain of the protein, encoded by exons 7-9 (71c). TPO mutations are a common form of thyroid dyshormonogenesis. Severe congenital hypothyroidism with goiter is present in the great part of patients, with a total IOD. Recently, a few patients with partial IOD have been reported (72,72a).

Dual Oxidase 2 and Dual Oxidase Maturation Factor 2 mutations (OMIM#607200 and 274900)

DUOX2 (formerly THOX2) and DUOXA2 are components of a nicotinamide adenine dinucleotide phosphate oxidase complex that produces hydrogen peroxide indispensable for TPO action.

The first mutation in DUOX2 has been reported in 2002. Heterozygous mutations have been found in a part of the patients, suggesting autosomal dominant and autosomal recessive inheritance both possible in this form (72b). Monoallelic mutations usually cause mild hypothyroidism; biallelic mutations are present in mild to severe hypothyroidism. In some cases, DUOX2 mutations lead to transient congenital hypothyroidism, with normalization of thyroid function at follow up. DUOX2 mutations usually cause partial IOD, but total IOD is also reported (72c). Mutations in DUOXA2 were described in patients detected by neonatal screening with mild CH. Partial IOD was found in these cases (72d).


Thyroglobulin Mutations (OMIM#274700)

Thyroglobulin (Tg) is a glycoprotein synthetized by the thyrocytes that serves as a matrix for thyroid hormones synthesis and storage in the follicles (68t). Tg is also in part released in the blood and it is a useful marker of thyroid tissue.

In CH, Tg serum determination can differentiate between a true and apparent athyreosis, the last with same residual dysgenetic tissue and Tg detectable.

In dyshomonogenesis, Tg levels are low in patients with Tg mutations, but are normal or high in the other defects of hormonogenesis (68t). CH due to Tg mutations is usually severe, with goiter in utero or at birth. Different mechanisms cause hypothyroidism in Tg mutations: a )Tg synthesis defects alter protein synthesis; b) Tg transport defects limit Tg excretion in the follicle; c) a abnormal structure of T impairs coupling of MIT and DIT; d) a large imperfect DNA inversion in Tg gene is a novel cause for CH (72e-72g).


Iodothyrosine Deiodinase Mutations cause Iodide Recycling defects (OMIM#274800)

DEAHAL 1(IYD) is the enzyme that regulates the recycling of iodide from MIT and DIT to the follicle, thus allowing the synthesis of thyroid hormones. Dietary Iodine is scarce in nature and it is the limitating factor for thyroid hormones synthesis. Failure of DEAHL1 cause iodotyrosine deiodinase deficiency, characterized by hypothyroidism, goiter and mental retardation. It is important to stress that these patients are not detected by neonatal screening for CH, probably because the maternal iodine protect for a period the newborn. Diagnosis is reported between 18 month and 16 years with hypothyroidism and mental retardation (72h). The first mutations in DEAHAL1 has been reported in 2008 (72i).

The use of MIT and DIT -as early markers to identify iodotyrosine deiodinase deficiency before mental retardation-is under investigation.

 

Central Congenital Hypothyroidism (CCH)

 

Central hypothyroidism (CCH) is caused by an insufficient thyroid hormone biosynthesis due to a defective stimulation by TSH, in the presence of an otherwise normal thyroid. This condition includes all causes of congenital hypothyroidism due to a pituitary or hypothalamic pathology (secondary or tertiary hypothyroidism). CCH was previously considered a very rare disease with a prevalence initially estimated to be 1:100000 in newborns (73). In more recent data, CCH had an incidence that could reach 1:16.000, as shown from results from screening for congenital hypothyroidism applied in the Neetherlands, based on T4/TSH/TBG determination (73a).

Also with this sophisticated method of screening, CCH is sometime not identified at birth, because the limiting step is “how low is a low T4”, low enough to be considered an effective cutoff value and allow the determination of TSH and TBG. Many cases are diagnosed in infancy or childhood, if not later in adulthood (73b). The majority of screening programs are based on TSH determination and a high index of suspicion is needed to identify CCH in the preclinical phase. Delayed diagnosis may result in neurodevelopment delay. More than 50% of children with CCH have moderate or severe hypothyroidism, so, if not treated, the risk of neurodevelopmental delay should not be underestimated (73c).

In the majority of cases identified early, TSH deficiency is a part of combined pituitary hormone deficiency. A timely correction of ACTH and cortisol deficiency, and/or GH deficiency may avoid life threatening emergencies.

CCH can be transient (mostly due to drugs or maternal hyperthyroidism), or permanent.

 

Genetic Central Hypothyroidism (Table 1)

 

Isolated Thyroid Stimulating Hormone deficiency

Two forms of non-efficient TSH are known, the first one is very rare and is due to defects in the receptor that regulates the action of TRH on thyrotropes (TRHR), the second form is due to several mutations in the β-subunit of TSH.

 

a)Thyrotropin-releasing hormone receptor (TRHR ) gene defects. TRHR mediates the correct action of TRH on thyrotropes toward the synthesis, glycosylation and secretion of TSH.

This is a very rare cause of central hypothyroidism. Mutations in TRHR gene have been described so far in 3 patients from 2 families, the first from Canada, the second from Italy, with autosomal recessive inheritance (73d,73e). Index cases were detected at 9 and 11 years for short stature and symptoms related to hypothyroidism. Neonatal hypothyroidism could not be proven because neonatal screening was based on TSH level. No psychomotor delay or intellectual deficit was reported in these children. TSH was in the low normal range with a suspected low bioactivity; T4 or FT4 were low, TRH test showed no response of TSH and PRL.

A compound heterozygosis with 2 different mutations in TRHR gene was found in the Canadian patient. The first mutation in the paternal allele was a premature stop codon R17X that completely inactivated protein function. The second one, on the maternal allele was a complex combination of mutations: 9-nucleotide deletion followed by a point mutation resulting in an in-frame deletion of three aminoacids (Ser115-Thir117) plus a missense change located at the cytoplasmatic end of the transmembrane domain of the receptor (73d).The Italian patient had a homozygous nonsense mutation (pR17X).

A novel homozygous missense mutation (P81R) in TRHR has been published in a female infant presented at age 19 days with prolonged jaundice due to isolated hyperbilirubinemia. Thyroid function showed CCH (TSH 2.2 mU/L (RR 0.4-3.5). FT4 7.9 pmol/L (RR10.7-21.8). She was treated with L-thyroxine and at 4 years of age growth and neurological development are in the normal range. The location of the mutated aminoacid (proline 81) in the second transmembrane helix underlines the functional role of this helix in hormone binding and receptor activation (73f).

 

b)TSH β Gene defects

TSH is a glycoprotein hormone with an α subunit common with FSH, LH and hCG, and a β-subunit, specific for TSH.

Mutations of the β-subunit of TSH are the cause of the most severe forms of central congenital hypothyroidism. All mutations described so far caused central hypothyroidism, either because truncated protein or alterations in key structural features required for heterodimeric integrity occur (74, 74a, 74b).

Another consequence of mutations of the β-subunit of TSH is the modification of bioactivity and immunoreacivity of the TSH heterodimer. Diagnosis of central hypothyroidism can be complicated because of impaired TSH immunoreactivity and/or bioactivity. For instance, TSH is not detectable when the heterodimer formation between TSHα and TSH β subunit is completely not allowed from mutations (i.e. p.G49, p.32), in other cases some mutant heterodimeric TSH is present and measurable in an immunoassay dependent manner (i. e. p.Q69, c.373 delT). TSH can be measurable but not shows normal bioactivity (74a). Interestingly, a variant (c223A>G, pR75) causing normal bioactive TSH, but with impaired immunoreactivity has been described (74c, 74d). These individuals are euthyroid, but erroneous diagnosis and inappropriate treatment have been reported.

In children affected with CCH due to mutations of the β-subunit of TSH, psychomotor and mental retardation can occur, depending on the time of diagnosis and treatment. Most are clinically diagnosed after 3 months of age because they are not identified by neonatal screening based on increased TSH levels. Hyperplastic pituitary, high levels of serum glycoprotein alfa-subunit and hypoplasic thyroid gland have been reported (74a). Several mutations have been reported, including missense, nonsense and frameshift mutations (74,74a), as well as slice mutations (74b). Recently a homozygous TSHβ mutation was found (74e). A novel missense mutation (c.2T>C) in which a methionin codon, is replaced by a threonine, has been very recently reported in a child with very low levels of TSH (0.45mU/l, (NR 0.4-3.5) and FT4.(<5.1 pmol/l (NR 13.8-22.5). This child was diagnosed at 3.5 months of age because feeding difficulties, somnolence, constipation and severe growth retardation. She was treated with L-thyroxine with a good response in growth, but she has severe neurodevelopmental deficits, with bilateral sensorineural deafness, nistagmus, motor and language development delay at age of 10. She was on autistic/Asperger spectrum and needed special education at school (74f).

 

Immunoglobulin superfamily member1 (IGSF1) gene defects

IGSF1 (immunoglobulin (Ig) superfamily member1) gene mutations were described in 2012 as a cause of central hypothyroidism, with an incidence of about 1:100.000 (75,75a). IGSF1 gene is located on X chromosome (Xq26.1) and encodes for a plasma membrane glycoprotein that is mainly expressed in the pituitary, brain and testes.

Several pathogenetic mutations in IGSF1 gene have been reported so far (75,75a,75b). An extensive case series, expanding the clinical phenotype has been published very recently (75c,75d). The first patient was diagnosed by neonatal screening in the Neetherlands where a screening program for congenital hypothyroidism that includes T4 determination (T4/TBG/TSH) is applied. Many other cases of central hypothyroidism were identified in this family and in others with an age at diagnosis ranging from 3 weeks to 69.9 years (75). Typical phenotype in adult males includes central hypothyroidism and macroorchidism (>30 ml by Prader orchidometer). Hypoprolactinemia and GH deficiency can be present. GH deficiency is usually transient and detectable in childhood. Body mass index tends to be elevated. Testicular volume is normal in childhood and increases at a normal age in puberty, but the testosterone rise is delayed, as well as the pubertal growth spurt and the appearance of secondary sexual characteristics. Thyroid volume is small, TSH is usually detectable, TSH response to TRH is diminished. No clear correlation genotype-phenotype has been established.

IGSF1 gene is located on X chromosome. Male are affected but 1/3 of females heterozygous carriers shows a milder phenotype, with central hypothyroidism, delayed menarche, mild prolactin deficiency and benign ovarian cysts sometime requiring surgical resection. Recently a familial form of isolated central hypothyroidism with neurological phenotypes due to a novel IGSF1 gene mutation has been reported from Israel (75e).

 

TSH deficiency in combined pituitary hormone deficiency

Central congenital hypothyroidism can be a component of combined pituitary hormone deficiency. This form represents the majority of cases detected by the neonatal screening when T4 determination is used (73b). Early diagnosis in these cases helps to prevent dangerous hypoglycemic and adrenal crisis due to associated GH and ACTH deficiency.

TSH deficiency can be present at diagnosis or occurs later, as a component of an evolving phenotype. In a minority of patients, mutations of known transcriptor factors (i.e POU1F1 ,PROP1,HESX1,LHX3,LHX4,SOX3 and OTX2) that are involved in pituitary development can be identified (76) (See Table 1).

Mutations in early transcriptor factors cause developmental abnormalities, i.e., septo-optic dysplasia, midline defects, holoprosencephaly, ocular or skeletal defects, intellectual impairment, associated with variable hypopituitarism. Mutations in HESX1, OTX2 and SOX3 have been found in patients with septo-optic dysplasia and TSH deficiency (76).

TSH deficiency in association with other pituitary hormone deficiencies may be associated with abnormal midline facial and brain structures (particularly cleft lip and palate, and absent septum pellucidum and/or corpus callosum) and should be suspected in any male infant with microphallus and persistent hypoglycemia (76a). One of the more common of these syndromes, septo-optic dysplasia, has been related in some cases to a mutation in the HESX 1 homeobox gene in some cases (76b). Other genetic causes of congenital hypopituitarism include molecular defects in the genes for the transcription factors LHX3 (76c), LHX4, POU1F 1 (76d) or PROP 1 (76d). POU1F 1 (Pit-1 in mice) is essential for the differentiation of thyrotrophs, lactotrophs and somatotrophs while PROP 1, a homeodomain protein that is expressed briefly in the embryonic pituitary, is necessary for POU1F 1 expression.

Defects of Thyroid Hormone Transport in Serum

For complete coverage of this and related areas visit the chapter entitled: “Defects of thyroid hormone transport in serum” by Samuel Refetoff, MD in this book.

Inherited abnormalities of the iodothyronine-binding serum proteins include TBG deficiency (partial or complete), TBG excess, transrethyretin (TTR) (prealbumin) variants and familial dysalbuminemic hyperthyroxinemia (FDH). In these conditions the concentration of free hormones is unaltered, but the abnormal total thyroxine concentrations can be misleading during neonatal screening and in the evaluation of thyroid function.

Impaired Sensitivity to Thyroid Hormone

For complete coverage of this and related areas visit the chapter entitled: “Impaired sensitivity to thyroid hormone: defects of transport, metabolism and action” by.Alexandra M. Dumitrescu, MD and Samuel Refetoff, MD, in this book.

Impaired sensitivity to thyroid hormone, previously known as “reduced sensitivity to thyroid hormone”, include defects in thyroid hormone action, transport and metabolism. They are classified in a)Thyroid hormone cell membrane transport defect (THCMTD),b) thyroid hormone metabolism defect (THMD) and c) thyroid hormone action defect that include Resistance to thyroid hormone (RTH) (77).The first defect, recognized almost 50 years ago, produces reduced sensitivity to TH and was given the acronym RTH, for resistance to thyroid hormone (77a, 77b). Its major cause, found in more than 3,000 individuals, is mutations in the TH receptor ß (THRB) gene. More recently mutations in the THRA gene were found to produce a different phenotype owing to the distinct tissue distribution of this TH receptor (77c, 77d). Two other gene mutations, affecting TH action, but acting at different sites have been identified in the last 10 years. One of them, caused by mutations in the TH cell-membrane transporter MCT8, with decreased T4 uptake into brain cells produces severe psychomotor defects (77e,77f). In this syndrome, first described as Allan Herdon Dudley syndrome, (77g) mutations in the monocarboxylate transporter 8 (MCT 8 gene, located on the X-chromosome), have been associated with male- limited hypothyroidism and severe neurological abnormalities, including global developmental delay, dystonia, central hypotonia, spastic quadriplegia, rotary nystagmus and impaired gaze and hearing (77e, 77f). Heterozygous females had a milder thyroid phenotype and no neurological defects. A defect of the intracellular metabolism of TH, identified in 11 members from 9 families, is caused by mutations in the SECISBP2 gene required for the synthesis of selenoproteins, including TH deiodinases (77h). Knowledge of the molecular mechanisms involved in mediation of TH action allows the recognition of the phenotypes caused by genetic defects in the involved pathways. While these defects have opened the avenue for novel insights into thyroid physiology, they continue to pose therapeutic challenges.

CAUSES OF TRANSIENT NEONATAL HYPOTHYROIDISM

Transient neonatal hypothyroidism should be distinguished from a ‘false positive’ result in which the screening result is abnormal but the confirmatory serum sample is normal. Causes of transient abnormalities of thyroid function in the newborn period are listed in Table 2. While iodine deficiency, iodine excess, drugs and maternal TSH receptor blocking antibodies are the most common causes of transient hypothyroidism, in some cases the cause is unknown.

 

TABLE 2. CAUSES OF TRANSIENT HYPOTHYROIDISM IN THE NEWBORN

· 2.1 PRIMARY HYPOTHYROIDISM
· ·       Prenatal or postnatal iodine deficiency or excess
· ·       Maternal antithyroid medication
· ·       Maternal TSH receptor blocking antibodies
· ·       Mild gene mutations (i.e. DUOX2, TSH-R )
· ·       Maternal hypothyroidism
· ·       Prematurity, VLBW
· ·       Drugs, (i.e. Dopamine, steroids)
  ·       Hypothyroxinemia (low T4, normal TSH)
· 2.2 CENTRAL HYPOTHYROIDISM
· ·       Prenatal exposure to maternal hyperthyroidism
· ·       Prematurity (particularly <27 weeks gestation)
· ·       Drugs

Iodine Deficiency or Excess

In addition to iodine deficiency, both the fetus and newborn infant are sensitive to the thyroid-suppressive effects of excess iodine, whether administered to the mother during pregnancy, lactation or directly to the baby (78). This occurs, in part because, as noted earlier, recovery from the thyroid-suppressive effect of iodine does not mature before 36 weeks gestation; however, other factors, including increased skin absorption are also likely to play a role. Reported sources of iodine have included drugs (e.g., potassium iodide, amiodarone), radiocontrast agents and antiseptic solutions (e.g., povidone-iodine) used for skin cleansing or vaginal douches. In contrast to Europe, iodine-induced transient hypothyroidism has not been documented frequently in North America (79). For other information see the chapter “Iodine deficiency disorders” in this book.

Maternal Antithyroid Medication

Transient neonatal hypothyroidism may develop in babies whose mothers are being treated with antithyroid medication (either propylthiouracil, PTU or methimazole, MMI) for the treatment of Graves ’ disease. Even maternal PTU doses of 200 mg or less have been associated with an effect on neonatal thyroid function, illustrating the increased fetal sensitivity to these drugs (80). Babies with PTU- or MMI-induced hypothyroidism characteristically develop an enlarged thyroid gland and if the dose is sufficiently large, respiratory embarrassment may occur. Both the hypothyroidism and goiter resolve spontaneously with clearance of the drug from the baby’s circulation. Usually replacement therapy is not required.

Maternal TSH Receptor Antibodies

Maternal TSH receptor blocking antibodies, a population of antibodies closely related to the TSH receptor stimulating antibodies in Graves’ disease, (81) may be transmitted to the fetus in sufficient titer to cause transient neonatal hypothyroidism. The incidence of this disorder has been estimated to be 1 in 180,000 (81a,81b). TSH receptor blocking antibodies most often are found in mothers who have been treated previously for Graves’ disease or who have the non goitrous form of chronic lymphocytic thyroiditis (primary myxedema). Occasionally these mothers are not aware that they are hypothyroid and the diagnosis is made in them only after congenital hypothyroidism has been recognized in their infants (81b). Unlike TSH receptor stimulating antibodies that mimic the action of TSH, TSH receptor blocking antibodies inhibit both the binding and action of TSH (see below). Because TSH-induced growth is blocked, these babies do not have a goiter. Similarly, inhibition of TSH-induced radioactive iodine uptake may result in a misdiagnosis of thyroid agenesis (81c). Usually the hypothyroidism resolves in 3 or 4 months. Babies with TSH receptor blocking-antibody induced hypothyroidism are difficult to distinguish at birth from the more common thyroid dysgenesis but they differ from the latter in a number of important ways (Table 3). They do not require lifelong therapy, and there is a high recurrence rate in subsequent offspring due to the tendency of these antibodies to persist for many years in the maternal circulation. Unlike babies with thyroid dysgenesis in whom a normal cognitive outcome is found if postnatal therapy is early and adequate, babies with maternal blocking-antibody induced hypothyroidism may have a permanent deficit in intellectual development if feto-maternal hypothyroidism was present in utero (27).

 

TABLE 3. CLINICAL FEATURES OF THYROID DYSGENESIS VERSUS TSH RECEPTOR

  • BLOCKING ANTIBODY INDUCED CONGENITAL HYPOTHYROIDISM
  Dysgenesis Blocking Ab
Severity of CH + to ++++ + to ++++
Palpable thyroid No No
123I uptake None to low None to normal
Clinical Course Permanent Transient
Familial risk No Yes
TPO Abs

Variable

 

 

 

 

eele

Variable
TSH Receptor Abs Absent Potent

Transient Central Hypothyroidism Due to Maternal Hyperthyroidism

Occasionally, babies born to mothers who were hyperthyroid during pregnancy develop transient hypothalamic-pituitary suppression (81,81a,81b,81c). This hypothyroxinemia is usually self-limited, but in some cases it may last for years and require replacement therapy (82). In general the titer of TSH receptor stimulating antibodies in this population of infants is lower than in those who develop transient neonatal hyperthyroidism (see below).

Prematurity

Hypothyroxinemia in the presence of a ‘ normal ’ TSH occurs most commonly in premature infants in whom it is found in 50% of babies of less than 30 weeks gestation. Often the free T4 when measured by equilibrium dialysis is less affected than the total T4 (83). The reasons for the hypothyroxinemia of prematurity are complex. As well as hypothalamic-pituitary immaturity mentioned earlier, premature infants frequently have TBG deficiency due to both immature liver function and undernutrition, and they may have “sick euthyroid syndrome”. They may also be treated with drugs that suppress the hypothalamic-pituitary-thyroid axis. Hypothyroxinemia of prematurity may be associated with adverse neurodevelopmental outcomes. L-T4 treatment overall has no proven benefit and can be harmful (83a). Long term outcome evaluation in young adults did not find association between transient hypothyroxinemia of prematurity and neurodevelopmental outcome (83b). Whether or not premature infants with hypothyroxinemia should be treated remains controversial at the present time (83c,83d,83e). Although several retrospective, cohort studies have documented a relationship between severe hypothyroxinemia and both developmental delay and disabling cerebral palsy in preterm infants <32 weeks gestation a causal relationship could not be determined since the serum T4 in premature infants, as in adults, has been shown to reflect the severity of illness and risk of death (83c).

Drugs

Drugs that suppress the hypothalamic-pituitary axis include known agents such as steroids and dopamine, but also aminophylline, caffeine and diamorphine, other commonly used in sick premature infants (84).

Other causes of hypothyroidism in infancy

Chronic lymphocytic thyroiditis

Chronic lymphocytic thyroiditis (CLT) is a rare disease in infancy, but if not recognized and treated, can cause severe hypothyroidism in a short time with permanent brain damage (85). CLT can be associated with other autoimmune disease as type 1 diabetes or a manifestation of IPEX syndrome (85a). In the cases described by Foley, no goitrous was found. Clinical manifestations and biochemical hypothyroidism (TSH ranged from >42 to 928 mU/L) were severe and very high levels of antibodies were detectable.

Lymphocytic thyroiditis has also been described in newborns with severe defects in tolerance and autoimmunity with immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome, a polyglandular disorder characterized by early-onset diabetes and colitis (85a,85b). IPEX disorders are an expanding spectrum of disease with mutations in FOXP3, CD25 deficiency, STAT5 deficiency and other.

Hepatic hemangiomas: consumptive hypothyroidism

Hepatic emangioendothelioma is a rare tumor typically presenting in infancy. Hypothyroidism is caused by a production of type 3 deiodinase by the vascular tumor (85c). D3 deoidinase increases inactivation of T4 and T3 to reverse T3 andT2 and large amount of LT4 (up to 94/ µg/kg/day) are needed to compensate this inactivation (85d). Frequent monitoring is required, adapting the LT4 treatment to the growing proliferative phase of the tumor. Today hemangioendotheliomas in infancy may successfully being treated with steroids and propranolol and may undergo spontaneous regression. Some babies underwent liver transplantation.

HYPERTHYROIDISM

Transient Neonatal Hyperthyroidism

Unlike congenital hypothyroidism which usually is permanent, neonatal hyperthyroidism almost always is transient and results from the transplacental passage of maternal TSH receptor antibodies. Hyperthyroidism develops only in babies born to mothers with the most potent stimulatory activity in serum (86,87,87a). This corresponds to 1-2% of mothers with Graves’ disease, or 1 in 50,000 newborns, an incidence that is approximately four times higher than is that for transient neonatal hypothyroidism due to maternal TSH receptor blocking antibodies (81a). Some mothers have mixtures of stimulating and blocking antibodies in their circulation, the relative proportion of which may change over time. Not surprisingly, the clinical picture in the fetus and neonate of these mothers is more complex and depends not only on the relative proportion of each activity in the maternal circulation at any one time but on the rate of their clearance from the neonatal circulation postpartum. Thus, one affected mother gave birth, in turn, to a normal infant, a baby with transient hyperthyroidism, and one with transient hypothyroidism (87b). In another neonate, the onset of hyperthyroidism did not become apparent until 1-2 months postpartum when the higher affinity blocking antibodies had been cleared from the neonatal circulation (87c). In the latter case, multiple TSH receptor stimulating and blocking antibodies were isolated from the maternal peripheral lymphocytes. Each monoclonal antibody recognized different antigenic determinants (“epitopes”) on the receptor and had different functional properties (87d).

Occasionally, neonatal hyperthyroidism may even occur in infants born to hypothyroid mothers. A prospective study showed that 40% of patients treated for Graves’ disease with radioactive iodine had TRAb detectable after 5 years (87e). In these situations, the maternal thyroid has been destroyed either by prior radioablation, surgery or by coincident destructive autoimmune processes so that potent thyroid stimulating antibodies, present in the maternal circulation, are silent in contrast to the neonate whose thyroid gland is normal (87d). Fetal/neonatal thyrotoxicosis can occur also in newborn from hypothyroid mothers with chronic lymphocytic thyroiditis (87f).

Clinical manifestations

Maternal TSH receptor antibody-mediated hyperthyroidism may present in utero. Fetal hyperthyroidism is suspected in the presence of fetal tachycardia (pulse greater than 160/min) especially if there is evidence of failure to thrive. Obstetric complications are common. Fetal goiter can by monitored by ultrasound. In the neonate infant most often the onset is during the first one-two weeks of life but can occur by 45 days. This is due both to the clearance of maternally-administered antithyroid drug (propylthiouracil, PTU, methimazole or carbimazole) from the infant ’s circulation and to the increased conversion of T4 to the more metabolically active T3 after birth. Rarely, as noted earlier, the onset of neonatal hyperthyroidism may be delayed until later if higher affinity blocking antibodies are also present. In the newborn infant, characteristic signs and symptoms include tachycardia, irritability, poor weight gain, and prominent eyes (Figure 5). Goiter, when present, may be related to maternal antithyroid drug treatment as well as to the neonatal Graves’ disease itself.

Figure 15.5. A baby with neonatal hyperthyroidism secondary to maternal Graves ‘disease. Note the prominent eyes in the baby and mother in whom Graves’ disease developed after radioiodine therapy for Hodgkin’s disease. In contrast, the father was unaffected.

Rarely, infants with neonatal Graves’ disease present with thrombocytopenia, jaundice, hepatosplenomegaly, and hypoprothrombinemia, a picture that may be confused with congenital infections such as toxoplasmosis, rubella, or cytomegalovirus (87g). In addition, arrhythmias and cardiac failure may develop and may cause death, particularly if treatment is delayed or inadequate. In addition to a significant mortality rate that approximates 20% in some older series, untreated fetal and neonatal hyperthyroidism is associated with deleterious long-term consequences, including premature closure of the cranial sutures (cranial synostosis), failure to thrive, and developmental delay (87h). The half-life of TSH receptor antibodies is 1 to 2 weeks. The duration of neonatal hyperthyroidism, a function of antibody potency and the rate of their metabolic clearance, is usually 2 to 3 months but may be longer.

Laboratory Evaluation

TSH receptor antibodies (TRAb) are Immunoglobulin of G class and freely cross the placenta. Different type of TRAb can be found: TRAb that bind to the TSH receptor and stimulates the production of thyroid hormones, (TSH receptor stimulating antibodies, TSI), TRAb that bind to the TSH receptor, do not stimulate the production of thyroid hormones and can block the binding of TSH (TSH receptor blocking antibodies TBI) .TSH receptor neutral antibodies have also been identified which do not block TSH binding and are unable to stimulate cAMP production (88)..

The receptor binding assays usually used to measure TRAb are not able to distinguish between TSH-receptor stimulating and blocking or neutral antibodies. Bioassays that measure TSI activity based on cAMP on cultured cells can be useful if TRAb are not detectable (88a,88b). The recent guidelines for management of hyperthyroidism (88c) and the updated guidelines for the management of thyroid disease during pregnancy released from the American Thyroid Association ATA (33b) both suggest to anticipate the determination of TRAb in pregnant women with Graves’ disease at 18-22 weeks instead of 20-24 weeks of gestation because a severe case of fetal Graves’ disease has occurred at 18 weeks of pregnancy (88d).

Because of the importance of early diagnosis and treatment, infants at risk for neonatal hyperthyroidism should undergo both clinical and biochemical assessment as soon as possible.

All neonates born from a woman with TRAb positivity in pregnancy should undergo determination of TRAb from cord blood at delivery. If TRAb are negative, the risk to neonatal hyperthyroidism is negligible (Sensitivity is around 100%). FT3, FT4 and TSH determination from cord blood did not predict neonatal hyperthyroidism. Determination of FT4 increase on day 3 to 5 seems to better indicate the onset of hyperthyroidism (88e) Situations that should prompt consideration of neonatal hyperthyroidism are listed in Table 4. A high index of suspicion is necessary in babies of women who have had thyroid ablation because in them a high titer of TSH receptor antibodies would not be evident clinically. Similarly, women with persistently elevated TSH receptor antibodies and with a high requirement for antithyroid medication are at an increased risk of having an affected child. The diagnosis of hyperthyroidism is confirmed by the demonstration of an increased concentration of circulating T4 (and free T4, and T3, if possible) accompanied by a suppressed TSH level in neonatal or fetal blood. The latter can be obtained by cordocentesis if someone experienced in this technique is available. Results should be compared with normal values during gestation. Fetal ultrasonography may be helpful in detecting the presence of a fetal goiter and in monitoring fetal growth. Demonstration in the baby or mother of a high titer of TSH receptor antibodies will confirm the etiology of the hyperthyroidism and, in babies whose thyroid function testing is normal initially, indicate the degree to which the baby is at risk.

 

TABLE 4. SITUATIONS THAT SHOULD PROMPT CONSIDERATION OF NEONATAL HYPERTHYROIDISM

·     Unexplained tachycardia, goiter or stare
·     Unexplained petechiae, hyperbilirubinemia, or hepatosplenomegaly
·     History of persistently high TSH receptor antibody titer in mother during pregnancy
·     History of persistently high requirement for antithyroid medication in mother during pregnancy
·     History of thyroid ablation for hyperthyroidism in mother
·     History of previously affected sibling

 

As noted in the case of TSH receptor blocking antibody-induced congenital hypothyroidism, the receptor binding assays are a cost-effective, rapid and technically feasible approach. In general, babies likely to become hyperthyroid have the highest TSH receptor antibody titer whereas if TSH receptor antibodies are not detectable, the baby is most unlikely to become hyperthyroid (87g, 89,89a). In the latter case, it can be anticipated that the baby will be euthyroid, have transient hypothalamic-pituitary suppression or have a transiently elevated TSH, depending on the relative contribution of maternal hyperthyroidism versus the effects of maternal antithyroid medication, respectively (89). Close follow up of all babies with abnormal thyroid function tests or detectable TSH receptor antibodies is mandatory.

Therapy

In the fetus, treatment is accomplished by maternal administration of antithyroid medication. Until recently PTU was the preferred drug for pregnant women in North America, but current recommendations suggest the use of MMI rather than PTU after the first trimester because of concerns about potential PTU-induced hepatotoxicity (123) (discussed under Graves’ disease, below). The goals of therapy are to utilize the minimal dosage necessary to normalize the fetal heart rate and render the mother euthyroid or slightly hyperthyroid.

In the neonate MMI (0.5 to 1.0 mg/kg/day) has been used initially in 3 divided doses. If the hyperthyroidism is severe, a strong iodine solution (Lugol’ s solution or SSKI, 1 drop every 8 hours) is added to block the release of thyroid hormone immediately. Often the effect of MMI is not as delayed in infants as it is in older children or adults, a consequence of decreased intrathyroidal thyroid hormone storage. Therapy with both antithyroid drug and iodine is adjusted subsequently, depending on the response. Propranolol (2 mg/kg/day in 2 or 3 divided doses) is added if sympathetic overstimulation is severe, particularly in the presence of pronounced tachycardia. If cardiac failure develops, treatment with digoxin should be initiated, and propranolol should be discontinued. Rarely, prednisone (2 mg/kg/day) is added for immediate inhibition of thyroid hormone secretion. Measurement of TSH receptor antibodies in treated babies may be helpful in predicting when antithyroid medication can be safely discontinued (87). Lactating mothers on antithyroid medication can continue nursing as long as the dosage of PTU or MMI does not exceed 400 mg or 40 mg, respectively. The milk/serum ratio of PTU is 1/10 that of MMI, a consequence of pH differences and increased protein binding, so one might anticipate less transmission to the infant, but concerns about potential PTU toxicity need to be considered. At higher dosages of antithyroid medication, close supervision of the infant is advisable.

A review about management of neonates born to mothers with Graves’ disease has been recently published (89b).

Permanent neonatal hyperthyroidism

Rarely, neonatal hyperthyroidism is permanent and is due to a germline mutation in the TSH receptor (TSH-R) resulting in its constitutive activation (90,90a,90b,90c). A gain of function mutation of the TSH-R should be suspected if persistent neonatal hyperthyroidism occurs in the absence of detectable TSH-R antibodies in the maternal circulation. Prematurity, low birth weight and advanced bone age are common. Most cases result from a mutation in exon 10 which encodes the transmembrane domain and intracytoplasmic tail of the TSH-R, a member of the G protein coupled receptor superfamily (90,90a,90b,90c). Less frequently, a mutation encoding the extracellular domain has been described (90d). An autosomal dominant inheritance has been noted in many of these infants; other cases have been sporadic, arising from a de novo mutation.

Early recognition is important because the thyroid function of affected infants is frequently difficult to manage medically (90a-90c), and, when diagnosis and therapy is delayed, irreversible sequelae, such as cranial synostosis and developmental delay may result (90c). For this reason early, aggressive therapy with either thyroidectomy or even radioablation has been recommended (90c).

Two clinical forms were described: the first one is the “familial non-autoimmune autosomal dominant hyperthyroidism” (FNAH). High variable age of manifestation from neonatal period to 60 years, with. variability also within the same family is reported. Goiter is present in children, with nodules in older age.

The second one is “Persistent sporadic congenital non autoimmune hyperthyroidism” (PSNAH) includes forms with sporadic (de novo) germline mutations in the TSH-R.

PSNAH is characterized by fetal-neonatal onset or within 11 months and more severe hyperthyroidism requiring early aggressive therapy. Guidelines about this rare condition have recently been published (90e).

McCune Albright syndrome

McCune Albright is a syndrome due to somatic activating mutations in Gsα gene, can rarely presents with neonatal hyperthyroidism (90f).

THYROID DISEASE IN CHILDHOOD AND ADOLESCENCE

Hypothyroidism and Thyroiditis

Chronic lymphocytic thyroiditis is the more common cause of acquired hypothyroidism in children and adolescents. Occasionally, patients with disorders classified as congenital hypothyroidism, i.e thyroid dysgenesis, inborn error of thyroid hormonogenesis, central hypothyroidism may be recognized later in childhood and adolescence.

Causes of hypothyroidism in children and adolescents are listed in table 5.

 

TABLE 5. CAUSES OF HYPOTHYROIDISM IN CHILDHOOD AND ADOLESCENCE

 

PRIMARY HYPOTHYROIDISM
A) Congenital
Thyroid dysgenesis
Inborn error of thyroid hormonogenesis
Thyroidal Gsα protein abnormalities (pseudohypoparathyroidism 1B)
B) Acquired
Autoimmune
Chronic Lymphocytic Thyroiditis
Reversible autoimmune hypothyroidism (silent and postpartum thyroiditis, cytokine-induced thyroiditis
Infiltrative: Cystinosis, Hemocromathosis, Thalassemia,-Langerhans Cell Histiocytosis
Infective: acute, subacute thyroiditis

Post ablative

Surgery

Thyroiditis due to I 131, external irradiation of non-thyroidal tumors (i.e. lymphomas, brain tumors, TBI
Iodine deficiency and iodine excess
Drugs: antithyroid agents, lithium, natural and synthetic goitrogenic chemicals, tyrosine kinase inhibitors, lithium, thionamides, aminosalicylic acid, aminoglutethimide
Goitrogens (cassava, water pollutants, cabbage, sweet potatoes, cauliflower, broccoli, soya beans)
CENTRAL HYPOTHYROIDISM
A)Congenital
Pituitary hypoplasia, septo-optic dysplasia, basal encephalocele
Functional defects in TSH biosynthesis and release
Mutations in genes encoding for TRH receptor, TSHß, pituitary transcription factors (Pit-1, PROP1, LHX3, LHX4, HESX1), or LEPr, IGSF1
B)Acquired
Tumors (pituitary adenoma, craniopharyngioma, meningioma, dysgerminoma, glioma, metastases)
Trauma surgery, irradiation, head injury
Infections
Vascular damage ischemic necrosis, hemorrhage, stalk interrruption,
Hypotalamic disorders
Drugs: dopamine; glucocorticoids; bexarotene; L-T4 withdrawal
“Peripheral” (extrathyroidal) hypothyroidism
Consumptive hypothyroidism (massive infantile hemangioma)
Mutations in genes encoding for MCT8, SECISBP2, TRα or TR β (impaired sensitivity to thyroid hormones)

 

 

Chronic Lymphocytic Thyroiditis

 

Autoimmune thyroid diseases (AITD) are defined by the lymphocytic infiltration of the thyroid (91). Usually antibodies against thyroid antigens as thyroperoxidase (TPOAb), thyroglobulin (TgAb), and anti-TSH receptor (TRAb) are detectable in serum. Thyroid antibodies in serum correlate with the presence of lymphocytic infiltrate in the thyroid gland. The clinical spectrum of AITD ranges from hypothyroidism to hyperthyroidism and include chronic lymphocytic thyroiditis (CLT) and Graves’ disease. CLT is the most common cause of hypothyroidism in children and adolescents (91,91a).

Graves’ disease and CLT are closely associated and in fact overlapping syndromes .Patients can move from one to the other category, depending upon the stage of their illness. For example, an individual might first be observed with thyroid enlargement and positive antibody tests for anti-thyroglobulin or anti-TPO antibodies, and thus qualify as having CLT. At a later stage, this individual might become hyperthyroid (Hashitoxicosis) and fit in the category of Graves’ disease. Or, the patient with hyperthyroidism might have progressive destruction of the thyroid, or develops blocking antibodies, and become hypothyroid or ultimately develop myxedema.

Incidence

The prevalence of CLT in children and adolescents was reported to be 1.2% by Rallison in 1975 (91b). In this 6 year-survey study 5179 school children were examined in Arizona. Goiter was evaluated by palpation (91b). More recently, a study from Sardinia in 8040 children and adolescents aged 6-15 years reported TPOAb detectable in 2.9% (91c). Similar results were found in Berlin with a prevalence of TPOAb of 3.4% (mean age 11 years) (91d) and in Greece after correction of iodine deficiency. In this study examining 440 children and adolescents aged 5-18 years a prevalence of TPOAb and TgAb was reported to be 4.6% and 4.3% respectively. The prevalence of CLT, confirmed by ultrasound was 2.5% (91e).

In childhood the most common age at presentation is adolescence, but the disease may occur at any age, even infancy. CLT in infancy is rare, but can cause in a short time severe hypothyroidism and permanent damage to CNS if not recognized and treated (85). The female/male ratio in AITD is up to 6:1, but In prepubertal age the female/male ratio is lower than reported in adolescents and adults.

Etiology and Pathogenesis

CLT is thought to be caused by a combination of genetic susceptibility and environmental factors. Both thyroid-specific genes and genes involved in immune recognition and/or response have been identified (91f, 91g). (See chapter Autoimmunity, by A Weetman for an exhaustive information). Some genes are common to both disorders and some tend to predominate only in Graves’ disease. AITD has a striking predilection for females, but in prepubertal age the female/male ratio is lower. A family history of autoimmune thyroid disease (both chronic lymphocytic thyroiditis and Graves’ disease) is found in 30% to 40% of patients. A study about familial clustering of juvenile AITD found thyroid antibodies detectable in 56% of mothers and 25% of fathers. Interestingly, HLA DQ alleles and antibody status in fathers influenced the susceptibility to AITD in children (91h). Siblings recurrence in childhood is 20-30% (91i). AITD are often associated with other autoimmune disorders. The plethora of associations and their familial occurrence indicates that a defect in the immune system may be more likely than primary defects in each organ, as these diseases often share similar genetic associations, including HLA, CTLA-4, PTPN22 and CD25 gene polymorphisms. It is also clear however that there is a difference in the kind of clustering of other autoimmune disease in CLT and Graves’ disease, presumably related to differences between these two types of thyroid disease in genetic predisposition (91j,91k). There is also an increased incidence of CLT in patients with certain chromosomal abnormalities as Down syndrome (91l) Turner syndrome (91m), Klinefelter syndrome (91n) as well as in patients with Noonan syndrome (91o).

Environmental factors as infection, environmental toxins, substances as iodine, selenium, stress, smoking, estrogens, drugs (amiodarone, interferon alfa, lithium) have been suggested as precipitating factors for CLT (91p). The precise environmental trigger has not been yet established. An epigenetic mechanism may be implicated (91q).

Clinical Manifestations

Both goitrous (Hashimoto’s thyroiditis) and nongoitrous (atrophic thyroiditis, also called primary myxedema) as variants of chronic lymphocytic thyroiditis have been distinguished. The term “Hashimoto’s thyroiditis” is often used as a synonymous of CLT, not necessary linked to the presence of goiter (91, 91a). Goiter, present in approximately two-thirds of children with CLT is caused by lymphocytic infiltration that may be extensive, with lymphoid germinal centers, TSH stimulation, or production of antibodies that stimulate thyroid growth (92). Progressive thyroid cell damage, with cell mediated cytotoxicity and follicular cell apoptosis, can change the apparent clinical picture from goitrous hypothyroidism to that of “atrophic” thyroiditis. Atrophic thyroiditis, or primary hypothyroidism/mixedema, is considered to be the end stage of CLT (91).

Children with chronic lymphocytic thyroiditis may be euthyroid, or may have subclinical or overt hypothyroidism. Occasionally, children may experience an initial thyrotoxic phase due to the discharge of preformed T4 and T3 from the damaged gland. Alternatively, as indicated above, thyrotoxicosis may be due to concomitant thyroid stimulation by TSH receptor stimulatory antibodies (Hashitoxicosis).

The onset of hypothyroidism in childhood is insidious. Affected children often are recognized either because of the detection of a goiter on routine examination or because of a poor interval growth rate present for several years prior to diagnosis (92a). Because the deceleration in linear growth tends to be more affected than weight gain, these children can be relatively overweight for their height, although they rarely are significantly obese (Figure 6). If the hypothyroidism is severe and longstanding, immature facies with an underdeveloped nasal bridge and immature body proportions (increased upper-lower body ratio) may be noted. Dental and skeletal maturation are delayed, the latter often significantly. Patients with central hypothyroidism tend to be even less symptomatic than are those with primary hypothyroidism.

Figure 15-6 Sequential changes in physical appearance in a young girl who presented at 15 years of age with amenorrhea and hyperprolactinemia secondary to severe hypothyroidism. Note her poor linear growth since at least 11 years of age.

The classical clinical manifestations of hypothyroidism can be elicited on careful evaluation, though they often are not the presenting complaints. These include sluggishness, lethargy, cold intolerance, constipation, dry skin or hair texture, and periorbital edema. Bradycardia and delayed deep tendon reflexes can be present. In severe, long-standing hypothyroid children pericardial and pleural effusions may occur. School performance is not usually affected, in contrast to the severe irreversible neuro-intellectual sequelae that occur frequently in inadequately treated babies with congenital hypothyroidism. Causes of hypothyroidism associated with a goiter (CLT, inborn errors of thyroid hormonogenesis, thyroid hormone resistance) should be distinguished from non goitrous causes (primary myxedema, thyroid dysgenesis, central hypothyroidism). The typical thyroid gland in a longstanding chronic lymphocytic thyroiditis is diffusely enlarged and has a rubbery consistency. Although the surface is classically described as ’pebbly’ or bosselated, occasionally asymmetric enlargement occurs and must be distinguished from thyroid neoplasia. Alternatively, the thyroid may be normal in size and consistency or not palpable at all. A palpable lymph node superior to the isthmus (“Delphian node”) is often found and may be confused with a thyroid nodule. The thyroid gland, in thyroid hormone synthetic defects, on the other hand, tends to be softer and diffusely enlarged.

In patients with severe hypothyroidism of longstanding duration, the sella turcica may be enlarged due to thyrotrope hyperplasia. There is an increased incidence of slipped femoral capital epiphyses in hypothyroid children. The combination of severe hypothyroidism and muscular hypertrophy which gives the child a “Herculean” appearance is known as the Kocher-Debre-Semelaigne syndrome (92b).

Puberty tends to be delayed in hypothyroid children in proportion to the retardation in the bone age, although in longstanding severe hypothyroidism, sexual precocity has been described. Females with sexual precocity have menstruation, and breast development but relatively little sexual hair. Multicystic ovaries, the etiology of which is unknown, may be demonstrated on ultrasonography. In other cases, galactorrhea or severe menses have been the presenting features. In boys, testicular enlargement may be found (92c). An elevated serum prolactin, the latter possibly due to raised TRH which is known to stimulate prolactin as well as TSH, has been described in some cases, but gonadotropin levels are not consistently elevated. It has been hypothesized that this syndrome of pseudopuberty in hypothyroid patients is due to cross- interaction of the extremely elevated serum TSH with the FSH receptor (92d). Consistent with the latter hypothesis, there is little increase in serum testosterone as might be expected if the FSH, but not luteinizing hormone (LH) receptor is involved and serum gonadotropins are frequently not increased.

Long term follow up studies of children with chronic lymphocytic thyroiditis have suggested that while most children who are hypothyroid initially remain hypothyroid, spontaneous recovery of thyroid function may occur, particularly in those with initial compensated hypothyroidism (93,93a, 93b). A recent five-years prospective study in children and adolescents affected with CLT showed that thyroid dysfunction increased from 27.3% to 47.4% (93c). Therefore, close follow up is necessary.

Although chronic inflammation, leading to neoplastic transformation, is a well-established clinical phenomenon, if CLT can increase the risk for thyroid nodules and thyroid cancer remains controversial. In the past autoimmune thyroiditis has been thought to be protective from thyroid cancer, but several studies both in adults and in children suggested the opposite. Thyroid nodules in healthy children in iodine replete regions are detected in up to 1.6% (94). High prevalence of thyroid nodules, ranging from 13% to 31%, has been reported in children and adolescents with CLT. In a multicentric pediatric retrospective study from Italy, nodules were found in 115/365 patients with CLT (31.5%), and papillary thyroid carcinoma in 11/115 (9.5%) (94a). In a recent study from Turkey, thyroid nodules were detected in 39/300 (13%) of cases of pediatric CLT and papillary thyroid carcinoma was diagnosed in 2 of the 12 cases that underwent FNAB (94b). Recently, in. a retrospective study from United States examining ultrasound characteristic of the thyroid in 154 children and adolescents with goiter, nodules were reported in 20/154 (13%) and PTC in 4/154 (2.5% ) of children. In this study, the same prevalence of nodules (17%) was found in TPOAb positive and TPOAb negative children. Interestingly, one case of PTC was first classified at ultrasound as pseudonodule. Only 15 % of nodules and none of the papillary thyroid carcinoma in these series (PTC) were palpable, although PTC has a diameter ranging from 1.2 to 2.6 cm (94c). A rare variant of PTC, the diffuse sclerosing variant, has also been reported in children with CLT(94d).

Associated Disease

AITD are frequently associated with other common autoimmune disorders as type 1 diabetes (94e,94f) and celiac disease. AITD can be also the first manifestation of autoimmune polyendocrine syndromes (APS1 and 2) (95,95a,95b). A pletora of other autoimmune conditions, organ or non-organ specific disease, can be associated with AITD in childhood and adolescence. CLT is more frequently associated with adrenal and β cell autoimmunity than Graves’ disease (91j). Early identification and treatment of these disorders may be critical and even preserve children from life-threatening events. Long term surveillance is required, because a second autoimmune disorder may occur any time.

Type 1 diabetes. CLT is the most common associated autoimmune disease in type 1 diabetes. In a ten-years observational study of children and adolescents with type 1 diabetes (mean age at diagnosis 10 years), the prevalence of TPOAb and TgAb at diagnosis was 15.4% and 14.4% respectively. The cumulative incidence increased especially in females in mid puberty. In this study about 14% of patients required treatment with L-thyroxine (95c). Children with AITD had islet cell antibodies in 2.3% (95d). Screening for AITD is suggested at diagnosis and every 2-3 years if negative (ADA and ISPAD recommendation). Thyroid function should be checked every year or more frequently if needed, because thyroid dysfunction (both hypothyroidism and hyperthyroidism) affects metabolic control. In overt hypothyroidism hypoglycemia can occur because glucose absorption may be slow and the sensitivity and rate of degradation of insulin is increased. Hepatic gluconeogenesis and peripheral glucose utilization are also reduced. Long term dyslipidemia may affect cardiovascular risk in these patients.

Hyperthyroidism in type 1 diabetic children can precipitate acute complications. In a study on 60456 children and adolescents with type 1 diabetes, hyperthyroidism was diagnosed in 276 (0.46%). Hyperthyroid state was associated with diabetic ketoacidosis, hypoglycemia and hypertension (95e). Life-long surveillance is required.

Celiac disease. Another strong association is with celiac disease, which is found 3 times more commonly in patients with AITD. Intriguingly the autoantibodies which are the hallmark of celiac disease, directed against transglutaminase, can bind to thyroid cells and thus could be implicated directly in thyroid dysfunction (95f). A recent meta-analysis showed that the prevalence of celiac disease in AITD patients in higher in children (6.2%) than in adults (1.2%) (95g). Prevalence of AITD in celiac patients is about 20%. The presence of celiac disease in type 1 diabetes seems to increase the risk for AITD (95h). Undiagnosed celiac disease causes malabsorption with or without gastrointestinal symptoms. Delayed linear growth may be the first manifestation as unexplained change in L-T4 requirement (95i).

Addison’s disease. Addison’s disease is also associated with AITD. In an old report, the prevalence of adrenal antibodies in children with AITD, was found to be 2.3%, while the great majority of children affected with Addison’s disease presented with CLT (95d). Addison’ disease is more often a component of autoimmune polyendocrine syndromes (APS1 and 2) (95,95a,95b). Addison's disease and/or type 1 diabetes mellitus and AITD occasionally co-exist and form the classical autoimmune polyendocrine syndrome type2 (Schmidt syndrome). Undiagnosed adrenal insufficiency, is a life-threatening condition and can be exacerbated by L-thyroxine therapy, because L-thyroxine increases cortisol clearance. Moreover, symptoms of overt hypothyroidism can overlap with adrenal insufficiency manifestations. Adrenal insufficiency is a rare but non-obvious diagnosis in childhood and should be considered in when autoimmune disorders are diagnosed.

Autoimmune gastritis. Autoimmune gastritis was first described in association with AITD as thyrogastric syndrome. Common clinical manifestations in adults are iron deficient or pernicious anemia (95j). Perhaps 45% of patients with autoimmune thyroiditis have circulating gastric parietal cell antibodies. Also in children with AITD, early manifestations of gastric autoimmunity has been reported, with a prevalence of gastric parietal cell antibodies of 30%. In this series, 45% of PCA positive children presented with increased gastrin plasma levels (a marker of atrophic body gastritis) (95l).

Autoimmune polyendocrine syndromes (APS1 and 2)

CLT can be the first manifestation of autoimmune polyendocrine syndromes (APS1 and 2) (95,95a,95b). APS1 tends to present in early childhood and is characterized primarily by mucocutaneous candidiasis, hypoparathyroidism and adrenal deficiency. APS1 is also defined as autoimmune polyendocrinopaty-candidiasis-ectodermal dystrophy (APECED). APS1 results from mutations in the AIRE (autoimmune regulator) gene. It is a rare autosomal dominant disorder with incomplete penetrance (96). In APS1, chronic lymphocytic thyroiditis is found in approximately 10% of patients. APS1 was originally described in Europe. Recently, a report from USA described different clinical features and diagnostic criteria in APECED patients from Western Hemisphere i.e., as initial signs urticarial eruptions, intestinal dysfunction, enamel dysplasia. Classical triad presentation (mucocutaneous candidiasis, hypoparathyroidism and adrenal deficiency) was delayed. Life threatening endocrine complications can be prevented by an early diagnosis (96a). APS2, tends to occur later in childhood or in the adult with a polygenic predisposition. APS2 can be clustering in families with heterogeneous clinical phenotypes. Other disorders, including vitiligo, celiac disease, myasthenia gravis, premature ovarian failure and chronic active hepatitis may be present (95,95a). An extensive review of these associations has been published (96b) and large population data bases have clarified the strength of the various associations in adults (95k,96c).

Many other autoimmune conditions, organ or non-organ specific disease, can be associated with AITD in childhood and adolescence. Increased prevalence of CLT has been found in juvenile idiopathic arthritis (96d), non-segmental vitiligo (96e) and alopecia areata (96f). CLT may be associated with chronic uriticaria (96g) and rarely with immune-complex glomerulonephritis (96h). High prevalence of antinuclear antibodies (ANA) has recently been reported in a series of 93 children (86 with CLT and 8 with GD) without overt rheumatic disorders. In this series ANA positivity was found in 71% of children, ENA positivity in 4% and anti-DNA antibodies in 1% (96i). Growth hormone deficiency on an autoimmune basis has been suggested in a small number of children, of whom 43% have CLT (92j-96k). Nevertheless this is a rare association, hypothetically a growth disorder in a child with CLT can be due to other causes than hypothyroidism as celiac disease or GH deficiency. It is important that clinicians are cognizant of these associations in order to maintain a high index of vigilance.

Laboratory Evaluation

Measurement of TSH is the best initial screening test for the presence of primary hypothyroidism. If the TSH is elevated, then evaluation of the free T4 will distinguish whether the child has subclinical (normal free T4) or overt (low free T4) hypothyroidism. Measurement of TSH, on the other hand, is not helpful in central hypothyroidism. In these cases hypothyroidism is demonstrated by the presence of a low free T4 accompanied by an “inappropriate ‘’ TSH. In the past TRH testing (TRH 7 mcg/kg) was sometimes utilized to distinguish a hypothalamic versus pituitary origin of the hypothyroidism; in hypothalamic hypothyroidism there tends to be a delayed peak in TSH secretion (60-90 minutes versus the normal maximal response at 15-30 minutes) whereas in hypopituitarism there usually is little or no TSH response. However TRH is no longer available in the USA. Furthermore, the reliability of this test in the pediatric range has been questioned (97). Occasionally mild TSH elevation is seen in individuals with hypothalamic hypothyroidism, a consequence of the secretion of a TSH molecule with impaired bioactivity but normal immunoreactivity. Thyroid hormone resistance is characterized by elevated levels of T4 and T3 and an inappropriately normal or elevated TSH concentration. Antibodies to Tg and TPO, the thyroid antibodies measured in routine clinical practice, are detectable in over 90% of patients with chronic lymphocytic thyroiditis. Therefore, they are useful as markers of underlying autoimmune thyroid damage, TPO antibodies being more sensitive. TSH receptor antibodies also are found in a small proportion of patients with chronic lymphocytic thyroiditis. When stimulatory TSH receptor antibodies are present, they may give rise to a clinical picture of hyperthyroidism, the coexistence of chronic lymphocytic thyroiditis and Graves’ disease is known as Hashitoxicosis. In one study, TSH receptor blocking antibodies were found in <10% of children and adolescents with chronic lymphocytic thyroiditis, patients overall, but in 17.8% of those with severe hypothyroidism (defined as a serum TSH concentration >20 mU/L). Unlike in adults, they were found in goitrous as well as nongoitrous patients, and, when present at a high concentration, appeared to persist indefinitely, suggesting that the presence of potent TSH receptor blocking antibodies in adolescent females might identify patients at risk of having babies with transient congenital hypothyroidism in the future (97a,97b).

Imaging studies (thyroid ultrasonography and/or thyroid uptake and scan) may be performed if thyroid antibody tests are negative or if a nodule is palpable. If no goiter is present, imaging studies are helpful in identifying the presence and location of thyroid tissue, and therefore, of distinguishing primary myxedema from thyroid dysgenesis. Inborn errors of thyroid hormonogenesis beyond a trapping defect are usually suspected by an increased radioiodine uptake, and a large gland on scan. Other etiologies of hypothyroidism usually are evident on history. Ultrasound (US) is useful to define size, anatomy, echogenity of the thyroid. Occasionally the finding of heterogeneous echogenicity on ultrasound examination has been described prior to the appearance of antibodies. Diffuse reduction in echogenity, (hypoechoic), and pseudonodules are common findings (98). Moreover, US can be useful in detecting unsuspected thyroid nodules and cancer. In a study examinating US characteristic of the thyroid in 154 children and adolescents with goiter, nodules were reported in 20/154 (13%) and PTC in 4/154 (2.5%) of children. None of the papillary thyroid carcinoma in these series (PTC) was palpable, although a PTC diameter ranging from 1.2 to 2.6 cm was found. Interestingly, one case of PTC was first classified as pseudonodule (94c). The diffuse sclerosing variant of PTC, with a typical US appearance, has also been reported in children with CLT (94d). If there is a cost-effective benefit in performing US in all cases of children with CLT and/or goiter deserves sufficiently powered prospective studies.

Therapy

The typical replacement dose of L-thyroxine (derived from congenital hypothyroidism) in overt severe hypothyroidism is about 4 to 6 mcg/kg/day for children 1 to 5 years of age, 3 to 4 mcg/kg/day for these ages 6 to10 years and 2-3mcg/kg/day for these 11 ages and older. Lower dose as 1 to 3 mcg/kg/day may be sufficient in less severe cases. The dose should be individually titrated as the lowest useful to keep TSH in the normal range and FT4 or T4 in the upper half of the reference range. L-thyroxine should be given once daily preferably half to 1 hour before meal. Somministration of preparates (i.e., calcium, soya), or drugs that can interfere with absorption should be avoided. T4 and TSH should be measured after the child has received the recommended dosage for at least 6-8 weeks. Once a euthyroid state has been achieved, patients should be monitored every 6 to 12 months. In patients with a goiter a somewhat higher L-thyroxine dosage is used so as to keep the TSH in the low normal range, and thereby minimize its goitrogenic effect.

Close attention is paid to interval growth and bone age as well as to the maintenance of a euthyroid state. Thyroid hormone replacement is not associated with significant weight loss in overweight children, unless the hypothyroidism is severe (99b). Some children with severe, long standing hypothyroidism at diagnosis may not achieve their adult height potential even with optimal therapy (99c), emphasizing the importance of early diagnosis and treatment. Treatment is usually continued indefinitely.

Treatment of children and adolescents with subclinical hypothyroidism (normal free T4, elevated TSH) is controversial (100). Because normalization of TSH is also possible if the patient is not symptomatic, a reasonable option is to reassess thyroid function in 3- 6 months prior to initiating therapy because of the possibility that the thyroid abnormality will be transient.

In adults in whom the risk of progression to overt hypothyroidism is significant, treatment has been recommended whenever the serum TSH concentration is >10 mU/L ; if the TSH is 6-10 mU/L treatment on a case by case basis is suggested (100a). In children and adolescents, a recent five-years prospective study showed that in patients with CLT thyroid dysfunction increased from 27.3% to 47.4% (93c). Some considerations about “to treat or not to treat” “subclinical hypothyroidism” in children with a known cause of thyroid failure as CLT may be useful.

Well designed and adequately powered trials needed to establish the advantages of treating “subclinical hypothyroidism” are not available in adults and seem to be very difficult in children, also because some of the clinical consequences- i.e cardiovascular events- of untreated mild hypothyroidism may hypothetically occur later in adult life.

In adults, variations in thyroid function within the reference range may be associated with adverse health outcome (100b), and some data suggesting clinical consequences of subclinical hypothyroidism are also available in children and adolescents. A positive relationship was found for TSH levels and systolic and diastolic blood pressure (100c), atherogenic lipid profiles, (100d) and other risk factors for cardiovascular diseases (100e).

Moreover, adult patients with a thyroid nodule and TSH in the upper tertiles of the reference range may be at increased risk of malignancy (100f). Given an individual set point for TSH, more than an absolute TSH value (i.e more or less 6 7-, 8 UI/mL) the decision about treatment may consider the temporal trend of TSH in a patient. An increase of TSH value over the time suggests the progression of the grade of hypothyroidism. Both the decisions to “wait and see” or “to treat” require monitoring the thyroid function and clinical follow up. A careful discussion about “the state of the art” must be taken with the child and the family.

Guidelines about the management of subclinical hypothyroidism in pregnancy and in children have been published by the European Thyroid Association in 2014 (100g), but it as an evolving and open field (100).

Thyroid Dysgenesis and Inborn Errors of Thyroid Hormonogenesis

Occasionally, patients with thyroid dysgenesis will escape detection by newborn screening and present later in childhood with non goitrous hypothyroidism or with an enlarging mass at the base of the tongue or along the course of the thyroglossal duct. Similarly, children with inborn errors of thyroid hormonogesis may only be recognized later in childhood because of the detection of a goiter.

Drugs or Goitrogens

In addition to antithyroid medication, a number of drugs used in childhood may affect thyroid function, including certain anticonvulsants, lithium, amiodarone, aminosalicylic acid, aminoglutethimide and sertraline (101-101a). Similarly, a large number of naturally occurring goitrogens (broccoli, cabbage, sweet potatoes, cauliflower, soya beans, cassava and water pollutants) have been identified. Both radioiodine therapy and thyroidectomy, occasionally used in childhood for the definitive treatment of Graves’ disease, frequently cause permanent hypothyroidism.

Worldwide, iodine deficiency continues to be an important cause of hypothyroidism, affecting at least 800 million people living largely in developing countries. In addition, even in certain parts of Europe, an estimated 100-120 million individuals are thought to have borderline iodine deficiency (101b). Although one rarely sees iodine deficiency in North America, an iodine sufficient area, a 6 year old boy with goitrous hypothyroidism has been described in whom iodine deficiency was due to multiple food allergies and severe dietary restriction (101c). In addition, the child consumed a large intake of thiocyanate-containing foods that blocked organification of iodine.

Miscellaneous Causes of Acquired Hypothyroidism

Rarely, the thyroid gland may be involved in generalized infiltrative or infectious disease processes that are of sufficient severity to result in a disturbance in thyroid function (i.e., (Langerhans cell histiocytosis) (101 d). Alternatively, hypothyroidism may be a long term complication of mantle irradiation for Hodgkin’s disease or lymphoma. External irradiation of brain tumors in the posterior fossa of the brain may be associated with both primary and secondary hypothyroidism because of the inclusion of the neck in the radiation field. Rarely, hypothyroidism has been reported in infants with large hemangiomas (85b,85c). In these cases, the hypothyroidism was shown to be due to increased inactivation of T4 by the D3 activity of these tumors.

Central Hypothyroidism

Secondary or tertiary hypothyroidism in less severely affected children with the congenital abnormalities noted earlier in this chapter, may be recognized only later in childhood. Alternatively, secondary or tertiary hypothyroidism may develop as a result of acquired damage to the pituitary or hypothalamus, i.e., by tumors (particularly craniopharyngioma), granulomatous disease, head irradiation, infection (meningitis), surgery or trauma. Usually other trophic hormones are affected, particularly growth hormone.

Impaired Sensitivity to Thyroid Hormone (Thyroid Hormone Resistance)

In contrast to the neonatal period, children with thyroid hormone resistance usually come to attention when thyroid function tests are performed because of poor growth, hyperactivity, a learning disability or other nonspecific signs or symptoms. A small goiter may be appreciated. Affected patients have a high incidence of attention deficit hyperactivity disorder (102). Thyroid hormone resistance has also been described in patients with cystinosis (102a).

Other causes of goiter: Colloid or Simple (Nontoxic) Goiter

Colloid goiter is the second most common cause of euthyroid thyroid enlargement in childhood after CLT. The etiology of colloid goiter is unknown. Not infrequently there is a family history both of goiter, chronic lymphocytic thyroiditis and Graves’ disease, leading to the suggestion that colloid goiter, too, might be an autoimmune disease. Immunoglobulins that stimulated thyroid growth in vitro have been identified in a proportion of patients with simple goiter, but their etiological role is controversial (103). It is important to distinguish patients with colloid goiter from chronic lymphocytic thyroiditis because of the risk of developing hypothyroidism in patients with chronic lymphocytic thyroiditis, but not colloid goiter. Whereas many colloid goiters regress spontaneously, others appear to undergo periods of growth and regression, resulting ultimately in the large nodular thyroid glands later in life.

Clinical Manifestations and Laboratory Investigation

Evaluation of thyroid function by measurement of the serum TSH concentration is the initial approach to diagnosis. In euthyroid patients, the most common situation, chronic lymphocytic thyroiditis should be distinguished from colloid goiter. Clinical examination in both instances reveals a diffusely enlarged thyroid gland. Therefore, the distinction is dependent upon the presence of elevated titers of TPO and Tg antibodies in chronic lymphocytic thyroiditis but not colloid goiter. All patients with negative thyroid antibodies initially should have repeat examinations because some children with chronic lymphocytic thyroiditis will develop positive titers with time.

Therapy

Thyroid suppression in children with a euthyroid goiter is controversial (103a). A significant decrease in goiter size in patients with chronic lymphocytic thyroiditis as assessed by standard deviation score on ultrasonography has been demonstrated recently in patients treated for 3 years (103b). However, the absolute difference quantitatively was not reported and so, whether or not this difference was significant clinically remains unclear. Given the variability in response in different patients, it would be reasonable to attempt a therapeutic trial in patients whose goiter is large.

Painful thyroid: Acute suppurative thyroiditis, subacute thyroiditis

Painful thyroid enlargement is rare in pediatrics and suggests the probability of either acute (suppurative) (106) or subacute thyroiditis (106a). Rarely chronic lymphocytic thyroiditis may be associated with intermittent pain and be confused with the latter disorders. In acute suppurative thyroiditis, progression to abscess formation with the potential of rupture may occur rapidly so prompt recognition and antibiotic therapy are essential (106b). Acute suppurative thyroiditis is a potentially life-threatening endocrine emergency. It is often preceded by an upper respiratory infection, and can be initially misdiagnosed in a young child presenting with high fever, sore throat, and severe dysphagia. A tender very painful swelling in the region of the thyroid gland is present and the abscess can progress to the surrounding tissues and to the skin. Recurrent attacks and involvement of the left lobe suggest a pyriform sinus fistula between the oropharynx and the thyroid as the route of infection (106c). In the latter case, surgical extirpation of the pyriform sinus will frequently prevent further attacks. The management of this condition has recently been reviewed (106,106b). Subacute viral thyroiditis (or de Quervain or granulomatous thyroiditis) it is rarely reported in childhood and adolescence, but cases at 2-3 years of age are known (106a). Usually subacute thyroiditis presents with sore throat, fever and firm, painful tender enlargement of the thyroid. Mild signs of hyperthyroidism can be overlooked. Subacute thyroiditis may occur as a acute, subacute or rarely chronic disorder. A painless variant has been described also in children. (106d). Therapy is usually symptomatic, because the disease is self-limiting. Sometimes treatment with prednisone (0.5-1mg/kg/die) for a short period (i.e. one week) can be useful.

THYROTOXICOSIS AND HYPERTHYROIDISM

Thyrotoxicosis is defined as the clinical syndrome of hypermetabolism resulting from increased free thyroxine (T4) and/or free triiodothyronine (T3) serum levels (107)). The term thyrotoxicosis is not synonymous with hyperthyroidism, the elevation in thyroid hormone levels caused by an increase in their biosynthesis and secretion by the thyroid gland (Table 6). For example, thyrotoxicosis can result from the destruction of thyroid follicles and thyrocytes in the various forms of thyroiditis, or it can be caused by an excessive intake of exogenous thyroid hormone. It should also be noted that the elevation of free thyroid hormone levels does not always result in thyrotoxicosis in all tissues. In the syndrome of Resistance to Thyroid Hormone (RTH), dominant negative mutations in the thyroid hormone receptor β ( TR β) result in decreased thyroid hormone action in tissues where TRβ is the predominant receptor, for example in the liver and the pituitary, whereas other tissues such as the heart, which express mainly TR α, show signs of increased thyroid hormone action. The determination of the etiology of thyrotoxicosis is of importance in order to establish a rational therapy.

 

TABLE 6. CAUSES OF THYROTOXICOSIS IN CHILDHOOD AND ADOLESCENCE

 

THYROTOXICOSIS DUE TO HYPERTHYROIDISM (increased production of T3, T4)
Autoimmune hyperthyroidism
·       Graves’ disease
·       Hashitoxicosis
Congenital non autoimmune hyperthyroidism
·       Sporadic (de novo Persistent sporadic congenital non autoimmune hyperthyroidism (PSNAH)
·       Hereditary familial non-autoimmune autosomal dominant hyperthyroidism (FNAH)
Autonomous functioning nodules
·       Toxic adenoma
·       Hyperfunctioning papillary or follicular carcinoma
·       Toxic multinodular goiter
·       McCune Albright disease
TSH-induced hyperthyroidism
·       TSH-producing pituitary adenoma

·       Thyroid Hormone resistance

Tumors

·       Hydatiform mole, choriocarcinoma

·       Struma ovari, teratoma (autonomous function of thyroid tissue in ovarian)

 

THYROTOXICOSIS WITHOUT HYPERTHYROIDISM-
Transient thyrotoxicosis (Release of stored hormones)
·       Chronic lymphocytic thyroiditis
·       Subacute thyroiditis
·       Silent thyroiditis
·       Drug-induced thyroiditis
·       Exogenous causes
·       Thyroid hormone ingestion
·       Iodine -induced hyperthyroidism (iodine, radiocontrast agents, amiodarone)

Graves’ Disease

Autoimmune thyroid disease (AITD), including Chronic lymphocytic thyroiditis and Graves’ disease share immunological abnormalities, histological changes in the thyroid, and genetic predisposition and associated diseases. (See chronic lymphocytic thyroiditis section). The clinical spectrum of AITD ranges from hypothyroidism to hyperthyroidism.

More than 95% of cases of hyperthyroidism in children and adolescents are due to Graves’ disease, (107a) an autoimmune disorder characterized by hyperthyroidism, goiter and a particular opthalmopathy. TSH receptor antibodies that mimic the action of TSH (TRAb), causing increased thyroid hormonogenesis and growth are specific of Graves’ disease, but other autoantibodies, as AbTPO and AbTg, are detectable.

Incidence

Graves’ disease is rare in children and adolescents. However, incidence rates of thyrotoxicosis below 15 years of age are increased in the last years. A study from Denmark reported an incidence of 1.58/100.000 person-years in the period 1998-2012 versus 0.79/100.000 person-years in 1982-1988 (107b). There is a strong female predisposition, the female:male ratio being 6 to 8:1. Although it can occur at any age, it is most common in adolescence. Prepubertal children tend to have more severe disease, to require longer medical therapy and to achieve a lower rate of remission as compared with pubertal children (107c). This appears to be particularly true in children who present at <5 years of age (107d). Graves’ disease has been described in children with other autoimmune diseases, both endocrine and non endocrine. These include diabetes mellitus, Addison’s disease, vitiligo, systemic lupus erythematosis, rheumatoid arthritis, myasthenia gravis, periodic paralysis, idiopathic thrombocytopenia purpura and pernicious anemia. (See also associated diseases in CLT). There is an increased risk of Graves’ disease in children with Down syndrome (trisomy 21) (107e).

Pathogenesis

The cause of Graves’ disease is unclear. For unknown reasons the immune system produces TSH receptor antibodies that mimic the action of TSH. Binding of ligand results in stimulation of adenyl cyclase and thyroid hormonogenesis and growth (107f, 107g). Presumably a complex interaction between genetic susceptibility (i.e., HLA, CTLA4, PTN22 genes) and environmental factors contribute. A familial history of AITD is often present, as well as for other autoimmune diseases.

Unlike chronic lymphocytic thyroiditis in which thyrocyte damage is predominant, the major clinical manifestations of Graves’ disease are hyperthyroidism and goiter. As noted earlier, TSH receptor blocking antibodies, in contrast, inhibit TSH-induced stimulation of adenyl cyclase. Both stimulatory and blocking TSH receptor antibodies bind to the extracellular domain of the receptor and appear to recognize apparently discrete linear epitopes in the context of a three-dimensional structure (107g). A number of different monoclonal stimulating Abs including one derived from a patient with Graves’ disease have now been generated (107h) and the crystal structure of the human monoclonal stimulating TSH receptor Ab complexed with a portion of the TSH receptor ectodomain has been accomplished (107i). Taken together, a picture has emerged of distinct but overlapping binding sites of both stimulating and blocking TSH receptor Abs and of TSH to the leucine rich TSH receptor ectodomain (107j). Current evidence suggests that it is the shed A subunit rather than the intact, holoreceptor that induces TSH receptor Abs leading to hyperthyroidism (107j). Studies employing monoclonal TSH receptor antibodies cloned from patients and recombinant mutant TSH receptor have demonstrated that there exist multiple TSH receptor antibodies each with different specificities and functional activities. There is evidence that stimulatory antibodies are mostly lambda and of the IgG1 subclass, strongly suggesting that they are monoclonal or pauciclonal (107k). Blocking antibodies, on the other hand, are not similarly restricted.

Clinical Manifestations

The major clinical manifestations of Graves’ disease are hyperthyroidism and goiter. Opthalmopathy is usually mild, pretibial myxedema and acropachy are not described in children and adolescents.

The onset of Graves’ disease in often insidious and a delay in diagnosis of several months is common, especially in prepubertal children (107c). In children below 4 years of age, the prolonged hyperthyroidism can be dangerous to the CNS (107d). Shortened attention span, and emotional lability may lead to behavioral and school difficulties. Sleep disturbances, and nightmares can occur. Some patients complain of polyuria and of nocturia, the result of an increased glomerular filtration rate. All but a few children with Graves disease present with some degree of thyroid enlargement, and most have symptoms and signs of excessive thyroid activity, such as tremors, inability to fall asleep, weight loss despite an increased appetite, diarrhea, proximal muscle weakness, heat intolerance and tachycardia. Acceleration in linear growth may occur, often accompanied by advancement in skeletal maturation (bone age). Adult height is not affected. In the adolescent child, puberty may be delayed. If menarche has occurred, secondary amenorrhea is a common concomitant. If sleep is disturbed, the patient may complain of fatigue. Clinical findings are usually related to hyperthyroid state and disappear with restoration to the euthyroid state (108).

Graves’ opthalmopathy in children and adolescents is reported in up to half of the children and is usually less severe than in adults. Eyelid retraction and “stare” are common and linked to hyperthyroid state (108a). Proptosis is subtle and often overlooked. Normal references for children should be used (108b). Some cases of prominent progressive proptosis requiring treatment have been reported (108c). Surgical therapy is infrequently necessary. In a series of 35 children with Graves’ opthalmopathy from Mayo Clinic 3 patients (8.6%) underwent transantral orbital decomprenssion for proptosis that caused discomfort and exposure keratitis and 1 patient (2.9%) required eyelid surgery. No compressive optic neuropathy was found (108d).

Laboratory Evaluation

The clinical diagnosis of hyperthyroidism is confirmed by the finding of increased concentrations of circulating thyroid hormones (T4 or, preferably, free T4 and T3 or FT3) and low- undetectable TSH. In hyperthyroidism, the circulating T3 concentration frequently is elevated out of proportion to the T4 because, like TSH, TSH receptor antibodies stimulate increased T4 to T3 conversion. Some patients may have at diagnosis high FT3 and normal FT4, a condition known as T3 thyrotoxicosis (109). Children with T3 thyrotoxicosis seem to be younger, with higher levels of TRAb and larger goiter than classical GD. The timing of T3 thyrotoxicosis onset is variable and can require higher doses of ATD to control hyperthyroidism (109a). Demonstration of a suppressed TSH excludes much rarer causes of thyrotoxicosis, such as TSH-induced hyperthyroidism and thyroid hormone resistance in which the TSH is inappropriately “normal” or slightly elevated. If the latter diseases are suspected, free α-subunit should be measured. Alternatively, an elevated T4 level in association with an inappropriately “normal” TSH may be due to an excess of thyroxine-binding globulins (either familial or acquired, for example a result of oral contraceptive use) or rarer binding protein abnormalities (for example, familial dysalbuminemic hyperthyroxinemia) (109b). In the latter cases, serum TBG concentration or electrophoresis of T4 binding proteins, respectively, should be measured. If pregnancy or an hCG-secreting tumor are suspected, serum or urinary hCG concentration can be measured. A low serum Tg can be demonstrated if thyrotoxicosis factitia is suspected (109c).

The diagnosis of Graves’ disease is confirmed by the demonstration of TSH receptor antibodies (TRAb) in serum. TRAb are disease specific antibodies and have a pathogenetic role in Graves’ disease. TRAb are usually determined by binding assays. Bioassays that measure Thyroid Stimulating Immunoglobulins activity based on cAMP on cultured cells can be useful if TRAb are not detectable by binding assays (88a,88b,88c). About 95% of children with GD have TRAb detectable. There is a positive correlation between severity of Graves’ disease and TR antibodies level. Higher levels of TRAb and thyroid hormones at presentation are associated with a need of prolonged ATD treatment (109d). Measurement of TSH receptor antibodies may be useful in distinguishing the toxic phase of chronic lymphocytic thyroiditis (TSH receptor antibody negative) from Graves’ disease. Tg and TPO antibodies are positive in 70% of children and adolescents with Graves’ disease but their measurement is not as sensitive or specific as measurement of TSH receptor antibodies. In contrast to adults, radioactive iodine uptake and scan are used to confirm the diagnosis of Graves’ disease only in atypical cases: for example, if measurement of TSH receptor antibodies is negative, or if a functioning thyroid nodule is suspected.

Therapy

The care of children with Graves’ disease can be complicated and requires physicians with expertise in this area. Treatment guidelines developed for adults guidelines cannot be simply applied to children. For instance, TRAb may be detectable in serum for several years, making the terms “remission” and “recidive” inapplicable in 1-2 years periods for the majority of children and adolescents.

The choice of which of the three therapeutic options (medical therapy, surgery radioactive iodine, or radioactive iodine) to use, should be individualized and discussed with the patient and his/her family. Each approach has its advantages and disadvantages with respect to efficacy, both short and long term complications, the time required to control the hyperthyroidism, and the requirement for compliance. In general, medical therapy with methimazole (MMI) is the initial choice of most pediatricians although radioiodine is gaining increasing acceptance, particularly in noncompliant adolescents, in children who are developmentally delayed, and in those about to leave home (for example, to go to college). Concern about the potential long term induction of cancer by RAI given to children is the discussed later. Alternately, surgery, the oldest form of therapy, may be the initial choice in specific cases if an experienced thyroid surgeon is available. ATA guidelines for hyperthyroidism including a pediatric section have recently been released (88c).

Medical therapy

The thiouracil compounds PTU, MMI and carbimazole (converted to MMI) exert their antithyroid effect by inhibiting the organification of iodine and the coupling of iodotyrosine residues on the Tg molecule to T3 and T4.

The aim of therapy with antithyroid drugs is to control hyperthyroidism for a period sufficient to go to spontaneous remission or until the child is old enough to afford definitive therapy as surgery or RAI. Remission is defined as a state of biochemically euthyroidism or hypothyroidism for one year or more after discontinuation of ATD and occurs in a minority of cases (see later).

Some important considerations arose in the last years: MMI is the drug that should be used, unless special conditions, because of the inacceptable risk of liver failure and transplantation (FDA Propilthyuracil warning) in patients using PTU. PTU can cause fulminant hepatic necrosis and death. The risk was estimated to be 1:2000 in children (110,110a,110b). Propylthiouracil and methimazole have for years been considered effectively interchangeable, and liver damage was considered a very rare event. Recently a commission appointed by the FDA reevaluated this problem, and concluded that the rare but severe complications of liver failure needing transplantation, and death, were sufficient to contraindicate the use of PTU as the normal first-line drug (110c). PTU can be used only in pediatric patients who are allergic to MMI, for a short term, and in whom permanent forms of therapy are not possible. MMI should be used alone, titrating the dosage at the lowest useful to maintain euthyroidism. The “block.and replace therapy”, adding L-thyroxine to MMI should be avoided, because it requires a higher dose of MMI, and the majority of side effect of MMI are dose dependent.

The initial dosage of MMI is 0.5 mg/kg/day (up to 1mg/kg/die, maximal dose 30 mg/die) given every 12 hours. The plasma half-life of methimazole in children is only 3-6 hours, but the drug is concentrated in the thyroid and maintains higher levels there for up to 24 hours after a dose (110d). The initial dosage of PTU is 5 mg/kg/day given every 8 hours. In severe cases, a beta-adrenergic blocker (atenolol, 25 to 50 mg daily or twice daily) can be added to control the cardiovascular overactivity until a euthyroid state is obtained.

Before FDA warning for PTU MMI was generally preferred over PTU because for an equivalent dose it requires taking fewer tablets, it has a longer half-life (and so, requires less frequent medication) and because it has a more favorable safety profile. PTU use has also been advocated in the first trimester of pregnancy. PTU but not MMI inhibits the conversion of T4 to the more active isomer T3.

Patients treated with MMI should be followed every 4 to 6 weeks until the serum concentration of T4 (or free T4 and total T3) normalizes. It should be noted that the TSH concentration may not return to normal until several months later. Therefore, measurement of TSH is useful as a guide to therapy only after it has normalized but not initially. Once the T4 and T3 have normalized, one can decrease the dosage of thioamide drug by 30% to 50%. Maintenance doses of MMI may be administered once daily. PTU may be given twice daily. Usually patients can be followed every 1-4 months once thyroid function has normalized.

As suggested by the ATA guidelines (88c), before starting therapy with ATD, a baseline complete blood count, including WBC with differential, and a liver including bilirubin, transaminases and alkaline phosphatase can be useful. This is because hyperthyroidism itself can determine low WBC count, and premorbid liver disease (i.e autoimmune hepatitis reported in 1% of GD) can exist (110e). Baseline information may help in a correct interpretation of side effects of MMI.

In most children and adolescents, circulating thyroid hormone levels can be normalized readily with antithyroid medication as long as compliance is not a problem. The optimal duration of therapy is controversial. There is no doubt that most children and adolescents, particularly prepubertal ones, require a longer course of therapy than adults. Therefore treatment guidelines developed for older individuals should not be applied to the young. In one retrospective study, TSH receptor Abs disappeared from the circulation in <20% of patients after 13-24 months of medical therapy (110f) in contrast to adults in most of whom TSH receptor Abs normalize by 6 to 12 months (110g,110h, 110i). In another study, approximately 25% of children remitted with every 2 years of therapy up to 6 years of treatment (110j). Equivalent results have been obtained by others (107c). In a recent prospective trial of 154 children with newly diagnosed Graves’ disease treated with carbimazole, 20% of children remitted after 4 years of therapy, 37% after 6 years and 45% after 8 years (110k). The median duration of therapy in most studies is 3 to 4 years, but therapy should be individualized. In patients treated with antithyroid drugs alone, a low drug requirement, small goiter, and lack of orbitopathy are favorable indicators that drug therapy can be tapered gradually and withdrawn. Lower initial degree of hyperthyroxinemia (T4<20 mcg/dL (257.4 nmol/L); T3:T4 ratio <20), lower initial TSH receptor Ab concentration (>4X upper limit of normal (111e) and postpubertal age are favorable prognostic indicators. Persistence of TSH receptor antibodies, on the other hand, indicates a high likelihood of relapse. Initial studies suggesting that combined therapy (i.e., antithyroid drug plus L-thyroxine) might be associated with an improved rate of remission (110l) have not been confirmed (110m).

Side effects

Side effects of drugs were reported in 20-30% of children treated both with PTU and MMI and major side effect are thought to be due to PTU. Cumulative data from more than 500 children (111) with Graves’ disease reported mild increase of liver enzymes in 28%, mild leucopenia in 26% skin reactions in 9%, arthritis in 2.4%, nausea in 1.1%, agranulocytosis and hepatitis in 0.4%. Rare complications can be loss of taste, hypothrombinemia, thrombocytopenia, aplastic anemia, nephrotic syndrome and death (111). Side effects of MMI occur in up to 19% of children. Urticaria, arthralgias, gastrointestinal problems and neutropenia (<1500 granulocytes/mm3) are the most common, myalgias (3%), and cholestatic liver injury (1%) were also reported in a series of 100 children with Graves’ disease exclusively treated with MMI. Side effects usually occur in the first 6 months of therapy (111a) but can occur any time.

Major side effects as Stevens-Johnson syndrome and vasculitis occur rarely (111). Vasculitis can be related with the development of anti-neutrophil-cytoplasmic antibodies (ANCA). ANCA positivity has been reported with MMI and PTU therapy and may develop after many years of therapy (111b). Manifestations of vasculitis typically are polyarthritis and purpuric skin lesions. Pulmonary and renal involvement are also described. In severe cases, glucocorticoids or other immunosuppressive therapy may be needed. Guma et al reported ANCA positivity in 67% of patients with Graves’ disease before medical treatment, suggesting an association with Graves’ disease, rather than a complication of antithyroid drugs (111c).

Rarely, more severe sequelae such as hepatitis, a lupus like syndrome, thrombocytopenia, and agranulocytosis may occur. Most reactions are mild and do not contraindicate continued use. The risk of agranulocytosis (<500 granulocytes/mm3) appears to be greatest within the first 3 months of therapy but it can occur at any time. There is some evidence that close monitoring of the white blood cell count during this initial time period may be useful in identifying agranulocytosis prior to the development of a fever and infection (111d), but most authors do not consider the low risk to be worth the cost of close monitoring. It is important to caution all patients to stop their medication immediately and consult their physician should they develop unexplained fever, sore throat, or gingival sores or jaundice. Unlike PTU, MMI is rarely associated with hepatocellular injury.

Children treated with PTU and MMI tends to excessive weight gain during the first 6 months of therapy and nutrition consultation should be considered if needed (111e). Approximately 10% of children treated medically will develop long term hypothyroidism, a consequence of coincident cell and cytokine-mediated destruction.

Patients with Graves’ disease showed a higher risk of thyroid cancer (111). The Collaborative Thyrotoxicosis Study Group found the incidence of thyroid carcinomas over 10-20 years of follow up 5 fold higher in adults with Graves’ disease treated with thionamides than in patients treated with definitive therapy (111g). Long term stimulation of TSAb can play a role. Patients treated for years with thionamides should be carefully monitored for the detection of thyroid nodules.

Surgery

Surgery, the second therapeutic modality, is performed less frequently now than in the past. The main argument favoring surgery is that it may correct the thyrotoxicosis with surety and speed, and result in less disruption of normal life and development that is associated with long-term administration of antithyroid drugs and the attendant constant medical supervision.

The most important limiting factor is the availably of a high-volume thyroid surgeon to reduce potential complications (112,112a,112b). Near-total thyroidectomy is the procedure of choice in order to minimize the risk of recurrence. Surgery usually is reserved for patients who have failed medical management, who have a markedly enlarged thyroid, who refuse radioactive iodine therapy, and for the rare patient with significant ophthalmopathy in whom radioactive iodine therapy is contraindicated. Often adolescents are unable to maintain the careful dosage schedule needed for control of the disease .and can choice a definitive treatment. Surgical complication rates are higher in younger children (112c). The most common potential complication is transient hypocalcemia which occurs in approximately 10% of patients. Starting therapy with calcitriol 3 days before surgery (0.25 to 0.5 µg twice a day), can reduce the need for calcium infusion and the length of stay (112c). Other, less common potential complications are keloid formation (2.8%), recurrent laryngeal nerve paralysis (2%), hypoparathyroidism (2%) and, rarely (0.08%) death (111). There are fewer complications with an experienced surgeon and when modern methods of anesthesia and pain control are used (112). Prior to surgery, it is important to treat with antithyroid medication in order to render the child euthyroid and prevent thyroid storm. Iodides (Lugols solution, 5 to 10 drops tid or potassium iodide, 2 to 10 drops daily or Na ipodate, 0.5-1 gm every 3 days) are added for 7 to 14 days prior to surgery in order to decrease the vascularity of the gland. L-thyroxine replacement therapy should be given within days of surgery. Following surgical thyroid ablation most patients become hypothyroid and require lifelong thyroid replacement therapy. On the other hand, if therapy is inadequate, hyperthyroidism may recur. Therefore long-term follow-up is mandatory.

131-I Therapy

Definitive therapy with either radioactive iodine or surgical thyroid ablation is usually reserved for patients who have failed drug therapy, developed a toxic drug reaction, or are noncompliant. In recent years, however, radioactive iodine is being favored increasingly, even as the initial approach to therapy (111). The advantages are the relative ease of administration, the reduced need for medical follow up and the lack of demonstrable long term adverse effects (111). The aim of RAI is to ablate completely the thyroid gland and thereby reduce the risk of future neoplasia. RAI should be administrated in a single dose.

Although a dose of 50 to 200 ï­Ci of 131I/estimated gram of thyroid tissue has been used, the higher dosage is recommended, particularly in younger children, in order to completely ablate the thyroid gland and thereby reduce the risk of future neoplasia. The size of the thyroid gland is estimated, based on the assumption that the normal gland is 0.5-1.0 gms/year of age, maximum 15-20 gms. The formula used is: Estimated thyroid weight in grams X 50-200 mcCi 131 -I/fractional 131I 24 hour uptake Thyroid size can be assessed by ultrasound because underestimation and consequent insufficient RAI treatment is frequent. Surgery may be indicated for goiters larger than 80 gr. Radioactive iodine therapy should be used with caution in children <10 years of age and particularly in those <5 years of age because of the increased susceptibility of the thyroid gland in the young to the proliferative effects of ionizing radiation (113). Pretreatment with antithyroid drugs prior to RAI therapy is advisable if the hyperthyroidism is severe. Thyroid hormone concentrations may rise transiently 4 to 10 days after RAI administration due to the release of preformed hormone from the damaged gland. Beta blockers may be useful during this time period. Similarly, analgesics may be employed if there is mild discomfort due to radiation thyroiditis. Other acute complications of RAI therapy (nausea, significant neck swelling) are rare. One usually sees a therapeutic effect within 6 weeks to 3 months. Worsening of ophthalmopathy, described in adults after RAI, does not appear to be common in childhood. However, if significant ophthalmopathy is present RAI therapy should be used with caution and pretreatment with steroids may be effective. Alternately, another permanent treatment modality (surgery) should be considered.

The question of an age limit below which RAI should not be used frequently arises. With lengthening experience these limits have been lowered. Several studies with average follow-up periods of 12 – 15 years attest to the safety of 131-I therapy in adults (111g,113a,113b). In two studies treated persons showed no tendency to develop thyroid cancer, leukemia, or reproductive abnormalities, and their children had no increase in congenital defects or evidence of thyroid damage (113c,113d). Franklyn and co workers (113e) reported on a population based study of 7417 patients treated with 131-I for thyrotoxicosis in England. They found an overall decrease in incidence of cancer mortality, but a specific increase in mortality from cancer of the small bowel (7 fold) and of the thyroid (3.25) fold 9 (113e). The absolute risk remains very low, and it is not possible to determine whether the association is related to the basic disease, or to radioiodine treatment.

There are less data about long term effects of RAI therapy in pediatric Graves’ disease. In an early report, 73 children and adolescents were so treated. Hypothyroidism developed in 43. Subsequent growth and development were normal (113f). In another group of 23 children treated with 131-I, there were 4 recurrences, at least 5 became hypothyroid, and one was found to have a papillary thyroid cancer 20 months after the second dose (113g). Safa et al. (113a) reviewed 87 children treated over 24 years and found no adverse effects except the well-known occurrence of hypothyroidism. Hamburger (115c) has examined therapy in 262 children ages 3 – 18 and concluded 131-I therapy to be the best initial treatment. Read et al (113h) reviewed experience with 131-I over a 36 year period, including six children under age 6, and 11 between 6 and 11 years. No adverse effects on the patients or their offsprings were found, and they advocate 131-I as a safe and effective treatment. In a review including approximately 1000 children with Graves’ disease treated with RAI and followed for <5 to >20 years to date, (111) there does not appear to be any increased rate of congenital anomalies in offspring nor in thyroid cancer. However, long term follow up data in a larger cohort are still lacking. The epidemic of thyroid cancer apparently induced by radioactive iodine isotopes in infants and children living around Chernobyl suggests caution in use of 131-I in younger children.

Since the possibility of a general induction of cancer by 131-I is of central concern, it is interesting to calculate the risk in children using the data presented by Rivkees et al (113i) who are proponents of use of RAI for therapy in young children. The risk of death from any cancer due specifically to radiation exposure is noted by these authors to be 0.16%/rem for children, and the whole body radiation exposure from RAI treatment at age 10 to be 1.45 rem/mCi administered. Rivkees et al advise treatment with doses of RAI greater than 160 uCi/gram thyroid, to achieve a thyroidal radiation dose of at least 150Gy (about 15000 rads). Assuming a reasonable RAIU of 50% and gland size of 40 gm, the administered dose would thus be 40(gm) x 160uCi/gm x 2 (to account for 50% uptake) =12.8 mCi. Thus the long term cancer death risk would be 12.8 (mCi) x 1.45 rem (per mCi) x 0.16% (per rem) = 3%. For a dose of 15mCi the theoretical incremental risk of a later radiation-induced cancer mortality would be 4% at age 5, 2% at age 10, and 1% at age 15.

Whether or not accepting a specific 2-4% risk of death from any cancer because of this treatment is of course a matter of judgment by the physician and family. However, this would seem to many persons to constitute a significant risk that might be avoided. We note that this is a theoretical risk, based on known effects of ionizing radiation to induce malignancies, but not so far proven in this setting.

Long term studies focused to establish an increased risk of non-thyroid malignancies in children treated with RAI for Graves’ diseases would require about 10.000 children treated below 10 years of age, thus today the decision should be taken on an individual base with the patient and the family. The choice between surgery and RAI therapy in Graves’ disease in children is one of the major long standing controversies in pediatric endocrinology. Most physicians remain concerned about the risks of carcinogenesis, and the experience of Chernobyl has accentuated this concern. Others believe that the risks of surgery and problems with antithyroid drug administration outweigh the potential risk of 131-I therapy. This problem was critically reviewed by Rivkees et al (113j). They point out the significant risks of reaction to antithyroid drugs, and of surgery. Surgery may have a mortality rate in hospital in children of about one per thousand operations, although this may have decreased in recent years. Among problems with radioactive iodide therapy, they note the whole body radiation, possibly worsening of eye disease, and the apparent lack of significant thyroid cancer risk so far reported among children treated with I-131 for Graves’ disease. They assumed that risk would be lower in children after age five, and especially after age ten, and if all thyroid cells were destroyed. They advise using higher doses of radioiodine to minimize residual thyroid tissue, and avoiding treatment of children under age five, but they believe that RAI is a convenient, effective, and useful therapy in children with Graves’ disease. However, as noted above in the section on risks related to use od 131-I, Rivkees own data indicate that treatment of children with conventional doses of RAI may induce a lifetime risk of any fatal cancer of over 2%, a very serious consideration (113i). Many physicians remain reluctant to use 131-I in children under age 15-18 as a first line therapy. Following thyroid ablation most patients become hypothyroid and require lifelong thyroid replacement therapy. On the other hand, if therapy is inadequate, hyperthyroidism may recur. Therefore longterm follow-up is mandatory.

Other Causes of Hyperthyroidism

Non autoimmune hyperthyroidism

Non autoimmune hyperthyroidism is caused by constitutive activation of the TSH receptor (TSHR) (Table 6). Two clinical forms including “familial non-autoimmune autosomal dominant hyperthyroidism (FNAH)” and “persistent sporadic congenital non autoimmune hyperthyroidism (PSNAH)” are described. FNAH is characterized by autosomal dominant inheritance and high variable age of manifestation from neonatal period to 60 years. Variability is present also within the same family. Goiter is present in children, with nodules in older age. PSNAH includes forms with sporadic (de novo) germline mutations in the TSHR. Guidelines about this rare condition have recently been published (90e).

Hyperfunctioning nodules

Hyperthyroidism may be caused by a functioning thyroid adenoma, or functioning thyroid carcinoma. Hyperfunctioning nodules are a rare cause of overt or subclinical hyperthyroidism. Somatic activating mutations within the genes encoding the TSH receptor or the Gs-alpha subunit can be detected (90f). Scintigraphy with Tc 99 or I 123 show hypercaptating nodule and absence of uptake of the surrounding thyroid parenchima. Hyperthyroidism can be controlled with methimazole. Autonomous nodules can be single or a part of multinodular goiter. A recent retrospective study on 31 pediatric cases from US indicated that 45% were overt hyperthyroid at diagnosis and 42% presented with multinodular goiter. Mean age at diagnosis was 15 years, with a range 3-18 yrs. Mean size of the autonomous nodule was 39 mm. In this series of 31 patients, only one patient developed a follicular carcinoma in the controlateral lobe seven years after lobectomy for a benign adenomatoid nodule (114). However, the risk of cancer has been reported up to one third of patients in a series of 31 patients from an iodine- deficient area (114a).

ATA Guidelines for pediatric thyroid nodules and cancer indicate surgery as treatment of children with overt hyperthyroidism due to hyperfunctioning nodules, and surgery is indicated in any nodule >4 cm, because of the decreased sensitivity of FNA to detect malignancy (114b).

Hyperthyroidism may be seen as part of the McCune Albright syndrome (90f) (Table 6). McCune Albright syndrome is due to somatic mutations in Gsα gene that can occur in different tissues as, skin, bones thyroid, adrenal glands.

TSH induced hyperthyroidism

Hyperthyroidism may be due to the inappropriate secretion of TSH by a pituitary adenoma, but thyroid hormone resistance should be excluded.

The syndrome of “inappropriate secretion of TSH” was described in 1975 to indicate two forms of central hyperthyroidism, characterized by high levels of FT3 and FT4 and non suppressed TSH levels(114c). TSH secreting pituitary adenomas are extremely rare in pediatric patients. Guidelines from the ETA has been recently released for these tumors (114d). It is important to consider that a pituitary tumor can be a manifestation of Multiple endocrine neoplasia type 1 and rarely of familial forms of isolated pituitary adenomas with AIP mutations (114e).

In thyroid hormone resistance (RTH) due to mutations of the β isoform of the thyroid hormone receptor hyperthyrodism TSH driven can occur. (See chapter entitled Impaired sensitivity to thyroid hormone. Defects of transport, metabolism and action. .Alexandra M. Dumitrescu, MD and Samuel Refetoff, MD, in this book for a detailed description of this condition).

Tumors secreting chorionic gonadotropin

Recently an adolescent female was described in whom hyperthyroidism resulted from an hCG-secreting hydatidiform mole (114f). Chorioncarcinoma, metastatic embryonal carcinoma of the testis can cause hyperthyroidism (114g).

Transient thyrotoxicosis

Thyrotoxicosis is caused by damage of thyroid cells and release of thyroid hormones stored in the gland. The duration of toxic phase (usually one to three months) depends on the amounts of the thyroid hormones released and the rate of metabolic clearance. Thyroid cell breakdown causes abrupt onset and short duration of symptoms.

Principal causes of transient thyrotoxicosis include:

  • Autoimmune thyroiditis (silent thyroiditis): no local symptoms of local inflammation are present.
  • Subacute Viral thyroiditis (or de Quervain or granulomatous thyroiditis) it is rarely reported in children and adolescents (106b). Usually presents with sore throat, fever and firm, painful tender enlargement of the thyroid. Mild signs of hyperthyroidism can be overlooked.
  • Acute bacterial thyroiditis is rarely a cause of transient thyrotoxicosis.
  • Drug-induced thyroiditis (amiodarone and thyrosine kinase inhibitors)

 

THYROID NODULES AND CANCER

For exhaustive information see also chapters “Thyroid nodules” and “Thyroid cancer “ By F. Pacini and Leslie de Groot in this book.

Recently, the first guidelines specifically elaborated for children with thyroid nodules and differentiated thyroid cancer have been published (114b). Hereditary syndromes (i.e. PTEN related sydromes, DICER1 syndrome, Carney complex, Familial adenomatous polyposis) associated with thyroid cancer in childhood are also been detailed (114b). Medullary thyroid carcinoma guidelines have also been revised, including genetic counseling and modified risk class for children with hereditary MTC (115a).

Thyroid nodules are rare in the first 2 decades of life, but when found, they are more likely to be carcinomatous than are similar masses in adults (115b). Follicular adenomas and colloid cysts account for the majority of benign nodules. Other causes of nodular enlargement include chronic lymphocytic thyroiditis and embryological defects, such as intrathyroidal thyroglossal duct cysts or unilateral thyroid agenesis. Like in adults, the most common form of thyroid cancer in childhood and adolescence is papillary thyroid carcinoma, but other histological types found in the adult may also occur (115c).

A high index of suspicion is necessary if the nodule is painless, of firm or hard consistency, if it is fixed to surrounding tissues or if there is a family history of thyroid cancer. Other worrisome findings include a history of rapid increase in size, associated cervical adenopathy, hoarseness or dysphagia. Even the findings of a cystic component or a functioning nodule, commonly used as favorable signs in adult patients, do not exclude the possibility of neoplasia (115c). Occasionally, thyroid cancer presents in childhood as unexplained cervical adenopathy, or neoplasia is found in patients who also have chronic lymphocytic thyroiditis (115c). The possibility of a rare medullary thyroid carcinoma should be considered if there is a family history of thyroid cancer or pheochromocytoma or if the child has multiple mucosal neuromas and a marfanoid habitus, findings suggestive of multiple endocrine neoplasia (MEN) types 2A and/or 2B (115d).

Children exposed previously to thyroid irradiation comprise a high-risk group. The increased risk of thyroid cancer in adults exposed during childhood to low levels of thyroid irradiation for benign conditions of the head and neck is well known (115e). The increased incidence of both benign and carcinomatous nodules in patients with Hodgkin disease who had received radiotherapy to the neck during childhood is also being documented increasingly (115f, 115g). Thyroid cancer is now known to be the most common second malignancy in childhood survivors of Hodgkin’s and is also seen with increased frequency in leukemia survivors (115h). Similarly, children exposed to high levels of radioactive iodine in the first decade of life or in utero, a consequence of the Chernobyl disaster, are at a markedly increased risk of developing papillary thyroid cancer (113). The risk of thyroid cancer is related to the dose of external irradiation and, unlike the 19 year average latency after low dose irradiation, the average latent period in survivors of Hodgkin disease appears to be only 9 years (115g). In Chernobyl victims, the latency was only 4 years (113). As compared with adults, there appears to be a higher prevalence of gene rearrangements in children with differentiated thyroid cancer, the clinical significance of which is unclear (115h).

Initial investigation of a thyroid nodule includes evaluation of thyroid function and TPO and Tg antibodies. A suppressed serum TSH concentration accompanied by an elevation in the circulating T4 and/or T3 suggests the possibility of a functioning nodule, which can be confirmed with a radionuclide scan. The finding of positive antibodies, on the other hand, usually indicates the presence of underlying chronic lymphocytic thyroiditis, but in some cases, positive antibodies may simply constitute evidence of an immune response to the presence of neoplastic cells. Ultrasonography provides information about whether the nodule is solid or cystic, and whether it is single or multifocal. Fine-needle aspiration biopsy, popular in the investigation of thyroid carcinoma in adults, is gaining increasing acceptance and is now considered to be the procedure of choice in the evaluation of nodules >0.5 cm (115k).

There is an increased incidence of both cervical node involvement and of pulmonary metastases at the time of diagnosis in children with thyroid carcinoma (115c). Nonetheless, the long term cancer specific mortality rate is no greater in children than in adults <40 years of age (115i). Guidelines specifically elaborated for management of children with thyroid nodules differentiated thyroid cancer have been published (114b). Excision of the tumor or lobe is the appropriate treatment for benign tumors and cysts, whereas total thyroidectomy with preservation of the parathyroid glands and recurrent laryngeal nerves is the initial therapy for malignant thyroid tumors. The latter procedure is followed by radioablation if there is evidence of residual gland or tumor after surgery. The issue of prophylactic lymph node dissection is controversial (115h). After radioiodine therapy, the dose of thyroxine is adjusted to keep the serum TSH concentration suppressed (between 0.05 mU/L and 0.1 mU/L in a sensitive assay). Measurement of serum Tg, a thyroid follicular cell-specific protein, is used to detect evidence of metastatic disease in differentiated forms of thyroid cancer, such as papillary or follicular carcinoma. This is best performed after a period (usually 6 weeks) of thyroxine withdrawal or after the exogenous administration of recombinant TSH (115). Measurement of circulating calcitonin is used as a tumor marker for medullary thyroid cancer (MTC), a C-cell derived malignancy (115m). Mutations of the RET protooncogene, detectable in nearly all familial forms of MTC, is of value in screening family members (115e, 115m). In families affected with multiple endocrine neoplasia type 2, screening of children as young as 5 years followed by total thyroidectomy has been successful in curing patients with microscopic MTC, an otherwise highly malignant neoplasm with a poor prognosis (115e). See Medullary Thyroid Carcinoma guidelines for updated genetic counseling and modified risk class for children with hereditary MTC (115a).

Optimal monitoring of patients with a history of thyroid irradiation during childhood remains controversial. Because of the insensitivity of clinical palpation, regular assessment of thyroid function (TSH and, as necessary free T4) as well as ultrasound examinations should be performed. There is evidence that thyroid suppression is associated with a reduction in the development of new nodules after partial surgical resection of an irradiated thyroid gland (115q) but whether it plays any role if the TSH is not elevated or in preventing neoplasia is unknown. Recently, a study that followed a cohort of 4338 5- years survivors of pediatric solid cancer suggested that chemotherapy (nitrosureas), splenectomy, and radiation dose to pituitary gland also play a role in predicting thyroid cancer risk (115r).

A retrospective study on the effects of total body irradiation (TBI) preceding hemopoietic cell transplation in childhood suggested short term and life-long monitoring for thyroid nodules and thyroid cancer in these patients (115s). Although it was a small size, retrospective study they found the time from TBI to thyroid carcinoma detection ranged from 2.2 years to 15.3 years. Follow up programs are advised for long term survivors of childhood cancer.

 

 

REFERENCES

  1. Missero C, Cobellis G, De Felice M, Di Lauro R. Molecular events involved in differentiation of thyroid follicular cells. Mol Cell Endocrinol. 1998;140:37-43.
  2. De Felice M, Di Lauro R. Thyroid development and its disorders: genetics and molecular mechanisms. Endocr Rev. 2004;25:722-46.
  3. Manley NR, Capecchi MR. The role of Hoxa-3 in mouse thymus and thyroid development. Development. 1995;121;1989-2003.
  4. Fagman H, Grande M, Edsbagge J, Semb H, Nilsson M. Expression of classical cadherins in thyroid development: maintenance of an epithelial phenotype throughout organogenesis. Endocrinology. 2003;144:3618-24.
  5. Fagman H, Liao J, Westerlund J, Andersson L, Morrow BE, Nilsson M. The 22q11 deletion syndrome candidate gene Tbx1 determines thyroid size and positioning. Hum Mol Genet. 2007;16:276-85.
  6. Fagman H, Grande M, Gritli-Linde A, Nilsson M. Genetic deletion of sonic hedgehog causes hemiagenesis and ectopic development of the thyroid in mouse. Am J Pathol. 2004;164:1865-72.
  7. Burrow GN, Fisher DA, Larsen PR. Maternal and fetal thyroid function. N Engl J Med. 1994;331:1072-8.
  8. Fisher DA, Klein AH. Thyroid development and disorders of thyroid function in the newborn. N Engl J Med. 1981;304:702-12.
  9. Thorpe-Beeston JG, Nicolaides KH, McGregor AM. Fetal thyroid function. Thyroid.1992;2:207-17.
  10. Williams FL, Simpson J, Delahunty C, et al. Developmental trends in cord and postpartum serum thyroid hormones in preterm infants. J Clin Endocrinol Metab. 2004;89:5314-20.
  11. Brown RS, Shalhoub V, Coulter S, et al. Developmental regulation of thyrotropin receptor gene expression in the fetal and neonatal rat thyroid: relation to thyroid morphology and to thyroid-specific gene expression. Endocrinology. 2000;141:340-5.
  12. Hume R, Simpson J, Delahunty C, et al. Human fetal and cord serum thyroid hormones: developmental trends and interrelationships. J Clin Endocrinol Metab. 2004;89:4097-103.
  13. De Nayer P, Cornette C, Vanderschueren M, et al. Serum thyroglobulin levels in preterm neonates. Clin Endocrinol. 1984;21:149-53.
  14. Sobrero G, Munoz L, Bazzara L, et al. Thyroglobulin reference values in a pediatric infant population. Thyroid. 2007;17:1049-54.
  15. Roti E, Gnudi A, Braverman LE. The placental transport, synthesis and metabolism of hormones and drugs which affect thyroid function. Endocr Rev.1983;4:131-49.
  16. Kester MH, Kaptein E, Van Dijk CH, et al. Characterization of iodothyronine sulfatase activities in human and rat liver and placenta. Endocrinology. 2002;143:814-9.
  17. Van der Geyten S, Segers I, Gereben B, et al. Transcriptional regulation of iodothyronine deiodinases during embryonic development. Mol Cell Endocrinol. 2001;183:1-9.
  18. Ruiz de Ona C, Obregon MJ, Escobar del Rey F, Morreale de Escobar G. Developmental changes in rat brain 5′-deiodinase and thyroid hormones during the fetal period: the effects of fetal hypothyroidism and maternal thyroid hormones. Pediatr Res. 1988;24:588-94.
  19. Morreale de Escobar G, Obregon MJ, Escobar del Rey F. Is neuropsychological development related to maternal hypothyroidism or to maternal hypothyroxinemia? J Clin Endocrinol Metab. 2000;85:3975-87.
  20. Ferreiro B, Bernal J, Goodyer CG, Branchard CL. Estimation of nuclear thyroid hormone receptor saturation in human fetal brain and lung during early gestation. J Clin Endocrinol Metab. 1988;67:853-6.

20a. Chan SY, Vasilopoulou E, Kilby MD. The role of the placenta in thyroid hormone delivery to the fetus. Nat Clin Pract Endocrinol Metab. 2009; 5:45-54.

20b. Patel J.Li H, Mortimer RH, Richard K. Delivery of maternal thyroid hormones to the fetus. Trends Endocrinol Metab. 2011;22:164-70.

20c. Burns R, O’Herlihy C, Smyth PP. The Placenta as a Compensatory Iodine Storage Organ. Thyroid. 2011; 21: 541-46.

20d. Burns R, O’Herlihy C, Smyth PP. Regulation of iodide uptake in placental primary cultures. Eur Thyroid J. 2013;2:243-51.

20e. Li H, Patel J, Mortimer RH, Richard K. Ontogenic changes in human placental sodium iodide symporter expression. Placenta. 2012;33:946-8.

  1. Vulsma T, Gons MH, de Vijlder JJ. Maternal-fetal transfer of thyroxine in congenital hypothyroidism due to a total organification defect or thyroid agenesis. N Engl J Med. 1989;321:13-6.
  2. Calvo RM, Jauniaux E, Gulbis B, et al. Fetal tissues are exposed to biologically relevant free thyroxine concentrations during early phases of development. J Clin Endocrinol Metab. 2002;87:1768-77.

22a. Contempre B, Jauniaux E, Calvo R, Jurkovic D, Campbell S, de Escobar GM. Detection of thyroid hormones in human embryonic cavities during the first trimester of pregnancy. J Clin Endocrinol Metab.1993; 77:1719-22.

22b. Bernal J. Thyroid hormone receptors in brain development and function. Nat Clin Practice. 2007; 3:249-259.

22c. Rovet JF. The role of thyroid hormones for brain development and cognitive function. Endocr Dev. 2014;26:26-43.

22d. Morreale de Escobar G, Obregon MJ, Escobar del Rey F: Maternal thyroid hormones early in pregnancy and fetal brain development. In: Best Practice & Research in Clinical Endocrinology and Metabolism: The Thyroid and Pregnancy. Editor: Glinoer D 2004; 18:225.

22e. Williams GR. Neurodevelopmental and neurophysiological actions of thyroid hormone. Neuroendocrinol. 2008;20:784-94.

22f. Schroeder AC, Privalsky ML. Thyroid hormones, T3 and T4, in the brain. Front Endocrinol. 2014;5:40. doi: 10.3389/fendo.2014.00040. eCollection 2014.

22g. Bernal J, Guadaño-Ferraz A, Morte B. Thyroid hormone transporters-functions and clinical implications. Nat Rev Endocrinol. 2015;11:406-17.

  1. Meinhold H, Dudenhausen JW, Wenzel KW, Saling E. Amniotic fluid concentrations of 3,3′,5′-tri-iodothyronine (reverse T3), 3,3′-di-iodothyronine, 3,5,3′-tri-iodothyronine (T3) and thyroxine (T4) in normal and complicated pregnancy. Clin Endocrinol.1979;10:355-65.

23a. Seror R, Amand G, Guibourdenche J, Ceccaldi PF, Luton D, Anti-TPO antibodies diffusion through the placental barrier during pregnancy. PloS One. 2014, 9:e84647.

23b. Zimmermann MB. The effects of iodine deficiency in pregnancy and infancy. Paediatr Perinat Epidemiol. 2012; 26 (Supp1):108-117.

  1. Boyages SC. Clinical review 49: Iodine deficiency disorders. J Clin Endocrinol Metab. 1993;77:587-91.
  2. Rovet J, Walker W, Bliss B, Buchanan L, Ehrlich R. Long-term sequelae of hearing impairment in congenital hypothyroidism. J Pediatr. 1996;128:776-83.
  3. de Zegher F, Pernasetti F, Vanhole C, Devlieger H, Van den Berghe G, Martial JA. The prenatal role of thyroid hormone evidenced by fetomaternal Pit-1 deficiency. J Clin Endocrinol Metab. 1995;80:3127-30.
  4. Matsuura N, Konishi J. Transient hypothyroidism in infants born to mothers with chronic thyroiditis–a nationwide study of twenty-three cases. The Transient Hypothyroidism Study Group. Endocrinol Jpn. 1990;37:369-79.

27a. Hallowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, Braveman LE. Serum T4 and thyroid antibodies in the United States population (1988-1994): National Health and Nutrition Examination Survay (NHANES III). J Clin Endocrinol Metab. 2002; 489-499.

  1. Man EB, Jones WS, Holden RH, Mellits ED. Thyroid function in human pregnancy. 8. Retardation of progeny aged 7 years; relationships to maternal age and maternal thyroid function. Am J Obstet Gynecol. 1971;111:905-16.
  2. Haddow JE, Palomaki GE, Allan WC, Williams JR, Knight GJ, Gagnon J, O’Heir CE, Mitchell ML, Hermos RJ, Waisbren SE, Faix JD, Klein RZ. Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child. N Engl J Med.1999;341:549-55.

29a. Smit BJ, Kok JH, Vulsma T, Briët JM, Boer K, Wiersinga WM. Neurologic development of the newborn and young child in relation to maternal thyroid function. Acta Paediatr. 2000;89:291-5.

29b. Li Y, Shan Z, Teng W, Yu X, Li Y, Fan C, Teng X, Guo R, Wang H, Li J, Chen Y, Wang W, Chawinga M, Zhang L, Yang L, Zhao Y, Hua T. Abnormalities of maternal thyroid function during pregnancy affect neuropsychological development of their children at 25-30 months. Clin Endocrinol. 2010;72:825-9.

  1. Pop VJ, Kuijpens JL, van Baar AL, Verkerk G, van Son MM, de Vijlder JJ, Vulsma T, Wiersinga WM, Drexhage HA, Vader HL. Low maternal free thyroxine concentrations during early pregnancy are associated with impaired psychomotor development in infancy. Clin Endocrinol. 1999;50:149-55.

30a. Pop VJ, Brouwers EP, Vader H, Vulsma T, van Baar AL, de Vijlder JJ. Maternal hypothyroxinaemia during early pregnancy and subsequent child development: a 3-year follow-up study. Clin Endocrinol. 2003;59:282-8.

30b. Henrichs J, Bongers-Schokking JJ, Schenk JJ, Ghassabian A, Schmidt HG, Visser TJ, Hooijkaas H, de Muinck Keizer-Schrama SM, Hofman A, Jaddoe VV, Visser W, Steegers EA, Verhulst FC, de Rijke YB, Tiemeier H. Maternal thyroid function during early pregnancy and cognitive functioning in early childhood: the generation R study. J Clin Endocrinol Metab. 2010;95:4227-34.

30c. Suárez-Rodríguez M, Azcona-San Julián C, Alzina de Aguilar V. Hypothyroxinemia during pregnancy: the effect on neurodevelopment in the child. Int J Dev Neurosci. 2012;30:435-8.

30d. Finken MJ, van Eijsden M, Loomans EM, Vrijkotte TG, Rotteveel J. Maternal hypothyroxinemia in early pregnancy predicts reduced performance in reaction time tests in 5- to 6-year-old offspring. J Clin Endocrinol Metab. 2013;98:1417-26.

30e. Julvez J, Alvarez-Pedrerol M, Rebagliato M, Murcia M, Forns J, Garcia-Esteban R, Lertxundi N, Espada M, Tardón A, Riaño Galán I, Sunyer J. Thyroxine levels during pregnancy in healthy women and early child neurodevelopment. Epidemiology. 2013;24:150-7.

30f. Vermiglio F, Lo Presti VP, Moleti M, Sidoti M, Tortorella G,Scaffidi G, Castagna MG, Mattina F, Violi MA, Crisà A, Artemisia A, Trimarchi F. Attention deficit and hyperactivity disorders in the offspring of mothers exposed to mild-moderate iodine deficiency:a possible novel iodine deficiency disorder in developed countries. J Clin Endocrinol Metab. 2004;89:6054-60.

30g. Modesto T, Tiemeier H, Peeters RP, Jaddoe VW, HofmanA, Verhulst FC, Ghassabian A. Maternal Mild Thyroid Hormone Insufficiency in Early Pregnancy and Attention-Deficit/Hyperactivity Disorder Symptoms in Children. JAMA Pediatr. 2015;169:838-45.

30h. Román GC, Ghassabian A, Bongers-Schokking JJ, Jaddoe VW,Hofman A, de Rijke YB, Verhulst FC, Tiemeier H. Association of gestational maternal hypothyroxinemia and increased autism risk. Ann Neurol. 2013;74:733-42.

30k. Gyllenberg D, Sourander A, Surcel HM, Hinkka-Yli-SalomäkiS, McKeague IW, Brown AS. Hypothyroxinemia During Gestation and Offspring Schizophrenia in a National Birth Cohort. Biol Psychiatry. 2015 19. pii: S0006-3223(15)00520-X.

30i. Ghassabian A, Bongers-Schokking JJ, de Rijke YB, van Mil N, Jaddoe VW, de Muinck Keizer-Schrama SM, Hooijkaas H, Hofman A, Visser W, Roman GC, Visser TJ, Verhulst FC, Tiemeier H. Maternal thyroid autoimmunity during pregnancy and the risk of attention deficit/hyperactivity problems in children: the Generation R Study.Thyroid. 2012; 22:178-86.

30j. Pop VJ, de Vries E, van Baar AL, Waelkens JJ, de Rooy HA, Horsten M, Donkers MM, Komproe IH, van Son MM,Vader HL. Maternal thyroid peroxidase antibodies during pregnancy: a marker of impaired child development? J Clin Endocrinol Metab. 1995;80:3561-6.

30l. Wasserman EE, Pillion JP, Duggan A, Nelson K, Rohde C, Seaberg EC, Talor MV, Yolken RH, Rose NR. Childhood IQ, hearing loss, and maternal thyroid autoimmunity in the Baltimore Collaborative Perinatal Project. Pediatr Res. 2012;72:525-30.

30m. Kooistra L, Crawford S, van Baar AL, Brouwers EP, Pop VJ. Neonatal effects of maternal hypothyroxinemia during early pregnancy. Pediatrics. 2006;117:161-7.

30n. Mirabella G, Westall CA, Asztalos E, Perlman K, Koren G, Rovet J. Development of contrast sensitivity in infants with prenatal and neonatal thyroid hormone insufficiencies. Pediatr Res. 2005;57:902-7.

30o. Ghassabian A, Bongers-Schokking JJ, Henrichs J, Jaddoe VW, Visser TJ, Visser W, de Muinck Keizer-Schrama SM, Hooijkaas H, Steegers EA, Hofman A, Verhulst FC, van der Ende J, de Rijke YB, Tiemeier H. Maternal thyroid function during pregnancy and behavioral problems in the offspring: the generation R study. Pediatr Res. 2011;69:454-9.

  1. Momotani N, Iwama S, Momotani K. Neurodevelopment in children born to hypothyroid mothers restored to normal thyroxine (T₄) concentration by late pregnancy in Japan: no apparent influence of maternal T₄ deficiency. J Clin Endocrinol Metab. 2012;97:1104-8.

31a. Liu H, Momotani N, Noh JY, Ishikawa N, Takebe K, Ito K. Maternal hypothyroidism during early pregnancy and intellectual development of the progeny. Arch Intern Med. 1994;154:785-7.

31b. Downing S, Halpern L, Carswell J, Brown RS Severe maternal hypothyroidism corrected prior to the third trimester is associated with normal cognitive outcome in the offspring. Thyroid. 2012; 22:625-30.

  1. Lazarus JH, Bestwick JP, Channon S et al. Antenatal thyroid screening and childhood cognitive function. N Engl J Med. 2012; 366:493-501.

32a. Casey B. Effect of treatment of maternal subclinical hypothyroidism or hypothyroxinemia on IQ in offspring. American Journal of Obstetrics & Gynecology. Supplement to JANUARY 2016 Abstr 2 S2.

32b. Su PY, Huang K, Hao JH, Xu YQ, Yan SQ, Li T, Xu YH, Tao FB. Maternal thyroid function in the first twenty weeks of pregnancy and subsequent fetal and infant development: a prospective population-based cohort study in China. J Clin Endocrinol Metab. 2011;96:3234-41.

32c. Korevaar TI, Muetzel R, Medici M, Chaker L, Jaddoe VW, de Rijke YB, Steegers EA, Visser TJ, White T, Tiemeier H, Peeters RP. Association of maternal thyroid function during early pregnancy with offspring IQ and brain morphology in childhood: a population-based prospective cohort study. Lancet Diabetes Endocrinol. 2016; 4:35-43.

32d. Samadi A, Skocic J, Rovet JF. Children born to women treated for hypothyroidism during pregnancy show abnormal corpus callosum development.Thyroid. 2015; 25:494-502.

32e. Lischinsky JE, Skocic J, Clairman H, Rovet J. Preliminary Findings Show Maternal Hypothyroidism May Contribute to Abnormal Cortical Morphology in Offspring. Front Endocrino . 2016;25:7-16.

32f. Williams FL, Watson J, Ogston SA, Visser TJ, Hume R, Willatts P. Maternal and umbilical cord levels of T4, FT4, TSH, TPOAb, and TgAb in term infants and neurodevelopmental outcome at 5.5 years. J Clin Endocrinol Metab. 2013;98: 829-38.

  1. Klein RZ, Haddow JE, Faix JD, et al. Prevalence of thyroid deficiency in pregnant women. Clin Endocrinol. 1991;35:41-6.

33a. Thangaratinam S, Tan A, Knox E, Kiliby MD, Franklyn J, Coomarasamy A. Association between thyroid autoantibodies and miscarriage and preterm birth: meta-analysis of evidence. BMJ. 2011; 342:d2616.

33b. Alexander EK, Pearce EN, Brent GA, Brown RS, Chen H, Dosiou C, grobman W, Laurberg P, Lazarus JK, Mandel SJ, Peeters R, Sullivan S. 2016 guidelines of the American Thyroid Association for the diagnosis and management of thyroid disease during pregnancy and postpartum. Thyroid 2017, Jan 6. doi: 10.1089/thy.2016.0457. [Epub ahead of print]

  1. Abuid J, Stinson DA, Larsen PR. Serum triiodothyronine and thyroxine in the neonate and the acute increases in these hormones following delivery. J Clin Invest..1973;52:1195-9.
  2. Bianco AC, Silva JE. Intracellular conversion of thyroxine to triiodothyronine is required for the optimal thermogenic function of brown adipose tissue. J Clin Invest. 1987;79:295-300.
  3. Houstek J, Vizek K, Pavelka S, et al. Type II iodothyronine 5 ′ -deiodinase and uncoupling protein in brown adipose tissue of human newborns. J Clin Endocrinol Metab. 1993;77:382-7.
  4. Mercado M, Yu VY, Francis I, Szymonowicz W, Gold H. Thyroid function in very preterm infants. Early Hum Dev. 1988;16:131-41.
  5. Nelson JC, Weiss RM, Wilcox RB. Underestimates of serum free thyroxine (T4) concentrations by free T4 immunoassays. J Clin Endocrinol Metab. 1994;79:76-9.
  6. Ares S, Escobar-Morreale HF, Quero J, et al. Neonatal hypothyroxinemia: effects of iodine intake and premature birth. J Clin Endocrinol Metab. 1997;82:1704-12.
  7. Frank JE, Faix JE, Hermos RJ, et al. Thyroid function in very low birth weight infants: effects on neonatal hypothyroidism screening. J Pediatr. 1996;128:548-54.
  8. Kok JH, Tegelaers WH, de Vijlder JJ. Serum thyroglobulin levels in preterm infants with and without the respiratory distress syndrome. I. Cord blood study. Pediatr Res. 1986;20:996-1000.
  9. Thorpe-Beeston JG, Nicolaides KH, Snijders RJ, Felton CV, McGregor AM. Thyroid function in small for gestational age fetuses. Obstet Gynecol. 1991;77:701-6.
  10. Zurakowski D, Di Canzio J, Majzoub JA. Pediatric reference intervals for serum thyroxine, triiodothyronine, thyrotropin, and free thyroxine. Clin Chem. 1999;45:1087-91.
  11. Fisher D. Next generation newborn screening for congenital hypothyroidism? J Clin Endocrinol Metab. 2005;90:3797-9.

44b. Marvin L. Mitchell, Ho-Wen Hsu, Inderneel Sahai and the Massachusetts Pediatric Endocrine Work Group. The increased incidence of congenital hypothyroidism: fact or fancy? Clin Endocrinol. 2011;75:806-10.

  1. Klein AH, Meltzer S, Kenny FM. Improved prognosis in congenital hypothyroidism treated before age three months. J Pediatr. 1972;81:912-5.
  2. Dussault JH. The anedoctal history of screening for congenital hypothyroidism. J Clin Endocrinol Metab. 1999;84:4332-4.
  3. Delange F. Neonatal screening for congenital hypothyroidism: results and perspectives. Horm Res.1997;48:51-61.
  4. Rovet J, Daneman D. Congenital hypothyroidism: a review of current diagnostic and treatment practices in relation to neuropsychologic outcome. Paediatr Drugs. 2003;5:141-9.

50a. Ford G, LaFranchi SH. Screening for congenital hypothyroidism: a worldwide view of strategies. Best Practice & Research Clinical Endocrinology & Metabolism. 2014;28:175-187.

50b. Wassner AJ, Brown RS. Congenital Hypothyroidism: recent advances. Curr Opin Endocrinol Diabetes Obes. 2015, 22:407-412.

50c. Olivieri A, Corbetta C, Weber G, Vigone MC, Fazzini C,Medda E, and The Italian Study Group for Congenital Hypothyroidism. Congenital hypothyroidism due to defects of thyroid development and mild increase of TSH at screening: data from the Italian registry of infants with congenital hypothyroidism. J Clin Endocrinol Metab. 2013; 98:140:3-1408.

50d. Harris KB and Pass KA. Increase of congenital hypothyroidism in New York State and in The United States. Molecular Genetics and Metabolism. 2007; 91:268-277.

50e. Hinton CF, Harris KB, Borgfeld L et al. trends in incidence of congenital hypothyroidism related to select demographic factors: data from the United States , California, Massachussets, New York and Texas. Pediatrics. 2010;125:S37-S47.

50f. Olivieri A, Fazzini C, Medda E. Multiple factors influencing the incidence of congenital hypothyroidism detected by neonatal screening. Horm Res Pediatr. 2015 83:86-93.

50g. Barry Y, Bonaldi C, Goulet V, Coutant R, Leger J, Paty AC, Delmas D, Cheillan D, Roussey M. Increased incidence of congenital hypotiroidism in France from 1982 to 2012: a nationwide multicenter analysis. Ann Epidemiol. 2016;26:100-105.

  1. LaFranchi SH, Hanna CE, Krainz PL, Skeels MR, Miyahira RS, Sesser DE. Screening for congenital hypothyroidism with specimen collection at two time periods: results of the Northwest Regional Screening Program. Pediatrics. 1985;76:734-40.

51a. Ford GA, Denniston S, Sessere D, Skeels MR, LaFranchi SH. Transient versus permanent congenital hypothyroidism after the age of 3 years in infants, detected on the first versus second newborn screening test in Oregon, USA. Horm Res Paediatr. 201616;86(3):169-177

51b. Leger J, Olivieri A, Donaldson M, Torresani T,Krude H, van Vliet G, Polak M, Butler G on behalf of ESPE-PES-SLEP-JSPE-APEG-APPES-ISPAE, and the Congenital Hypothyroidism Consensus Conference Group. European Society for Pediatric Endocrinology Consensus Guidelines on Screening, Diagnosis, and management of congenital hypothyroidism. Horm Res Pediatr. 2014;81:80-103.

51c. Leger J, Olivieri A, Donaldson M, Torresani T, Krude H, van Vliet G, Polak M, Butler G on behalf of ESPE-PES-SLEP-JSPE-APEG-APPE-ISPAE, and the Congenital Hypothyroidism Consensus Conference Group. European Society for Pediatric Endocrinology Consensus Guidelines on Screening, Diagnosis, and management of congenital hypothyroidism. J Clin Endocrinol Metab. 2014; 99:363-384.

  1. Lanting CI, van Tijn DA, Loeber JG, Vulsma T, de Vijlder JJ, Verkerk PH. Clinical effectiveness and cost-effectiveness of the use of the thyroxine/thyroxine-binding globulin ratio to detect congenital hypothyroidism of thyroidal and central origin in a neonatal screening program. Pediatrics. 2005;116:168-73.
  2. van Tijn DA, de Vijlder JJ, Verbeeten B, Jr., Verkerk PH, Vulsma T. Neonatal detection of congenital hypothyroidism of central origin. J Clin Endocrinol Metab. 2005; 90:3350-9.

53a. Nesbesio TD, McKenna MP, Nabhan ZM et al. Newborn screening results in children with central hypothyroidism. J Pediatr. 2010;156:990-3.

53b. Adachi M, Soneda A, Asakura Y, Muroya K, Yamagami Y, Hirahara F. Mass screening of newborns for congenital hypothyroidism of central origin by free thyroxine measurement of blood samples on filter paper. Eur J Endocrinol. 2012; 166:829-838.

53c. Soneda, A, Adachi M, Muroya K, Asakura Y, Yamagami Y, Hirahara F. Overall usefulness of newborn screening for congenital hypothyroidism by using free thyroxine measurements. Endocr J 2014; 61:025-1-30.

  1. American Academy of Pediatrics AAP Section on Endocrinology and Committee on Genetics, and American Thyroid Association Committee on Public Health: Newborn screening for congenital hypothyroidism: recommended guidelines. Pediatrics. 1993;91:1203-9.

54a. Peters C, Brooke I, Ifederu A, Langham S, Hindmarsh P, Cole TJ. Defining the newborn blood spot screening reference interval for TSH: impact of ethnicity. J Clin Endocrinol Metab. 2016; 101:3445-3449.

  1. 55. Fisher DA. Effectiveness of newborn screening programs for congenital hypothyroidism: prevalence of missed cases. Pediatr Clin North Am. 1987;34:881-90.
  2. Adams LM, Emery JR, Clark SJ, Carlton EI, Nelson JC. Reference ranges for newer thyroid function tests in premature infants. J Pediatr. 1995;126:122-7.

56a. Vigone MC, Caiulo S, Di Frenna M et al. Evolution of thyroid function in preterm infants detected by screening for congenital hypothyroidism. J Pediatrics. 2014;164: 1296-1302.

56b. LaFranchi SH. Screening preterm infants for congenital hypothyroidism: better the second time around. J Pediatr. 2014; 164:1259-1261.

56c. Zoller RT, Rovet J. Timing of thyroid hormone action in the developing brain: clinical observation and experimental findings. J Neuroendocrinol. 2004:16:809-818.

56d. Spencer CA, LoPresti JS, Patel A et al. Application of a new chemiluminometric thyrotropin assay to subnormal measurement. J Clin Endocrinol Metab. 1990;70:453-460.

56e. Parazzini C, Baldoli C, Scotti G, Triulzi F. Terminal zones of myelination: MR evaluation of children aged 20-40 months. Am J Neuroradiol. 2002:23:1669-1673.

56f. Gagne N, Parma J, Deal C, Vassart G, Van Vliet G. Apparent congenital athyreosis contrasting with normal plasma thyroglobulin levels and associated with inactivating mutations in the thyrotropin receptor gene: are athyreosis and ectopic thyroid distinct entities? J Clin Endocrinol Metab. 1998;83:1771-5.

  1. Smith DW, Klein AM, Henderson JR, Myrianthopoulos NC. Congenital hypothyroidism–signs and symptoms in the newborn period. J Pediatr. 1975;87(6 Pt 1):958-62.

57a. Karakoc-Aydiner E, Turan S, Akpinar I, Dede F, Isguven P, Adal E, Guran T, Akcay T, Bereket A. Pitfalls in the diagnosis of thyroid disgenesis by thyroid untrasonography and scintigraphy. Eur J Endocrinol. 2012; 166:43 -48.

  1. Brown RS, LaFranchi S, Rose SR. Patient information page from the hormone foundation. Congenital hypothyroidism. J Clin Endocrinol Metab. 2009;94:1835-6.

58a. Carswell JM, Gordon JH, Popovsky E, Hale A, Brown RS. Generic and brand-name L-thyroxine are not bioequivalent for children with severe congenital hypothyroidism. J Clin Endocrinol Metab. 2013;98: 610-617.

  1. Bongers-Schokking JJ, de Muinck Keizer-Schrama SM. Influence of timing and dose of thyroid hormone replacement on mental, psychomotor, and behavioral development in children with congenital hypothyroidism. J Pediatr. 2005;147:768-74.
  2. Heyerdahl S, Oerbeck B. Congenital hypothyroidism: developmental outcome in relation to levothyroxine treatment variables. Thyroid. 2003;13:1029-38.
  3. Simoneau-Roy J, Marti S, Deal C, Huot C, Robaey P, Van Vliet G. Cognition and behavior at school entry in children with congenital hypothyroidism treated early with high-dose levothyroxine. J Pediatr. 2004;144:747-52.
  4. Rovet JF. In search of the optimal therapy for congenital hypothyroidism. J Pediatr. 2004;144:698-700.
  5. Dimitropoulos A, Molinari L, Etter K, et al. Children with congenital hypothyroidism: long-term intellectual outcome after early high-dose treatment. Pediatr Res. 2009;65:242-8.
  6. Fisher DA. The importance of early management in optimizing IQ in infants with congenital hypothyroidism. J Pediatr. 2000;136:273-4.

64a. Leger J. Congenital hypothyroidism: a clinical update of long-term outcome in young adults. Eur J Endocrinol. 2015;172, R67-R77.

64b. Leger J, Ecosse E, Roussey M, Lanoe JL, Larroque B. French Congenital hypothyroidism study group. Subtle health impairment and socioeducational attainment in young adult patients with congenital hypothyroidism diagnosed by neonatal screening: a longitudinal population-based cohort study. J Clin Endocrinol Metab. 2011;96:1771-82.

64c. Lichtenberer-Geslin L, Dos Santos S, Hassani Y, Ecosse E, Van Den Abbele T, Leger J. Factors associated with hearing impairment in patient with congenital hypothyroidism treated since the neonatal period: a national population-based study. J Clin Endocrinol Metab. 2013; 98:3644-3652.

64d. Leger J, dos Santos S, larroque B, Ecosse E. Pregnancy outcomes and relationship to treatment adequacy in women treated early for congenital hypothyroidism: a longitudinal population-based study. J Clin Endocrinol Metab. 2015; 100: 800-809.

64e. Lof C, Patyra K, Kuulasmaa T, Vangipurapu J, Undeutsch H, Jaeschke H, Pajunen T, Kero A, Krude H, Biebermann, Kleinau G, Kuhnen P, Rantakari K, Miettinen P, Kirjavainen T, Pursiheimo JP, Mustila T, Jaaskelainen J, Ojaniemi M, Toppari J, Ignatius J, Laakso M, Kero J. Detection of novel gene variants associated with congenital hypothyroidism in a finnish patient cohort. Thyroid. 2016; 26:1215-1224.

 

64f. Nicholas AK, Serra EG, Cangui H, Alyaarubi S, Ullah I, Schoenmakes E, Deeb A, Habeb AM, AI Maghamsi M, Peters C, Nathwani N, Aycan Z, Saglam H, Bober E, Dattani M, Shenoy S, Murray PG, Babiker A, Willensen R, Thankarmony A, Lyons G, Irwin R, Padidela R, Tharian K, Davies JH, Puthi V, Park SM, Massoud AF, Gregory JW, Albanese A, Pease-Gevers E, Martin H, Brugger K, Maher ER, Chatterje K, Anderson CA, Schoenmakers N. Comprehensive screening of eight known causative genes in congenital hypothyroidism with gland-in-situ. J Clin Endocrinol Metab. 2016; 12:4521-31.

  1. Siebner R, Merlob P, Kaiserman I, Sack J. Congenital anomalies concomitant with persistent primary congenital hypothyroidism. Am J Med Genet. 1992;44:57-60.
  2. Kumar J, Gordillo R, Kaskel FJ, Druschel CM, Woroniecki RP. Increased prevalence of renal and urinary tract anomalies in children with congenital hypothyroidism. J Pediatr. 2009;154:263-6.
  3. Perry R, Heinrichs C, Bourdoux P, et al. Discordance of monozygotic twins for thyroid dysgenesis: implications for screening and for molecular pathophysiology. J Clin Endocrinol Metab. 2002;87:4072-7.

67a. Brown RS, Demmer LA. The etiology of thyroid dysgenesis-still an enigma after all these years. J Clin Endocrinol Metab. 2002;87:4069-71.

  1. Fort P, Lifshitz F, Bellisario R, et al. Abnormalities of thyroid function in infants with Down syndrome. J Pediatr. 1984;104:545-9.

68a. Stoupa A, Kariyawasam D, Carre’ A, Polak M. Update of thyroid development genes. Endocrinol Metab Clin N Am. 45:243-246, 2016.

68b. Szinnai G. genetics of normal and abnormal thyroid development in humans. Best Pract Res Clin Endocrinol Metab. 2014; 28:133-50.

68c. Satoshi Narumi and Tobonobu Hasegawa. TSH resistance revised. Endocrine Journal. 2015, 62:393-398.

68d. Calebiro D, Gelmini G, Cordella D et al. Frequent TSH receptor genetic alterations with variable signaling impairment in a large series of children with nonautoimmune isolated hyperthyrotropinemia. J Clin Endocrinol Metab. 2012; 97: E156-160.

 

68e. Carre A, Szinnai G, Castanet M et al. Five new TTF1/NKX2-1 mutations in brain–lung-thyroid syndrome: rescue by PAX8 synergism in one case. Hum Mol Genet. 2009;18:2266-76.

 

68f. de Filippis T, Marelli F, Vigone MC, Di Frenna M, Weber G, Persani L. Novel NKX2-1 frameshift mutations in patients with athypical phenotypes of the brain-lung-thyroid syndrome. Eu Thyroid J. 2014; 3: 227-233.

68g. Thowarth A, Schittert-Hubener S , Schrumpf P et al. Comprehensive genotyping and clinical characterization reveal 27 novel NKX2-1 mutations and expand the phenotypic spectrum. J Med Genet. 2014;51:375-387.

68h. Stenson PD, Mort M, Ball EV et al. The human gene mutation database: building a comprehensive mutation repository for clinical and molecular genetics, diagnostic testing and personalized genomic medicine. Hum Genet. 2014;133: 1-9.

68i. Narumi S, Muroya K, Asakura Y et al. Transcription factor mutations and congenital hypothyroidism: systematic genetic screening of a population-based cohort of Japanese patients. J Clin Endocrinol Metab. 2010: 95: 1981-5.

68j. Fernandez LP, Lopez- Marquez A, Santisteban P. Thyroid transcription factors in development, differentiation and disease. Nat Rev Endocrinol. 2015; 11:29-42.

68k. Fernandez LP, Lopez-Marquez A, Martinez AM, Gomez-Lopez G, Santisteban P l. New insights into FOXE1 functions: identification of direct FOXE1 targets in thyroid cells. PLoS One. 2013, 8:e62849.

68m. Carre A Hamza RT, Kariyawasam D, et al. A novel FOXE1 mutation (R73S) in Bamforth-Lazarus syndrome causing increased thyroid gene expression. Thyroid. 2014:24:649-65.

68n. Dentice M, Cordeddu V, Rosica A et al. Missense mutation in the transcription factor NKX2-5: a novel molecular event in the pathogenesis of thyroid dysgenesis. J Clin Endocrinol Metab. 2006;91:1428-1433.

 

68o. Opitz R, Hitz MP, Vandernoot I et al. Functional zebrafish studies based on human genotyping point to netrin-1 as a link between aberrant cardiovascular development and thyroid dysgenesis. Endocrinology. 2015;156:377-388.

68q. Porazzi P, Marelli F, Benato F,de Filippis T,Calebiro D, Argenton F, Tiso N, Persani L . Disruptors of global and JAGGED1 mediated notch signaling affect thyroid morfogenesis in the zebrafish. Endocrinology. 2012; 153:5645-5658.

68r. De Filippis T, Marelli F. Nebbia G, Porazzi P, Corbetta S, Fugazzola L, Gastaldi R, Vigone MC, Biffanti R, Frizziero D, Mandara’ L, Prontera P, Salerno M, Maghnie M, Tiso N, Radetti G, Weber G, Persani L. JAG1 loss- of- function variations as a novel predisposing event in the pathogenesis of congenital thyroid defects. J Clin Endocrinol Metab. 2016;101:861-870.

68s. Knobel M, Madeires-Neto G. An outline of inherited disorders of the thyroid hormone generating system. Thyroid. 2003; 13:771-801.

  1. Portulano C, Paroder-Belenitsky M, Carrasco N. The Na+/I symporter (NIS): mechanism and medical impact. Endocr Rev. 2014;35 :106-149.
  2. Everett LA, Glaser B, Beck JC, Idol JR, Buchs A, Heyman M, Adawi F, Hazani F, Nassir E, Baxevanis AD, Sheffied VC, Green ED. Pendred syndrome is caused by mutations in putative sulphate transporter gene (PDS). Nat Genet. 1997;17:411-422.
  3. 71. Kopp P. Pendred’s syndrome: identification of the genetic defect a century after its recognition. Thyroid. 1999;9:65-9.

71a. Ladsous M, Vlaeminck-Guillem V, Dumur V, Vincent C, Dubrulle F, Dhanens CM, Wemeau JL. Analysis of the thyroid phenotype in 42 patients with Pendred syndrome and non syndromic enlargement of the vestibular aqueduct. Thyroid. 2014; 24:639-648.

71b. Bizhanova A, Kopp P. Genetics and phenomics of Pendred syndrome. Mol Cell Endocrinol. 2010;322:83-90.

  1. Nascimiento AC, Guendes DR, Santos CS, Knobel M, Rubio IG, Medeiros-Neto G. Thyroperoxidase gene mutations in congenital goitrous hypothyroidism with total and partial iodide organification defect. Thyroid. 2003;13:1145-51.

 

72a. Delodoey J, Pfarr N, Vuissoz JM, Parma J, Vassart G, Biesterfeld S, Phlenz J, Van Vliet G. Pseudodominant inherence of goitrous congenital hypothyroidism caused byTPO mutations; molecular and in silico studies. J Clin Endocrinol Metab. 2008;93:627-633.

 

72b. Moreno JC,Bikker H, Kempers MJE, vanTrotsenburg AS, Baas F, deVijlder JJM, Vulsma T, Ris-Stalpers C. Inactivating mutations in the gene for thyroid oxidase 2 (THOX2) and congenital hypothyroidism. N Engl J Med. 2002; 347:95-102.

 

72c. Muzza M, Rabbiosi S, Vigone MC, Zamproni I, Cirelio V, Maffini M, Maruca K, Schoenmarkers N, Beccaria L, Gallo F, Park SM, Beck-Peccoz P, Persani L, Weber G, Fugazzola L. The clinical and molecular characterization of patients with dyshormonogenetic congenital hypothyroidism reveals specific diagnostic clues for DUOX2 defects. J Clin Endocrinol Metab. 2014;99:E544-E553.

 

72d. Zamproni I, Grasberger H, Cortinovis F, Vigone MC, Chiumello G, Mora S, Onigata K, Fugazzola L, Refetoff S, Persani L, Weber G. Biallelic inactivation of the dual oxidation maturation factor 2 (DUAXA2) gene as a novel cause of congenital hypothyroidism. J Clin Endocrinol Metab. 2008; 93:605-610.

 

72e. Ieri T, Cochaux P, Targovinik HM, Suzuki M, Shimoda S, Perret J, Vassart G. A 3’splice site mutation in the thyroglobulin gene responsible for congenital goiter with hypothyroidism. J Clin Invest..1991:88:1901-1905.

 

72f. Medeiros-Neto G, Kim PS, Yoo SE, Vono J, Targovnik H, Camargo R, Hossain SA, Arvan P. Congenital hypothyroid goiter with deficient thyroglobulin. Identification of an endoplasmic reticulum storage disease with induction of molecular chaperones. J Clin Invest. 1996; 98:2838-2844.

 

72g. Citterio CE, Rossetti LC, Souchon PF, Morales C, Thouvard-Viprey M, Salmon-Musial AS, Mauran PL, Doco-Fenzy M, Gonzales-Sarmiento R, Rivolta CM, De Brasi CD, Targovink HM. Novel mutational mechanism in the thyroglobulin gene: imperfect DNA inversion as a cause for hereditary hypothyroidism. Mol Cell Endocrinol. 2013; 381:220-229.

 

72h. Iglesias A, Garcia-Nimo L, Cocho de Juan JA, Moreno JC. Towards the pre-clinical diagnosis of hypothyroidism caused by iodotyrosine deiodinase (DEHAL1) deficts. Best Prac Res Clin Endocrinol Metab. 2014; 28:151-159.

 

72i. Moreno JC, Klootwijk W, van Toor H, Pinto G, D’Allessandro M, Leger A, Goundie D, Polak M, Gruters A, Visser TJ. Mutations in the iodotyrosine deiodinase gene and hypothyroidism. N Eng J Med. 2008;358:1856-1859.

  1. Fisher DA, Dussault JH, Foley TP, Klein AH, LaFranchi S, Larsen PR, Mitchell ML, Murphey WH, Walfish PG. Screening for congenital hypothyroidism: results of screening one million North American infants. J Pediatr. 1979;94: 700-705.

73a. Lanting CI, van Tijn DA, Loeber JG, Vulsma T, de Vijder JJ, Verkerk PH. Clinical effectiveness and cost-effectiveness of the use of the thyroxine-binding globulin ratio to detect congenital hypothyroidism of thyroidal and central origin in a neonatal screening program. Pediatrics. 2005;116:168-173.

73b. Kempers MJ, Lanting CI, van Heijst AF, van Trotsenburg AS, Wiedijk BM. De Vijder JJ,Vulsma T. Neonatal screening for congenital hypothyroidism based on thyroxine, thyrotropin and thyroxine-binding globulin measurement potentials and pitfalls. J Clin Endocrinol Metab.2006; 91:3370-3376.

73c. Zwaveling-Soonawala N, van Trotsenburg AS, Verkerk PH. The severity of congenital hypothyroidism of central origin should not be underestimated. J Clin Endocrinol Metab. 2015:100:E297-E300.

73d. Collu R, Tang J, Castagne’ J, Legace’ G, Masson N, Huot C, Deal C, Delvin E, Faccenda F, Edne KA, Van Vliet G. A novel mechanism for isolated central hypothyroidism: Inactivating mutations in the thyrotropin releasing-hormone receptor gene. J Clin Endocrinol Metab. 1997:82:1561-1565.

73e. Bonomi M, Busnelli M, Beck-Peccoz P, Costanzo D, Antonica F, Dolci C, Pilotta A, Buzi F, Persani L. A family with complete resistance to thyrotropin-releasing hormone receptor gene. N Engl J Med. 2009;360:731-734.

 

73f. Koulouri O, Nicholas AK, Shoenmakers E, Mokrosinki J, Lane F, Cole T, Kirk J, Farooqi IS, Chatterjee VK, Gurnell M, Shoenmakers N. A novel thyrotropin-releasing hormone receptor missense mutation (P81R) in central congenital hypothyroidism. J Clin Endocrinol Metab. 2016; 101:847-851.

 

  1. Dacou-Voutekatis C, Feltquate DM, Drakopoulou M, Kourides IA, Dracopoli NC. Familial hypothyroidism caused by a nonsense mutation in the thyroid stimulating hormone beta-subunit gene. Am J Hum Genet. 1990;46:988-993.

 

74a. Bonomi M, Proverbio MC, Weber G, Chiumello G, Beck-Peccoz P, Persani L. Hyperplastic pituitary gland, high serum glycoprotein hormone alfa-subunit, and variable circulating thyrotropin (TSH) levels as a hallmark of central hypothyroidism due to mutations of the TSH beta gene. J Clin Endocrinol Metab. 2001;86:1600-1604.

 

74b. Bequadano MS, Ciaccio M, Dujovne N et al. Two novel mutations of the TSH- β-subunit gene underlying congenital central hypothyroidism undetectable in neonatal TSH screening. J Clin Endocrinol Metab. 2010;95:E98-E103.

 

74c. Drees JC, Stone JA, Reamer CR, Arboleda VF, Huang K, Hrynkow J, Greene DN, Petrie MS, Hoke C, Lorey TS et al. Falsely undetectable TSH in a cohort of Sauth Asian euthyroid patients. J Clin Endocrinol Metab. 2014;99:1171-1179.

 

74d. PappaT, Johannesen J, Schemberg N, Torrent M, Dumitriescu A, Refetoff S. A TSHB variant with impaired immunoreactivity but intact biological activity and its clinical implications. Thyroid. 2015; 25:869-876.

 

74e. Hermanns P, Couch R, Leonard N, Klotz C, Pohlenz J. A novel deletion in the thyrotropin β-subunit gene identified by array comparative genomyc hybridization analysis causes central congenital hypothyroidism in a boy originating from Turkey. Horm ResPediatr. 2014;82:201-205.

 

74f. Nicholas AK, Jaleel S, Kyons E, Shoenmakers E, Dattani MT, Crowne E, Bernhard B, Kirk J, Roche EF, Chatterjee VK, Shoenmakers N. Molecular spectrum of TSHβ subunit gene defects in central hypothyroidism in the UK and Ireland. Clin Endocrinol. 2016; Jun 30 doi:10111/cen. 13149.

  1. Sun Y, Bak B, Schoenmaker N, van Trotsenburg AS, Oostdijk W, Voshol P et al. Loss-of-function mutations in IGSF1 cause an X-linked syndrome of central hypothyroidism and testicular enlargement. Nat Genet. 2012; 44:1375-1381.

75a. Tajima T, Nakamura A, Ishizu K. A novel mutation of IGSF1 in a Japanese patient of congenital central hypothyroidism without macroorchidism. Endocr J. 2013; 60:245-249.

75b. Nakamura A, Bak B, Silander TL, Lam J, Hotsubo T, Yorifuji T, Ishizu K, Bernard DJ, Tajima T. Three novel IGSF1 mutations in four Japanese patients with X-linked congenital central hypothyroidism. J Clin Endocrinol Metab. 2013;98:E1682-E1691.

75c. Joustra SD, Schoenmakers N, Persani L, Campi I, Bonomi M, Radetti G, Beck-Peccoz P, Zhu H, Davis TM, Sun Y, Corssmit EP, Appelman-Dijkstra NM, Heinen CA, Pereira AM, Varewick AJ, Janssen JA, Endert E, Hennekam RC, Lombardi MP, Mannens MM, Bak B, Bernard DI, Breuning MH, Chatterijee K et al. The IGSF1 deficiency syndrome:characteristics of male and females patients. J Clin Endocrinol Metab. 2013; 98:4942-4952.

75d. Joustra SD, Heinen CA, Schoenmakers N, Bonomi M, Ballieux MO, Turgeon D, Bernard E, Fliers E, van Trotsenburg ASP, Losekoot M, Persani L, Wit JM, Biermasz NR, Pereira AM, Oostdijk W, on behalf of the IGSF1 Clinical Care Group. IGSF1 Deficiency: lessons from an extensive case series and recommendations for clinical management. J Clin Endocrinol Metab. 2016; 101:1627-1636.

75e. Tenenbaum-Rakover Y, Turgeon MO, London S,Hermanns P,Pohlenz J, Bernard DJ, Bercovich D. Familial central hypothyroidism caused by a novel IGSF1 gene mutation. Thyroid. 2016 ;26:1693-1700..

  1. Alatzoglou KS, Dattani MT. Genetic forms of hypopituitarism and their manifestations in neonatal period. Early Hum Dev. 2009;85:705-712.

76a. Brown RS, Bhatia V, Hayes E. An apparent cluster of congenital hypopituitarism in central Massachusetts: magnetic resonance imaging and hormonal studies. J Clin Endocrinol Metab. 1991;72:12-8.

76b. Dattani ML, Martinez-Barbera J, Thomas PQ, et al. Molecular genetics of septo-optic dysplasia. Horm Res. 2000; 53 Suppl 1:26-33.

76c. Machinis K, Pantel J, Netchine I, et al. Syndromic short stature in patients with a germline mutation in the LIM homeobox LHX4. Am J Hum Genet. 2001;69:961-8.

76d. Parks JS, Brown MR. Transcription factors regulating pituitary development. Growth Horm IGF Res. 1999;9 Suppl B:2-8; discussion -11.

  1. Refetoff S, Bassett JH, Beck-Peccoz P et al. Classification and proposed nomenclature for inherited defects of thyroid hormone action, cell transport and metabolism. Thyroid. 2014; 24:407.

77a. Refetoff S, Weiss RE, Usala SJ. The syndromes of resistance to thyroid hormone. Endocr Rev. 1993; 14:348-399.

77b. Weiss RE, Balzano S, Scherberg NH, Refetoff S. Neonatal detection of generalized resistance to thyroid hormone. Jama. 1990;264:2245-50.

77c. Bochukova E, Schoenmakers N, Agostini E, et al. A mutation in the thyroid hormone receptor alpha gene. N Engl J Med. 2012;366:243-9.

77d. Moran C, Chatterje K, Resistance to thyroid hormone due to defective thyroid hormone receptor alpha. Best Pract Res Clin Endocrinol Metab. 2015; 29: 647.

77e. Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S. A novel syndrome combining thyroid and neurological abnormalities is associated with mutations in a monocarboxylate transporter gene. Am J Hum Genet. 2004;74:168-75.

77f. Friesema EC, Gruters A, Biebermann H, Krude H, von Moers A, Reeser M, Barrett TG, Mancilla EE, Svensson J, Kester MH, Kuiper GG, Balkassmi S, Uitterlinden AG, Koehrle J, Rodien P, Halestrap AP, Visser TJ. Association between mutations in a thyroid hormone transporter and severe X-linked psychomotor retardation. Lancet 2004; 364:1435-1437.

77g. Allan W, Erdon CN, Dudley FC. Some exemples of the inheritance of mental deficiency apparently sex-linked idiocy and microcefaly. Am J Ment Defic. 1944, 48:325.

77h. Dumitrescu AM, Liao XH, Abdullah MS, et al. Mutations in SECISBP2 result in abnormal thyroid hormone metabolism. Nat Genet. 2005;37:1247-52.

  1. l’Allemand D, Gruters A, Beyer P, Weber B. Iodine in contrast agents and skin disinfectants is the major cause for hypothyroidism in premature infants during intensive care. Horm Res. 1987;28:42-9.
  2. Brown RS, Bloomfield S, Bednarek FJ, Mitchell ML, Braverman LE. Routine skin cleansing with povidone-iodine is not a common cause of transient neonatal hypothyroidism in North America: a prospective controlled study. Thyroid.1997;7:395-400.
  3. Cheron RG, Kaplan MM, Larsen PR, Selenkow HA, Crigler JF, Jr. Neonatal thyroid function after propylthiouracil therapy for maternal Graves’ disease. N Engl J Med. 1981;304:525-8.
  4. Mitsuda N, Tamaki H, Amino N, Hosono T, Miyai K, Tanizawa O. Risk factors for developmental disorders in infants born to women with Graves disease. Obstet Gynecol. 1992;80(3 Pt 1):359-64.

81a. Brown RS, Bellisario RL, Botero D, et al. Incidence of transient congenital hypothyroidism due to maternal thyrotropin receptor-blocking antibodies in over one million babies. J Clin Endocrinol Metab. 1996;81:1147-51.

81b. Brown RS, Keating P, Mitchell E. Maternal thyroid-blocking immunoglobulins in congenital hypothyroidism. J Clin Endocrinol Metab. 1990;70:1341-6.

 

81c. Connors MH, Styne DM. Transient neonatal ‘athyreosis’ resulting from thyrotropin-binding inhibitory immunoglobulins. Pediatrics. 1986;78:287-90.

  1. Mandel SH, Hanna CE, LaFranchi SH. Diminished thyroid-stimulating hormone secretion associated with neonatal thyrotoxicosis. J Pediatr. 1986;109:662-5.
  2. Deming DD, Rabin CW, Hopper AO, Peverini RL, Vyhmeister NR, Nelson JC. Direct equilibrium dialysis compared with two non-dialysis free T4 methods in premature infants. J Pediatr. 2007;151:404-8.

83a van Wassener-Leemhuis A, Ares S, Golombek S, Kok J, Paneth N, Kase J, LaGamma E. Thyroid hormone supplementation in preterm infants born before 28 weeks of gestational age and neurodevelopmental outcome at age 36 months. Thyroid. 2014; 24:1162-1169.

83b. Hollanders JJ, Israels J, van del Pal SM, Verkerk PH, Rotteveel J, Finken MJ, Duch POPS-19 collaborative study group. No Association Between Transient Hypothyroxinemia of Prematurity and Neurodevelopmental Outcome in Young Adulthood. J Clin Endocrinol Metab. 2015; 100:4648-53.

83c. Williams FL, Visser TJ, Hume R. Transient hypothyroxinaemia in preterm infants. Early Hum Dev. 2006;82:797-802.

83d. van Wassenaer AG, Kok JH, de Vijlder JJ, et al. Effects of thyroxine supplementation on neurologic development in infants born at less than 30 weeks’ gestation. N Engl J Med. 1997;336:21-6.

83e. van Wassenaer AG, Westera J, Houtzager BA, Kok JH. Ten-year follow-up of children born at <30 weeks’ gestational age supplemented with thyroxine in the neonatal period in a randomized, controlled trial. Pediatrics. 2005;116:e613-8.

  1. Williams FL, Ogston SA, van Toor H, Visser TJ, Hume R. Serum thyroid hormones in preterm infants: associations with postnatal illnesses and drug usage. J Clin Endocrinol Metab. 2005;90:5954-63.
  2. Foley TP, Jr., Abbassi V, Copeland KC, Draznin MB. Brief report: hypothyroidism caused by chronic autoimmune thyroiditis in very young infants. N Engl J Med. 1994;330:466-8.

85a. Verbansky JW, Chatila TA. Immune dysregulation, Poliendocrinopathy, Enteropathy, X linked (IPEX) and IPEX related disorders: an evolving web of heritable autoimmune disease. Curr Opin Pediatr. 2013; 25:708-714.

85b. Wildin RS, Smyk-Pearson S, Filipovich AH. Clinical and molecular features of the immunodysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. J Med Genet. 2002;39:537-45.

85c. Huang SA, Tu HM, Harney JW et al. Severe hypothyroidism caused by type 3 iodothyronine dieiodinase in infantile hemangiomas. N Eng J Med. 2000;343: 185-9.

85d. Balatz AE, Athanassaki I, Gunn SK, Tatevian N, Huang SA, Haymond MW, Karaviti L. Rapid resolution of consumptive hypothyroidism in a child with hepatic hemangioendothelioma following liver transplantation. Ann Cli Lab Sc. 2007;37:280-284.

  1. Zakarija M, McKenzie JM. Pregnancy-associated changes in the thyroid-stimulating antibody of Graves’ disease and the relationship to neonatal hyperthyroidism. J Clin Endocrinol Metab. 1983;57:1036-40.
  2. Skuza KA, Sills IN, Stene M, Rapaport R. Prediction of neonatal hyperthyroidism in infants born to mothers with Graves disease. J Pediatr. 1996;128:264-8.

87a. Abeillon-du Payret, Chikh K, Bossart N, Bretones P, Gaucherand P, Claris O, Charrie’ A, Raverot V, Orgiazzi J, Borzon- Chazot F, Bournaud C. Predictive value of maternal second-generation thyroid-binding inhibitory immunoglobulin assay for neonatal autoimmune hyperthyroidism. Eur J Endocrinol. 2014, 171:451-460.

87b. Fort P, Lifshitz F, Pugliese M, Klein I. Neonatal thyroid disease: differential expression in three successive offspring. J Clin Endocrinol Metab. 1988;66:645-7.

87c. Zakarija M, McKenzie JM, Munro DS. Immunoglobulin G inhibitor of thyroid-stimulating antibody is a cause of delay in the onset of neonatal Graves’ disease. J Clin Invest. 1983;72:1352-6.

87d. Kohn LD, Suzuki K, Hoffman WH, et al. Characterization of monoclonal thyroid-stimulating and thyrotropin binding-inhibiting autoantibodies from a Hashimoto’s patient whose children had intrauterine and neonatal thyroid disease. J Clin Endocrinol Metab. 1997;82:3998-4009.

87e. Launberg P, Wallin G, Tallstedt L, Abraham-Nording M, Lundell G, Terring O. TSH-receptor autoimmunity in Graves’ disease after therapy with antithyroid –drugs, surgery or radioiodine: a 5 year prospective randomized study. Eur J Endocrinol. 2008; 158:69-75.

87f. Kiefier FW, Kierbemass-Screhof K, Steiner M, Worda C, Kaisprian G, Tanja D, Kahaly G, Gess A. Fetal/neonatal thyrotoxicosis in a newborn from a hypothyroid woman with Hashimoto thyroiditis, J Clin Endocrinol Metab. 2017;102:6-9.

87g. Neal PR, Jansen RD, Lemons JA, Mirkin LD, Schreiner RL. Unusual manifestations of neonatal hyperthyroidism. Am J Perinatol. 1985;2:231-5.

87h. Daneman D, Howard NJ. Neonatal thyrotoxicosis: intellectual impairment and craniosynostosis in later years. J Pediatr. 1980;97:257-9.

  1. Morshed SA, Ando T, Latif R, Davies TF. Neutral antibodies to the TSH receptor are present in Graves' disease and regulate selective signaling cascades. Endocrinology. 151:5537-49, 2010.

88a. Barbesino G, Tomer Y. Clinical utility of TSH receptor antibodies. J Clin Endocrinol Metab. 2013;98:2247-2255.

88b. Diana T, Brown RS, Bossowski A, Segni M, Niedziela M, Konig J, Bossowka A, Ziora K, Smith J, Pitz S, Kanitz M, Kahaly GJ. Clinical relevance of thyroid-stimulating autoantibodies in pediatric Graves’ disease. A multicenter study. J Clin Endocrinol Metab. 2014;99:1648-1655.

88c. Ross DS, Burch HB, Cooper DS, Greenlee MC, Launberg P, Maia AL, Rivkees S, Samuels M, Sosa JA, Stan MN, Walter MA. 2016 American Thyroid Association Guidelines for Diagnosis and management of hyperthyroidism and other causes of thyrotoxicosis.Thyroid.. 2016;2:1343-1421.

88d. Donnelly MA, Wood C, Casey B, Hobbins J, Barbour LA. Early severe fetal Graves disease in a mother after after thyroid ablation and thyroidectomy. Obstetric Gynecol. 2015; 125:141-152.

88e. Besancon A, Beltrand J, Le Gac I, Luton D, Polak M. Management of neonates born to women with Graves’ disease: a cohort study. Eur J Endocrinol. 2014;170:855-862.

  1. Matsuura N, Konishi J, Fujieda K, et al. TSH-receptor antibodies in mothers with Graves’ disease and outcome in their offspring. Lancet. 1988;1:14-7.

89a. Tamaki H, Amino N, Aozasa M, et al. Universal predictive criteria for neonatal overt thyrotoxicosis requiring treatment. Am J Perinatol. 1988;5:152-8.

89b. Van der Kaay D, Wasserman JD, Palmert MR. Management of neonates born to mothers with Graves’ disease. Pediatrics. 2016;137:e20151878.

  1. de Roux N, Polak M, Couet J, et al. A new mutation of the thyroid-stimulating hormone receptor in a severe neonatal hyperthyroidism. J Clin Endocrinol Metab. 1996;81:2023-6.

90a. Holzapfel HP, Wonerow P, von Petrykowski W, Henschen M, Scherbaum WA, Paschke R. Sporadic congenital hyperthyroidism due to a spontaneous germline mutation in the thyrotropin receptor gene. J Clin Endocrinol Metab. 1997;82:3879-84.

90b. Schwab KO, Gerlich M, Broecker M, Sohlemann P, Derwahl M, Lohse MJ. Constitutively active germline mutation of the thyrotropin receptor gene as a cause of congenital hyperthyroidism. J Pediatr. 1997;131:899-904.

90c. Kopp P, Muirhead S, Jourdain N, Gu WX, Jameson JL, Rodd C. Congenital hyperthyroidism caused by a solitary toxic adenoma harboring a novel somatic mutation (serine281–>isoleucine) in the extracellular domain of the thyrotropin receptor. J Clin Invest .1997;100:1634-9.

90d. Gruters A, Schoneberg T, Biebermann H, et al. Severe congenital hyperthyroidism caused by a germ-line neo mutation in the extracellular portion of the thyrotropin receptor. J Clin Endocrinol Metab. 1998;83:1431.

90e. Paschke R, Niedzela M, Vaidya B, Persani L, Rapaport B, Leclere J. The management of familial and persistent sporadic non-autoimmune hyperthyroidism caused by thyroid stimulating hormone receptor germline mutations. Eur Thyroid J. 2012;1:142-147.

90f. Yoshimoto M. Nakayama M, Baba T, Uehara y, Nikawa N, Ito M, Tsuji Y. A case of neonatal McCune Albright syndrome with Cushing syndrome and hyperthyroidism. Acta Paediatr Scand. 1991;80:984-987.

  1. Davies TF, Amino N. A new classification for human autoimmune thyroid disease. Thyroid. 1993; 3: 331-333,

91a. Fisher DA, Pandian MR, Carlton E. Autoimmune disease: an expanding spectrum. Pediatr Clin North Am. 1987; 34: 907-18.

91b. Rallison ML, Dobyns FR, Keating FR, Rall JE, Tyler FH, Occurrence and natural history of chronic lymphocytic thyroiditis in childhood. J Pediatr. 1975; 86: 675-82.

91c. Loviselli A, Veluzzi F, Mossa P, Cambosu MA, Secci G, Atzeni F, Taberlet A, Balestrieri A, Martino E, Grasso L, Songini M, Bottazzo GF, Mariotti S. Sardinian Schoolchildren Study Group. The Sardinian Autoimmunity Study: 3. Studies on circulating antithyroid antibodies in Sardinian schoolchildren: relationship to goiter prevalence and thyroid function.Thyroid. 2001;1: 849-57.

91d. Kabelitz M, Liesenkotter KP, Stach B, Willigerodt H, Stablen W, Singendok W, Jager-Roman E, Litzenborger H, EhnertB, Gruters A. The prevalence of anti-thyroid peroxidase antibodies and autoimmune thyroiditis in children and adolescents in an iodine replete area. Eur J Endocrinol. 2003;48:301-307.

91e. Kaloumenou I, Mastorakos G, Alevizaki M, Duntas LH, Mantzou E, Ladopoulos C, Antoniou A, Chiotis D, Popassotiriou I, Chousos GP, Dacou-Voutetakis-C. Thyroid autoimmunity in schoolchildren in an area with long-standing iodine sufficiency: correlation with gender, pubertal stage, and maternal thyroid autoimmunity. Thyroid. 2008;18:747-58.

91f. Davies TF. Really significant genes for autoimmune thyroid disease do not exist–so how can we predict disease? Thyroid. 2007;17:1027-9.

91g. Brown RS. Autoimmune thyroid disease: unlocking a complex puzzle. Curr Opin Pediatr. .2009;21:523-8.

91h .Segni M, Pani MA, Pasquino AM, Badenhoop K. Familial clustering of juvenile thyroid autoimmunity: higher risk is conferred by human leukocyte antigen DR3-DQ2 and thyroid peroxidase antibody status in fathers. J Clin Endocrinol Metab 2002; 87: 3779-82.

91i.Segni M, Wood J, Pucarelli I, Toscano V, Toscano R, Pasquino AM. Clustering of autoimmune thyroid diseases in children and adolescents: a study of 66 families. J Pediatr Endocrinol Metab. 2001, 14 Supp 5:1271-5.

91j. Wiebolt J, Achterbergh R, den Boer A, van der Leij S, Marsch E, Suelmann B, de Vries R, van Haeften TW. Clustering of additional autoimmunity behaves differently in Hashimoto's patients compared with Graves' patients. Eur J Endocrinol. 2011;164:789-94.

91k. Jenkins RC, Weetman AP. Disease associations with autoimmune thyroid disease. Thyroid. 2002;12:975-986.

91l. Karlsson B, Gustafsson G, Ivarson SA, Anneren G, Thyroid dysfunction in Down’s syndrome: relation to age and thyroid autoimmunity. Arch Dis Child. 1998;79:243-245.

91m. Eisheikh M, Wass JA, Conway GS. Autoimmune thyroid disease in women with Turner ‘s syndrome-the association with karyotype. Clin Endocrinol. 2001; 55:223-226,

91n. Seminog OO, Seminog AB, Yeates D, Goldacre MJ. Associations between Klinefelter’s syndrome and autoimmune diseases: English national recortd linkage studies. Autoimmunity. 2015;48: 125-280.

91o. Quaio CR, Carvalho JF, da Silva CA, Bueno C, Brasil AS, Pereira AC, Jorge AA, Malaquias AC, Kim CA, Bertola DR. Autoimmune disease and multiple autoantibodies in 42 patients with RASopathies. Am J Med Genet. 2012; 158A: 1077-82.

91p. Brent GA. Enviromental exposures and autoimmune thyroid disease. Thyroid. 2010;20:755-761.

91q. Eshler DC, Hasam A, Tomer Y. Cutting edge: the etiology of autoimmune thyroid disease. Clin Rev Allergy Immunol. 2011; 41:190-197.

  1. Brown RS. Immunoglobulins affecting thyroid growth: a continuing controversy. J Clin Endocrinol Metab. 1995;80:1506-8.

92a. deVries L, Bulvik S, Philllip M. Chronic autoimmune thyroiditis in children and adolescents at presentation and during long-term follow up. Arch Dis Child. 2009; 94:33-7.

92b. Najjar SS. Muscular hypertrophy in hypothyroid children: the Kocher-Debre-Semelaigne syndrome; a review of 23 cases. J Pediatr. 1974;85:236-9.

92c. Hopwood NJ, Lockhart LH, Bryan GT. Acquired hypothyroidism with muscular hypertrophy and precocious testicular enlargement. J Pediatr. 1974;85:233-6.

92d. Anasti JN, Flack MR, Froehlich J, Nelson LM, Nisula BC. A potential novel mechanism for precocious puberty in juvenile hypothyroidism. J Clin Endocrinol Metab. 1995;80:276-9.

  1. Maenpaa J, Raatikka M, Rasanen J, Taskinen E, Wager O. Natural course of juvenile autoimmune thyroiditis. J Pediatr. 1985;107:898-904.

93a. Moore DC. Natural course of ‘subclinical’ hypothyroidism in childhood and adolescence. Arch Pediatr Adolesc Med. 1996;150:293-7.

93b. Lazar L, Frumkin RB, Battat E, Lebenthal Y, Phillip M, Meyerovitch J. Natural history of thyroid function tests over 5 years in a large pediatric cohort. J Clin Endocrinol Metab. 2009;94:1678-82.

93c. Aversa T, Corrias A, Salerno MC, Tessaris D, Di Mase R, Valenzise M, Corica D, De Luca F, Wasniewka M. Five-years prospective evaluation of thyroid function test evolution in children with Hashimoto’s thyroiditis presenting with either euthyroidism or subclinical hypothyroidism. Thyroid. 2016, 26:1450-1456.

  1. Hayashida N,Imaizumi M, Shimura H, Okubo N, Asari Y, Nigarawa T, Midorikava Sm Kotani K, Nakaji S, Otssuru A, Akamizu T, Kitaoka M, Suzuki S, Taniguchi N, Yamashita S, Takamura N. Thyroid ultrasound findings in children from three Japanese prefectures: Aomori, Yamanashi and Nagasaki. Plos One. 2013,8:e83220.

94a. Corrias A, Cassio A, Weber G, Mussa A, Wasniewska M, Rapa A, Gastaldi R, Einaudi S, Baronio F, Vigone MC, Messina MF, Bal M, Bona G, De Santis C; for the study for thyroid diseases of the Italian Society for Pediatric Endocrinology and Diabetology. Thyroid nodules and cancer in children and adolescents affected by autoimmune thyroiditis. Arch Pediatr Adolesc Med. 2008; 162:526-531.

94b. Keskin M, Savas-Erdeve S, Aycan Z. Co-existence of thyroid nodule and thyroid cancer in children and adolescents with Hashimoto thyroiditis: a single-center study. Horm Res Pediatr. 2016; 85:181-187.

94c. Kambalpalli M, Gupta A, Prasad UR, Francis GL. Ultrasound characteristics of the thyroid in children and adolescents with goiter: a single center experience. Thyroid. 2015; 25:176-182.

94d. Jeong SH, Hong HS, Lee FH, Kwak JJ. Papillary thyroid carcinoma arising in children and adolescent Hashimoto’s thyroiditis: ultrasonographic and pathologic findings. Int J Endocrinolol. 2016, 2397690.

94e. Gilani BB, MacGillivray MH, Voorhess ML, Mills BJ, Riley WJ, MacLaren NK. Thyroid hormone abnormalities at diagnosis of insulin-dependent diabetes mellitus in children. J Pediatr. 1984;105:218-22.

94f. Kakleas K, Soldatou A, Karachaliou F, Karavanaki K. Associated autoimmune diseases in children and adolescents with type 1 diabetes. Autoimmunity Reviews. 2015; 14:781-797.

  1. Neufeld M, Maclaren N, Blizzard R. Autoimmune polyglandular syndromes. Pediatr Ann. 1980;9:154-62.

95a. Eisenbarth GS, Gottlieb PA. Autoimmune polyendocrine syndromes. N Engl J Med. 2004;350:2068-2079.

95b. Betterle C, Greggio NA, Volpato M. Clinical review 93: Autoimmune polyglandular syndrome type 1. J Clin Endocrinol Metab. 1998;83:1049-55.

95c. Kordonouri O, Harmann R, Deiss D, Wilms M, Gruters-Kieslich A. Natural course of autoimmune thyroiditis in type 1 diabetes: association with gender, age, diabetes duration and and puberty. Arch Dis Child. 2005; 90:411-414.

95d. Bright GM, Blizzard RM, Kaiser DM, Clarke WL. Organ-specific autoantibodies in children with common endocrine disease. J Pediatr. 1982; 100:8-14.

95e. Dost A, Roher TR, Frolich-Reitherer E, Bollow E, Karges B, Bockmann A, Harmann J, Holl RW: DPV initiative and the German Competence Network Diabetes Mellitus. Horm Res Paediatr. 2015; 84: 190-8.

95f. Naiyer AJ, Shah J, Hernandez L, Kim SY, Ciaccio EJ, Cheng J, Manavalan S, Bhagat G, Green PH. Tissue transglutaminase antibodies in individuals with celiac disease bind to thyroid follicles and extracellular matrix and may contribute to thyroid dysfunction. Thyroid. .2008;18:1171-8.

95g. Roy A, Laszowska M, Sundstrom J, Lebwohl B, green PH, Kampe O, Ludvigsoon JF. Prevalence of celiac disease in patients with autoimmune thyroid disease: a meta-analysis. Thyroid. 2016; 26:880-90.

95h. Kurien M, Mollazadegan K, Sanders DS, Ludvigssson JF. Celiac disease increased risk of thyroid disease in patients with type 1 diabetes: a nationwide cohort study. Diab Care. 2016 39:371-375.

95i. Virili C, Bassotti G, Santaguida MG, Iourio R, Del Duca SC, Mercuri V, Pocarelli A, Gargiulo P. Gargano L, Centanni M. Atypical celiac disease as a cause of increased need for thyroxine: a systematic study. J Clin Endocrinol Metab. 2012; 97:E419-22.

95j. Boelaert K, Newby PR, Simmonds MJ, Holder RL, Carr-Smith JD, Heward JM, Manji N, Allahabadia A, Armitage M, Chatterjee KV, Lazarus JH, Pearce SH, Vaidya B, Gough SC, Franklyn JA. Prevalence and relative risk of other autoimmune diseases in subjects with autoimmune thyroid disease. Am J Med. 2010;123:183.e1-9.

95k Irvine WJ, Davies SH, Teitelbaum S, Delamore IW, Williams AW. The clinical and pathological significance of gastric parietal cell antibody. Ann NY Acad Sci.1965; 124:657-91.

95l. Segni M, Borrelli O, Pucarelli I, Delle Fave G, Pasquino AM, Annibale B. Early manifestations of gastric autoimmunity in patients with juvenile autoimmune thyroid diseases. J Clin Endocrinol Metab. 2004; 4944-8.

  1. Scott HS, Heino M, Peterson P, et al. Common mutations in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients of different origins. Mol Endocrinol.1998;12:1112-9.

96a. Ferre EM, Rose SR, Rosenzweig SD, Burbelo PD, Romito KR, Niemela JE, Rosen LB, Break TJ, Gu W, Hunsberger S, Browne SK, Hsu AP, Rampertaap S, Swamydas M, Collar AL, Kong HH, Lee CR, Chascsa D, Simcox T, Pham A, Bondici A, Natarajan M, Monsale J, Kleiner DE, Quezado M, Alevizos I, Moutsopoulos NM, Yockey L, Frein C, Soldatos A, Calvo KR, Adjemian J, Similuk MN, Lang DM, Stone KD, Uzel G, Kopp JB, Bishop RJ, Holland SM, Olivier KN, Fleisher TA, Heller T, Winer KK, Lionakis MS. Redifined clinical features and diagnostic criteria in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. JCI insight. 2016;1(13):e88782.

96b. Jenkins RC, Weetman AP. Disease associations with autoimmune thyroid disease. Thyroid. 2002; 12:975-986.

96c. Fallahi P, Ferrari SM, Ruffilli I, Elia G, Biricotti M, Vita R, Benvenga S, Antonelli A. The association of other autoimmune diseases in patients with autoimmune thyroiditis: Review of the literature and report of a large series of patients. Autoimm Rev. 2016;15:1125-1128.

96d. Stagi S, Giani T, Simonini G, Falcini F. Thyroid function, autoimmune thyroiditis and coeliac disease in juvenile idiopathic arthritis. Reumatology. 2005; 44:517-20.

96e. Kroon MW, Vrijman C, Chandeck C, Wind BS, Wolkerstofer A, Luiten RM, Bos JD, Geskus RB, van Trosenburg P, van der Veen JP. High prevalence of autoimmune thyroiditis in children and adolents with vitiligo. Horm Res Paediatr. 2013;79:137-44.

96f. Noso S, Park C, Babaya N, Hiromine Y, Harada T, Ito H, Taketomo Y, Kanto K, Oiso N, Kawada A, Suzuki T, Kawabata Y, Ikegami H. Organ specificity in autoimmune diseases: thyroid and islet autoimmunity in alopecia areata. J Clin Endocrinol Metab. 2015; 100:1976-1983.

96g. Leznoff A, Josse RG, Denburg J, Dolovich J. Association of chronic urticaria and angioedema with thyroid autoimmunity. Arch Dermatol. 1983;119:636-40.

96h. O’Regan S, Fong JS, Kaplan BS, Chadarevian JP, Lapointe N, Drummond KN. Thyroid antigen-antibody nephritis. Clin Immunol Immunopathol. 1976;6:341-6.

96i. Segni M, Pucarelli I, Truglia S, Turriziani I, Serafinelli C, Conti F. High prevalence of antinuclear antibodies in children with thyroid autoimmunity. J Immunol Res 2014, 150239.

96j. Manetti L, Lupi I, Morselli L, Albertini S, Cosottini M, Grasso L, Genovesi M, Pinna G, Mariotti S, Bogazzi F, Bartalena L, Martino E. Prevalence and functional significance of antipituitary antibodies in patients with autoimmune and non-autoimmune thyroid diseases. J Clin Endocrinol Metab. 2007.92:2176-81.

96k. De Bellis A, Bellastella G, Maiorino MI, Aitella E, Lucci E, Cozzolino D, Bellastella A, Bizzarro A, Giugliano D, Esposito K, Italian Autoimmune Hypophysitis network group. Longitudinal behavior of autoimmune GH deficiency: from childhood to transition age. Eur J Endocrinol. 2016;174:381-7.

  1. Mehta A, Hindmarsh PC, Stanhope RG, Brain CE, Preece MA, Dattani MT. Is the thyrotropin-releasing hormone test necessary in the diagnosis of central hypothyroidism in children?. J Clin Endocrinol Metab. 2003;88:5696-703.

97a. Feingold SB, Smith J, Houtz J, et al. Prevalence and functional significance of thyrotropin (TSH) receptor blocking antibodies in children and adolescents with chronic lymphocytic thyroiditis. J Clin Endocrinol Metab. 2009;94: 4742-8.

97b. Brown RS, Alter CA, Sadeghi-Nejad A. Severe unsuspected maternal hypothyroidism discovered after the diagnosis of thyrotropin receptor-blocking antibody-induced congenital hypothyroidism in the neonate: failure to recognize and implication to the fetus Horm Res Pediatr. 2015; 83:132-8.

  1. Pedersen OM, Aadal NP, Larssen TB, Varhuag JE, Myking O, Vik-Mo H. The value of ultrasonography in predicting autoimmune thyroid disease. Thyroid. 2000, 10:251-259.
  2. Rovet JF, Daneman D, Bailey JD. Psychologic and psychoeducational consequences of thyroxine therapy for juvenile acquired hypothyroidism. J Pediatr. 1993;122:543-9.

99a. Van Dop C, Conte FA, Koch TK, Clark SJ, Wilson-Davis SL, Grumbach MM. Pseudotumor cerebri associated with initiation of levothyroxine therapy for juvenile hypothyroidism. N Engl J Med.1983;308:1076-80.

99b. Lomenick JP, El-Sayyid M, Smith WJ. Effect of levo-thyroxine treatment on weight and body mass index in children with acquired hypothyroidism. J Pediatr. 2008. 152:96-100.

99c. Rivkees SA, Bode HH, Crawford JD. Long-term growth in juvenile acquired hypothyroidism: the failure to achieve normal adult stature. N Engl J Med. 1988;318:599-602.

  1. Salerno M, Capalbo D, Cerbone M, De Luca F. Subclinical hypothyroidism in childhood- current knowledge and open issues. Nat Rev Endocrinol. 2016; 734-746.

100a. Surks MI, Ortiz E, Daniels GH, et al. Subclinical thyroid disease: scientific review and guidelines for diagnosis and management. Jama. 2004;291:228-38.

100b. Taylor P, Razvi S, Pearce Sh, Colin D. A Review of the clinical consequences of variation in thyroid function whitin the reference range. J Clin Endocrinol Metab. 2013; 98: 3562-3571,

100c. Ittermann T, Thamm M, Wallschofski H, Rettig R, Volzke H. Serum thyroid-stimulating hormone levels are associated with blood pressure in children and adolescents. J Clin Endocrinol Metab. 2012; 97:828-834.

100d. Witte T, Ittermann T, Thamm M, Ribiet NB, Votzke H. Association between serum thyroid stimulating hormone levels and serum lipids in children and adolescents: a population-based study on german youth. J Clin Endocrinol Metab. 2015;100:2090-7.

100e. Nader NS, Bahn RS, Johnson ,MD, Weaver AL, Singh R, Kumar S. Relationships between thyroid function and lipid status or insulin resistance in a pediatric population. Thyroid. 2010;20:1333-1339.

100f. McLeod DS, Watters KF, Carpenter AD, Landenson PW, Cooper DS, Ding EL. Thyrotropin and thyroid cancer diagnosis: a systematic review and dose-response meta-analysis. J Clin Endocrinol Metab. 2012; 97:2682-2692.

100g. Lazarus J, Brown RS, Daumerie C, Hubalewska-Dydejczyk A, Negro R, Vaydya B. European thyroid association guidelines for the management of subclinical hypothyroidism in pregnancy and in children. Eur Thyroid J. 2014; 3: 76-94.

  1. Surks MI, Sievert R. Drugs and thyroid function. N Engl J Med. 1995;333:1688-94.

101a. McCowen KC, Garber JR, Spark R. Elevated serum thyrotropin in thyroxine-treated patients with hypothyroidism given sertraline. N Engl J Med. 1997;337:1010-1.

101b. Delange FM. Iodine Deficiency. In: Braverman LE, Utiger,R.D., ed. Werner & Ingbar’s The Thyroid. 8th ed. Philadelphia: Lippincott Williams & Wilkins; 2000.

101c. Pacaud D, Van Vliet G, Delvin E, et al. A Third World endocrine disease in a 6-year-old North American boy. J Clin Endocrinol Metab. 1995;80:2574-6.

101d. Donadieu J, Rolon MA, Thomas C, et al. Endocrine involvement in pediatric-onset Langerhans ’ cell histiocytosis: a population-based study. J Pediatr. 2004;144:344-50.

  1. Hauser P, Zametkin AJ, Martinez P, et al. Attention deficit-hyperactivity disorder in people with generalized resistance to thyroid hormone. N Engl J Med. 1993;328:997-1001.

102a. Bercu BB, Orloff S, Schulman JD. Pituitary resistance to thyroid hormone in cystinosis. J Clin Endocrinol Metab. 1980;51:1262-8.

  1. van der Gaag RD, Drexhage HA, Wiersinga WM, et al. Further studies on thyroid growth-stimulating immunoglobulins in euthyroid nonendemic goiter. J Clin Endocrinol Metab. 1985;60:972-9.
  2. Rother KI, Zimmerman D, Schwenk WF. Effect of thyroid hormone treatment on thyromegaly in children and adolescents with Hashimoto disease. J Pediatr. 1994;124:599-601.
  3. Svensson J, Ericsson UB, Nilsson P, Olsson C, Jonsson B, Lindberg B, Ivarsson SA. Levothyroxine treatment reduces thyroid size in children and adolescents with chronic autoimmune thyroiditis. J Clin Endocrinol Metab. 2006. 91:1729-34.
  4. Smith SL, Pereira KD Suppurative thyroiditis in children. A management algorithm. Pediatr Emerg Care. 2008; 24:764-767.

106a. Greene JN. Subacute thyroiditis. Am J Med. 1971:51:97.

106b. Paes JE, Burman KD,.Cohen J, Franklyn J, McHemry CR, Shoham S, Kloos RT. Acute bacterial suppurative thyroiditis: a clinical review and expert opinion. Thyroid. 2010; 20:247-255.

106c. Mali VP, Prabhakaran K. Recurrent acute thyroid swellings because of pyriform sinus fistula. J Pediatr Surg. 2008;43:e27-30.

106d. Radfar N,Kenny FM, Larsen PR. Subacute thyroiditis in a lateral thyroid gland:evaluation of the pituitary-thyroid axis during the acute distructive and the recovery phases. J Pediatr. 1975; 87:34.

  1. Braverman L, Utiger R: Introduction to thyrotoxicosis. In: Braverman L, Utiger R eds. The Thyroid. 9th ed. Philadelphia: Lippincott Williams & Wilkins; 453-455, 2005.

107a. Graves RJ: Clinical lectures. London Med Surg J (pt 2):516, 1835.

107b. Kjaer H, Andersen SM, Hansen D. Increasing incidence of juvenile thyrotoxicosis in Denmark: a nationwide study, 1998-2012. Horm Res Paediatr. 2015;84: 102-107.

107c. Shulman DI, Muhar I, Jorgensen EV, Diamond FB, Bercu BB, Root AW. Autoimmune hyperthyroidism in prepubertal children and adolescents: comparison of clinical and biochemical features at diagnosis and responses to medical therapy. Thyroid. 1997;7:755-60.

107d. Segni M, Leonardi E, Mazzoncini B, Pucarelli I, Pasquino AM. Special features of Graves ’ disease in early childhood. Thyroid. 1999;9:871-7.

107e. Goday-Arno A, Cerda-Esteva M, Flores-Le-Roux JA, Chillaron-Jordan JJ, Corretger JM, Cano-Perez JF. Hyperthyroidism in a population with Down syndrome (DS). Clin Endocrinol. 2009;71:110-4.

107f. Rapoport B, Chazenbalk GD, Jaume JC, McLachlan SM. The thyrotropin (TSH) receptor: interaction with TSH and autoantibodies. Endocr Rev. 1998;19:673-716.

107g. Smith BR, Sanders J, Furmaniak J. TSH receptor antibodies. Thyroid. 2007;17:923-38.

107h. Sanders J, Evans M, Premawardhana LD, et al. Human monoclonal thyroid stimulating autoantibody. Lancet. 2003;362:126-8.

107i. Sanders J, Miguel RN, Bolton J, et al. Molecular interactions between the TSH receptor and a thyroid-stimulating monoclonal autoantibody. Thyroid. 2007;17:699-706.

107j. Rapoport B, McLachlan SM. The thyrotropin receptor in Graves’ disease. Thyroid .2007;17:911-22.

107k. Williams RC, Jr., Marshall NJ, Kilpatrick K, et al. Kappa/lambda immunoglobulin distribution in Graves’ thyroid-stimulating antibodies. Simultaneous analysis of C lambda gene polymorphisms. J Clin Invest. 1988;82:1306-12.

  1. Segni M, Gorman CA. The aftermath of childhood hyperthyroidism. J Pediatr Endocrinol Metab. 2001; Suppl 5 1277-82.

108a. Goldstein SM, Katowitz WR, Modhang T, Katowitz JA. Pediatric Thyroid-Associated Orbitopathy: The children’s hospital of Philadelphia experience and literature review. Thyroid. 2008;18:997-999.

108b. Nucci P, Brancato R, Bandello F, Alfarano R, Bianchi S. Normal exophtalmometric values in children. Am J Opthal 1989;108:582-584.

108c. Liu GT, Helher KL, Katowitz JA, Kazim M, Moazami G, Moshang T, Teener JW, Sladeky J, Volpe NJ, Galetta SL. Prominent proptosis in childhood thyroid eye disease. Opthalmology. 1996;103:779-784.

108d. Durairaj VD, Bartley GB, Garrity JA. Clinical features and treatment of Graves’ opthalmopathy in pediatric patients. Opthal Plast Reconstr Surg 2006; 22:7-12.

  1. Sterling K, Refetoff S, Selenkow HA. T3 thyrotoxicosis due to elevated serum triiodothyronine levels. JAMA. 1970;213:571-5.

109a. Harvengt J, Boizeau P, Chevenne D, Zenaty D, Paulsen A, Simon D, Crepon SG, Alberti C, Carel JC, Leger J. Triiodothyronine- predominant Graves’ disease in childhood:detection and therapeutic inplications. Eur J Endocrinol. 2015; 172: 715-23.

109b. Ruiz M, Rajatanavin R, Young RA, Taylor C, Brown R, Braverman LE, Ingbar SH. Familial dysalbuminemic hyperthyroxinemia: a syndrome that can be confused with thyrotoxicosis. N Engl J Med. 1982;306:635-9.

109c. Mariotti S, Martino E, Cupini C, et al. Low serum thyroglobulin as a clue to the diagnosis of thyrotoxicosis factitia. N Engl J Med. 1982;307:410-2.

109d. Kaguelidou F, Alberti C, Castanet M, Guitteny MA, Czernichow P, Leger J. Predictors of autoimmune hyperthyroidism relapse in children after discontinuation of anthythyroid drug treatment. J Clin Endocrinol Metab. 2008; 93:3817-3826.

  1. Ichiki Y, Akahoshi M, Yamashita N, et al. Propylthiouracil-induced severe hepatitis: a case report and review of the literature. J Gastroenterol. 1998;33:747-50.

110a. Russo MW, Galanko JA, Shrestha R, Fried MW, Watkins P. Liver transplantation for acute liver failure from drug induced liver injury in the United States. Liver Transpl. 2004;10:1018-23.

110b. Rivkees SA, Mattison DR. Ending propylthiouracil-induced liver failure in children. N Engl J Med. 2009;360:1574-5.

110c. Bahn RS, Burch HS, Cooper DS, Garber JR, Greenlee CM, Klein IL, Laurberg P, McDougall IR, Rivkees SA, Ross D, Sosa JA, Stan MN. The Role of Propylthiouracil in the Management of Graves’ Disease in Adults: report of a meeting jointly sponsored by the American Thyroid Association and the Food and Drug Administration. Thyroid. 2009;19:673-4.

110d. Okuno A, Yano K, Inyaku F, Suzuki Y, Sanae N, Kumai M, Naitoh Y. Pharmacokinetics of methimazole in children and adolescents with Graves’ disease. Acta Endocrinol.1987. 115:112-118.

110e. Rivkees SA. Pediatric Graves’ disease: management in the post-propilthyuracil era .Inter J Pediatr Endocrinolol. 2014; 10:1-10.

110f. Smith J, Brown RS. Persistence of thyrotropin (TSH) receptor antibodies in children and adolescents with Graves’ disease treated using antithyroid medication. Thyroid. 2007;17:1103-7.

110g. Fenzi G, Hashizume K, Roudebush CP, DeGroot LJ. Changes in thyroid-stimulating immunoglobulins during antithyroid therapy. J Clin Endocrinol Metab. 1979;48:572-6.

110h. Teng CS, Yeung RT. Changes in thyroid-stimulating antibody activity in Graves’ disease treated with antithyroid drug and its relationship to relapse: a prospective study. J Clin Endocrinol Metab. 1980;50:144-7.

110i. Bliddal H, Kirkegaard C, Siersbaek-Nielsen K, Friis T. Prognostic value of thyrotrophin binding inhibiting immunoglobulins (TBII) in longterm antithyroid treatment, 131I therapy given in combination with carbimazole and in euthyroid ophthalmopathy. Acta Endocrinol. 1981;98:364-9.

110j. Collen RJ, Landaw EM, Kaplan SA, Lippe BM. Remission rates of children and adolescents with thyrotoxicosis treated with antithyroid drugs. Pediatrics. 1980;65(3):550-6.

110k. Leger J, Gelwane G, Kaguelidou F et al. Positive impact of long-term antithyroid drug treatment on the outcome of children with Graves disease: natural long-term cohort study. J Clin Endocrinol Metab. 2012; 97:110-9.

110l. Hashizume K, Ichikawa K, Sakurai A, et al. Administration of thyroxine in treated Graves’ disease. Effects on the level of antibodies to thyroid-stimulating hormone receptors and on the risk of recurrence of hyperthyroidism. N Engl J Med. 1991;324:947-53.

110m. McIver B, Rae P, Beckett G, Wilkinson E, Gold A, Toft A. Lack of effect of thyroxine in patients with Graves’ hyperthyroidism who are treated with an antithyroid drug. N Engl J Med 1996;334:220-4.

  1. Rivkees SA, Sklar C, Freemark M. Clinical review 99: The management of Graves’ disease in children, with special emphasis on radioiodine treatment. J Clin Endocrinol Metab. 1998;83:3767-76.

111a. Rivkees SA, Stephenson K, Dinauer C. Adverse events associated with methimazole therapy of Graves’ disease in children. Int J Pediatr Endocrinol. 2010; 176970.

111b. Wada N, Mukai M, Kohono M, Notoya A, Ito T, Yoshioka N. Prevalence of serum anti-myeloperoxidase antineutrophil cytoplasmatic antibodies (MPO-ANCA) in patients with Graves’ disease trated with prophilthyuracil and thiamazole. Endocr J. 2002; 49: 329-334.

111c. Guma M, Salinas I, Reverter JL, Roca J, Valls-Roc M, Juan M, Olive’ A. Frequency of antineutrophil cytoplasmic antibody in Graves’ disease patients treated with methimazole. J Clin Endocrinol Metab. 2003, 88:2141-6.

111d. Tajiri J, Noguchi S, Murakami T, Murakami N. Antithyroid drug-induced agranulocytosis. The usefulness of routine white blood cell count monitoring. Arch Intern Med. 1990;150:621-4.

111e. van Veenendaal NR, Rivkees SA. Treatment of pediatric Graves’ disease is associated with excessive weight gain. J Clin Endocrinol Metab. 2011; 96:3257-63.

111f. Mazzaferri EL. Thyroid cancer and Graves’ disease. J Clin Endocrinol Metab. 1990; 70:825-829.

111g. Dobyns BM, Sheline GE, Workman JB, Tompkins EA, McConahey WM, Becker DV. Malignant and benign neoplasm of the thyroid in patients treated for hyperthyroidism: a report of the cooperative thyrotoxicosis therapy follow-up study. J Clin Endocrinol Metab. 1974; 38:976-988.

  1. Sherman J, Thompson GB, Lteif A, et al. Surgical management of Graves disease in childhood and adolescence: an institutional experience. Surgery. 2006;140:1056-61. Discussion 61-2.

112a.Tuggle CT, Roman SA, Wang TS, Thomas DC, Uldelsman R, Ann Sosa J. Pediatric endocrine surgery: who is operating on our children? Surgery. 2008; 144:869-877, Discussion 877.

112b. Kundel A, Thompson GB, Richards ML, Qiu LX, Cai Y, Schwenk FW, Lief AN, Pittock ST, Kumar S, Tebben PJ Hay ID, Grant CS. Pediatric endocrine surgery: a 20 year experience at the Mayo Clinic. J Clin Endocrinol Metab. 2014. 99: 399-406.

112c. Beuer CK, Solomon D, Donovan P, Rivkees SA, Udelsman R. Effect of patient age on surgical outcome for Graves’ disease: a case control study of 100 consecutive patients at a high volume thyroid surgical center. Int J Pediatr Endocrionol. 2013;1: 9856.

  1. Nikiforov Y, Gnepp DR, Fagin JA. Thyroid lesions in children and adolescents after the Chernobyl disaster: implications for the study of radiation tumorigenesis. J Clin Endocrinol Metab. 1996;81:9-14.

113a. Safa AM, Schumacher P, Rodriguez-Antunez A. Long-term follow-up results in children and adolescents treated with radioactive iodine (131-Iodine). N Engl J Med. 1975; 292:167-171.

113b. Holm LE, Dahlqvist I, Israelsson A, Lundell GM. Malignant thyroid tumors after 131-iodine therapy. N Engl J Med. 1980; 303:188-191.

113c. Hamburger JI. Management of hyperthyroidism in children and adolescents. J Clin Endocrinol Metab. 1985; 60:1019.

113d. Hayek A, Chapman E, Crawford JD. Long-term results of treatment of thyrotoxicosis in children and adolescents with radioactive iodine. N Engl J Med .1970; 283:949, 953.

113e. Franklyn JA, Maisonneuve P, Sheppard M, Betteridge PB.Bovie P. Cancer incidence and mortality after radioiodine treatment for hyperthyroidism: a population-based cohort study. Lancet. 1999;353:2111-5

113f. Starr P, Jaffe HL, Oettinger L Jr. Late results of 131-I treatment of hyperthyroidism in seventy-three children and adolescents. J Nucl Med.1964; 5:81-9.

113g. Kogut MD, Kaplan SA, Collipp PJ, Tiamsic T, Boyle D. Treatment of hyperthyroidism in children. N Engl J Med.1965; 272:217-22.

113h. Read CH Jr, Tansey MJ, Menda Y. A 36-year retrospective analysis of the efficacy and safety of radioactive iodine in treating young Graves’ patients. J Clin Endocrinol Metab. 2004;89:4229-33.

113i. Rivkees SA, Dinauer C. An optimal treatment for pediatric Graves’ disease is radioiodine. J Clin Endocrinol Metab. 2007;92:797-800.

113j. Rivkees SA. Controversies in the management of Graves’ disease in children. J Endocrinol Invest. 2016;39:1247-1257.

  1. Ly S, Frates MC, Benson CB, Peters HE, Grant FD, Drubach LA, Voss SD, Feldman HA, Smith JR, Barletta J, Hollowell M, Cibas ES, Moore FD, Modi BM, Shamberger RC, Huang SA. Features and outcome of autonomous thyroid nodules in children: 31 consecutive patients seen at a single center. J Clin Endocrinol Metab. 2016;101:3856-3862.

114a. Niedzela M, Breborowicz D, Trejster E, Koeman E. Hot nodules in children and adolents in western Poland from 1996 to 2000: clinical analysis of 31 patients. J Pediatr Endocrinol Metab. 2002;15:823-830.

114b. Francis GL, Waguenspack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID, Luster M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S. Management guidelines for children with thyroid nodules and differentiated thyroid cancer. Thyroid. 2015:716-759.

114c. Gershergorn MC, Weintraub BD. Thyrotropin-induced hyperthyroidism caused by selective pituitary resistance to thyroid hormone. A new syndrome of “inappropriate secretion of TSH”. J Clin Invest. 1975;56:633-642.

114d. Beck-Peccoz P, Lania A, Beckers A, Chatterrjee K, Wernau JL. European Thyroid association guidelines for the diagnosis and treatment of thyrotropin-secreting pituitary tumors. Thyroid J. 2013;2-76-82.

114e. Daly AF, Tichomirova MA, Petronians P, Heliovaara E, Jaffrain-Rea ML, Barlus R et al. Clinical characteristics and therapeutic responses.in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study. J Clin Endocrinol Metab. 2010; 95:E373-E383.

114f. Misra M, Levitsky LL, Lee MM. Transient hyperthyroidism in an adolescent with hydatidiform mole. J Pediatr. 2002;140:362-6.

114g. Hershman JM. Human chorionic gonadotropin and the thyroid: hyperemesis gravidarum and throphoblastic tumors. Thyroid. 1999; 9:653-657.

115a. Wells SA, Asa SL, Dralle H, Elisei R, Evans DB, Gagel RF, Lee N, Machens A, Moley JF, Pacini F, Raue F, Frank-Raue K, Robison B, Rosental MS, Santoro M, Schulberger M, Shah M, Waguespack SG, American Thyroid Association Guidelines task force on medullary thyroid carcinoma. Thyroid. 2015; 25:567-610.

115b. Hung W, Anderson KD, Chandra RS, et al. Solitary thyroid nodules in 71 children and adolescents. J Pediatr Surg. 1992;27:1407-9.

115c . Schlumberger M, De Vathaire F, Travagli JP, et al. Differentiated thyroid carcinoma in childhood: long term follow-up of 72 patients. J Clin Endocrinol Metab. 1987;65:1088-94.

115d . Flannery TK, Kirkland JL, Copeland KC, Bertuch AA, Karaviti LP, Brandt ML. Papillary thyroid cancer: a pediatric perspective. Pediatrics 1996;98(3 Pt 1):464-6.

115e. Lairmore TC, Frisella MM, Wells SA, Jr. Genetic testing and early thyroidectomy for inherited medullary thyroid carcinoma. Ann Med. 1996;28:401-6.

115f. Sarne D, Schneider AB. External radiation and thyroid neoplasia. Endocrinol Metab Clin North Am. 1996;25:181-95.

115g. Healy JC, Shafford EA, Reznek RH, et al. Sonographic abnormalities of the thyroid gland following radiotherapy in survivors of childhood Hodgkin’s disease. Br J Radiol. 1996;69:617-23.

115h. Soberman N, Leonidas JC, Cherrick I, Schiff R, Karayalcin G. Sonographic abnormalities of the thyroid gland in longterm survivors of Hodgkin disease. Pediatr Radiol. 1991;21:250-3.

115k. Dinauer CA, Breuer C, Rivkees SA. Differentiated thyroid cancer in children: diagnosis and management. Curr Opin Oncol. 2008;20:59-65.

115i. Waguespack SG, Francis G. Initial management and follow up of differentiated thyroid cancer in children. J Natl Compr Canc Netw. 2010; 8:1289-300.

115l. Gharib H, Goellner JR. Fine-needle aspiration biopsy of thyroid nodules. Endocr Pract .1995;1:410-7.

115m. Zimmerman D, Hay ID, Gough IR, et al. Papillary thyroid carcinoma in children and adults: long-term follow-up of 1039 patients conservatively treated at one institution during three decades. Surgery. 1988;104:1157-66.

115o. Ladenson PW, Braverman LE, Mazzaferri EL, et al. Comparison of administration of recombinant human thyrotropin with withdrawal of thyroid hormone for radioactive iodine scanning in patients with thyroid carcinoma. N Engl J Med. 1997;337:888-96.

115p. Wohllk N, Cote GJ, Evans DB, Goepfert H, Ordonez NG, Gagel RF. Application of genetic screening information to the management of medullary thyroid carcinoma and multiple endocrine neoplasia type 2. Endocrinol Metab Clin North Am. 1996;25:1-25.

115q. Fogelfeld L, Wiviott MB, Shore-Freedman E, et al. Recurrence of thyroid nodules after surgical removal in patients irradiated in childhood for benign conditions. N Engl J Med. 1989;320:835-40.

115r. de Vathaire F1, Haddy N1, Allodji RS1, Hawkins M1, Guibout C1, El-Fayech C1, Teinturier C1, Oberlin O1, Pacquement H1, Diop F1, Kalhouche A1, Benadjaoud M1, Winter D1, Jackson A1, Bezin Mai-Quynh G1, Benabdennebi A1, Llanas D1, Veres C1, Munzer M1, Nguyen TD1, Bondiau PY1, Berchery D1, Laprie A1, Deutsch E1, Lefkopoulos D1, Schlumberger M1, Diallo I1, Rubino C. Thyroid Radiation Dose and Other Risk Factors of Thyroid Carcinoma Following Childhood Cancer. J Clin Endocrinol Metab. 2015 ;100:4282-90.

115s. Vivanco M1, Dalle JH, Alberti C, Lescoeur B, Yakouben K, Carel JC, Baruchel A, Léger J. Malignant and benign thyroid nodules after total body irradiation preceding hematopoietic cell transplantation during childhood. Eur J Endocrinol. 2012 ;167):225-233.

Endocrine Hypertension in Childhood

ABSTRACT

Hypertension in children is an important health issue and deserves a greater awareness among health care providers and the general population. When evaluating a suspected hypertensive child, it is essential that clinicians utilize proper tools to measure and interpret the blood pressure (BP) readings. The preferred method is auscultation using a mercury sphygmomanometer connected to the appropriate size cuff. Systolic blood pressure (BP) is determined by the onset of the "tapping" Korotkoff sounds (K1) while diastolic DBP is defined as the fifth Korotkoff sound (K5), or the disappearance of Korotkoff sounds. Automated devices can be used for BP measurement in newborns and young infants, in whom auscultation is difficult. An elevated BP reading obtained with an oscillometric device should be repeated with auscultation. To determine percentile of BP, the values are compared to normal BP in children and adults adjusted for age, sex and height. For complete coverage of this and related areas in Endocrinology, visit our free web-books, www.endotext.org and www.thyroidmanager.org.

INTRODUCTION

Hypertension is defined as average systolic BP and/or diastolic BP that is ≥95th percentile for gender, age, and height on ≥3 occasions (1, 2). Regulation of systemic BP is a function of three components: intravascular volume, cardiac output and peripheral resistance. The effect(s) of steroids on one or more of these components contribute to BP control. The binding of glucocorticoids (GCs) to its receptor enhances the vascular smooth muscle response to vasopressive agents. Activation of the mineralocorticoid (MC) receptor by the ligands leads to an increase in sodium resorption which results in water retention and intravascular volume expansion. These hemodynamic changes affect peripheral resistance and cardiac output, which in turn regulates systemic BP.

The human adrenal gland is composed of a cortex and a medulla. While adrenal medulla produces bioamines that act as vasopressors, the cortex secretes classes of steroids. In the cortices, there are three distinct zones, each having a characteristic steroid profile (figure 1). In the outer most unit, zona glomerulosa, MCs are produced. The main MC in a physiologic state is aldosterone. Principal regulators of aldosterone secretion are the renin-angiotensin system and the serum potassium concentration. Other regulators, such as the adrenocorticotropic hormone (ACTH), atrial natriuretic factors of cardiac origin and local dopamine secreted within the adrenal, play minor roles. Decreases in intravascular volume result in increased secretion of renin by the renal juxtaglomerular apparatus. Renin acts as a proteolytic enzyme by cleaving angiotensinogen, and changes it to angiotensin I. Angiotensin I is then cleaved and activated by angiotensin-converting enzyme (ACE) in the lung and in other peripheral sites. Angiotensin II and its metabolite, angiotensin III, possess vasopressor and potent aldosterone secretory activity (figure 2). Once bound to the mineralocortiocoid receptor (MR), aldosterone enhances sodium resorption and the subsequent osmotic reabsorption of water through sodium-permeable channels in the apical membranes of the epithelial cells lining the distal tubules and collection ducts of the kidney. This results in an expanded blood volume and suppression of renin secretion. Potassium excretion also occurs as an ion exchange from the aldosterone effect.

In the middle adrenal zone, zona fasiculata, GC are produced. The principal GC in humans is cortisol, which serves many physiologic roles including glucose homeostasis and vascular integrity. The hypothalamic pituitary-adrenal or HPA axis determines the threshold for circulating GC concentration.

The inner zone, or zona reticuralis, is where adrenal androgens are produced (see chapter 3 in the Adrenal Physiology and Disease section). The clinical significance of its overproduction is evident in 11β-hydroxylase deficiency (11β-OHD). In this deficiency, steroid precursors proximal to the block shunted to androgen pathways which leads to virilization of the affected individual (see below).

Endocrine hypertension in children is usually mediated by the MC activities of cortisol, aldosterone and adrenal steroidogenic precursors with MC activity. Frequently in these cases, elevated BP is associated with suppressed renin activity, indicating a form of hypertension related with volume-overload and salt-sensitivity.

In the past few decades, considerable progress has been made toward unraveling the molecular genetics of some rare, or extremely rare, monogenic forms of hypertension (1).

CONGENITAL ADRENAL HYPERPLASIA (CAH); 11Β-OHD AND 17-OHD,

These include the following well-characterized disorders: two forms of

-hydroxylase deficiencies, glucocorticoid-remediable hyperaldosteronism (familial hyperaldosteronism type I), apparent mineralocorticoid excess, and Liddle's Syndrome. This chapter describes the important causes of endocrine hypertension in children as

well as some conditions with a similar presentation (Fig. 3).

Figure 3 Monogenetic mineralocorticoid hypertension syndromes. PRA = Plasma renin activity; FH = family hy- peraldosteronism; CAH = congenital adrenal hyperplasia; DOC = deoxycortisol; F/E = cortisol/cortisone ratio.

STEROID 11β- HYDROXYLASE DEFICIENCY CONGENTIAL ADRENAL HYPERPLASIA

CAH is a family of disorders characterized by enzymatic defects in one of the cortisol production steps. Steroid 11β-OHD is the second most common cause of CAH, accounting for 5-8% of all CAH cases (3). It occurs 1 in 100,00 live births (4) in the general population, but is more common in populations of North African origin (5).

Deficiency of 11β-hydroxylation causes a decrease in the conversion of 11-deoxycortisol (S) and 11-deoxycorticosterone (DOC) to cortisol and corticosterone, respectively (figure 1). Reduced cortisol feedback gives rise to an increase in ACTH secretion. Excessive ACTH secretion in turns leads to overproduction of precursors proximal to the enzyme block. These precursors serve as substrates for the unimpeded androgen pathways; therefore adrenal androgen secretion is increased. Virilization and hypertension are the salient clinical features of 11β-OHD.

The severity of in utero virilization of the external genitalia can vary from mild to severe, such that it is not uncommon to misassign an 11β-OHD affected female as a male (6,7). Males and females may manifest signs of androgen excess at any phase of postnatal development, including precocious pubic hair, advanced somatic and epiphyseal development, and central precocious puberty later in childhood. Without treatment, early epiphyseal maturation results in short stature.

Hypertension is a less consistent feature than virilization in 11β-OHD CAH. Despite failure of aldosterone production, upstream accumulation of deoxycorticosterone (DOC), a weak MC, causes salt retention and hypertension. Hypertension is usually not identified until later in childhood or in adolescence, although its appearance in an infant 3 months of age has been documented (8). In addition, hypertension correlates variably with biochemical values, or with the degree of virilization. Some of the severely virilized females were normotensive, whereas mildly virilized patients experienced severe hypertension, leading to fatal vascular accidents (9). An unusual presentation of neonatal salt wasting has also been reported (10). The complications of long standing uncontrolled hypertension, such as cardiomyopathy, retinal vein occlusion, and blindness have been reported in 11β-OHD patients (11,12). Potassium depletion develops concomitantly with sodium retention, but hypokalemia is variable.

Hormonal characteristics include elevation of compound S, DOC and androgens. Elevation of 17α-hydroxyprogesterone occurs, but not as greatly as in 21-hydroxylase deficiency (21OHD) CAH. Tetrahydro-11-deoxycortisol and tetrahydrodeoxycorticosterone, the principal metabolites of compound S and DOC, are significantly increased in the urine. Urinary 17-ketosteroids are elevated, reflecting the raised serum levels of adrenal androgens. Renin production is suppressed secondary to MC -induced sodium retention and volume expansion. Aldosterone production is low due to low serum potassium and low plasma renin.

Steroid 11β-OHD CAH is the result of autosomal recessive mutations in CYP11B1 gene. More than 50 mutations, including missense/nonsense, splicing, small/ gross deletions, insertions and complex rearrangement, which are responsible for 11β-OHD CAH have been described in CYP11B1 gene (14). A homozygous deletion of hybrid CYP11B2/CYP11B1, a reciprocal product of the recombination event as found in glucocorticoid remediable aldosteronism (GRA), leads to clinical phenotypes of neonatal salt wasting (due to diminished aldosterone synthase acitivity). This patient (10) also has 11β-OHD deficiency.

Treatment

Cortisol administration provides cortisol replacement and normalizes ACTH. This in turn removes the drive for oversecretion of DOC and in most cases brings about remission of hypertension, if diagnosed early in life. The goal is to replace deficient steroids while minimizing adrenal sex hormone and GC excess. Serum DOC and androgens are thus the indices of the adequate hormonal control. Plasma renin activity is also useful as a therapeutic index. In poor control cases with 11β-OHD, plasma renin is suppressed.

Similar to 21OHD CAH, oral hydrocortisone is preferred, because it is identical to physiologic GC. Typical dosing is 10–15 mg/m2·d in divided doses. Long-acting GCs may be an option at or near the completion of linear growth. Titration of the dose should be aimed at maintaining androgen levels at age and sex-appropriate levels and normalization of renin. Concurrently, over-treatment should be avoided because it can lead to Cushing syndrome. Depending on the degree of stress, stress dose coverage may require doses of up to 50-100 mg/m2/day. Each family must be given injectable hydrocortisone for emergency use (at the dose of 25 mg for infants, 50 mg for young children and 100 mg for adolescents and adults, intramuscularly). In the event of surgical procedure, a total of 5-10 times the daily maintainance dose (depending on the nature of the surgical procedure) may be required over the first 24 hours. Hydrocortisone dosage can be tapered down to maintenance dose during the first few days postoperatively, provided that there is no complication. Stress dose should not be given in the form of dexamethasone because of the delayed onset of action.

In children with advanced bone age, initiation of therapy may precipitate central precocious puberty, requiring treatment with a GnRH agonist. Growth hormone therapy improves height deficit in patients with poor height prediction (13). In patients with long duration of hypertension before diagnosis, additional spironolactone, calcium channel blockers or amiloride may be necessary. Reconstructive surgery of external genitalia should be performed by experienced surgeons.

Prenatal diagnosis and treatment can be accomplished using extracted fetal DNA for CYP11B1 analysis (4,15,16). An established protocol of prenatal treatment in 21OHD CAH can be applied to 11β-OHD CAH (also see Chapter 8 – Congenital Adrenal Hyperplasia)

STEROID -17 HYDROXYLASE DEFICIENCY CONGENTIAL ADRENAL HYPERPLASIA

17-OHD results from mutations in the cytochrome P450C17 enzyme which functions both as steroid 17α-hydroxylase and as 17, 20-lyase (17). The structural gene for cytochrome P450C17 (CYP17A1) has been mapped to chromosome 10q24.3 (18). Over 50 mutations in this gene have been described. Nucleotide substitution, causing missense or nonsense alterations, accounts for the majority of the patients reported (14). It is a rare disease identified in approximately 120 patients worldwide. The enzyme deficiency causes diminished production of cortisol and sex steroids, whose production requires the 17, 20-lyase function of the same 17α- hydroxylase enzyme (Figure 1). Because both adrenals and gonads share the enzyme defect, there is decreased biosynthesis of (i) androgens, results in an undervirilized phenotype in males (46,XY) at birth, and a failure of male pubertal development. (ii) estrogen, results in females at pubertal age presenting with primary amenorrhea and lack of development of secondary sex characteristics.

Reciprocal elevation of ACTH, due to low cortisol, increases synthesis of DOC and corticosterone via the unaffected 17-deoxy pathway. Therefore hypertension and hypokalemia may comprise the primary presentation at any age or can be associated with the abnormal sexual phenotype. As in 11β- OHD, the formation of aldosterone is reduced secondary to suppressed renin as a result of excess DOC.

Treatment

Treatment strategy in this condition is similar to other forms of CAH in term of GC replacement therapy and stress dose (see chapter 8 Congenital Adrenal Hyperplasia). In addition to GC, sex hormone replacement that is appropriate to sex of rearing is indicated at a developmentally appropriate time to allow patients to resemble their peers. (See also treatment section in Chapter 11 – 46,XY Disorders of Sexual Development)

GLUCOCORTICOID REMEDIABLE ALDOSTERONISM

GRA, also known as familial hyperaldosteronism type I (FH I), was first described by Sutherland et al. in 1966 (19). It is an autosomal dominant form of low renin hypertension characterized by hyperaldosteronism. Aldosterone secretion is controlled by ACTH rather than angiotensin II, and for this reason, the unique distinguishing feature of GRA is the complete and rapid suppression of aldosterone by exogenous GC (dexamethasone) administration.

GRA produces a volume expansion, salt-sensitive form of low renin hypertension. Variable presentation is not uncommon; hypertension is invariably present, but hypokalemia and metabolic alkalosis may be absent. The disease is characterized by early onset of moderate to severe hypertension with hyperaldosteronism and low renin values and by high incidence of premature cerebrovascular events. Additionally, children demonstrate normal growth and development, which distinguishes this disorder from 11β-OHD and apparent mineralocorticoid excess (AME) The serum aldosterone is elevated and plasma renin activity is suppressed, but the aldosterone-renin ratio is typically not as high as with primary aldosteronism (PA) caused by an aldosterone-producing adenoma.

Circadian measurement of plasma steroids in GRA patients has not only revealed excessive production of aldosterone following ACTH stimulation, but excessive secretion of two normally rare steroids: 18-hydroxycortisol and 18-oxocortisol (20). This can be explained by the molecular genetic finding of a chimeric gene between CYP11B1 and CYP11B2--two genes that reside within a 30-kilobase stretch on chromosome 8 that results from an unequal crossing over during meiotic reduction. CYP11B1 encodes 11β-hydroxylase, the enzyme that catalyzes the last step in cortisol synthesis in the zona fasiculata; CYP11B2 encodes aldosterone synthase, the enzyme that catalyzes the last step in aldosterone synthesis in the zona glomerulosa. The product of this chimera thus carries aldosterone synthase enzymatic activity but is regulated by ACTH. Indeed, direct genetic screening for the presence of the chimeric gene can be performed by the long template PCR method with oligonucleotides specific for CYP11B1 and CYP11B2. This test is 100% sensitive and specific, has a relatively low cost, and is more rapid and reliable, compared to conventional dexamethasone suppression test (21). However, both dexamethasone administration and genetic testing are of importance in making the diagnosis.

Treatment

Children with GRA who are treated with GCs usually experience resolution of their hypertension within 2 weeks after initiation of therapy. The recommended doses are similar to CAH during childhood and adulthood (also see Chapter 8 – Congenital Adrenal Hyperplasia), because the aim is to suppress ACTH secretion. Hydrocortisone is preferred during childhood period when dexamethasone is used in adults. A low sodium diet is recommended to lower BP because of the salt-sensitive volume expansion; this will also minimize potassium wasting. Typically, potassium supplement is not required. Normalization of urinary hybrid steroid levels and abolition of ACTH-regulated aldosterone production is not a requisite for hypertension control and, if used as a treatment goal, may unnecessarily increase the risk of Cushingoid side effects (22). The response to GCs is variable in adults, often requiring additional use of antihypertensive medications, such as spironolactone, amiloride and triamterene. It has been shown that even in the absence of hypertension, aldosterone excess is associated with increased left ventricular wall thicknesses and reduced diastolic function, initial changes that lead to cardiovascular morbidities. This leads to the recommendation to treat normotensive subjects diagnosed with FH I (23).

APPARENT MINERALOCORTICOID EXCESS

AME is a rare inherited form of hypertension caused by 11 β-hydroxysteroid dehydrogenase type 2 (11 β-HSD) deficiency. The disorder was first described biochemically and hormonally in 1977 by New et al in a Native American girl with severe hypertension (24). The syndrome is caused by non functional mutations in HSD11B2 gene on chromosome16q22. More than 40 causative mutations have been described. (14) In the past 4 decades since the original description of the disease, published data only included less than 100 patients worldwide.

AME defined an important “pre-receptor” pathway in steroid hormone action and their specificities to the receptor. The exploration and elucidation of this disease opened a new area in receptor biology as a result of the demonstration that the specificity of the MR function depends on a metabolic enzyme (11ßHSD2) rather than the receptor itself (25,26). This enzyme functions to protect the MR by inactivating cortisol to its inactive metabolite cortisone, thereby enabling the mineralocorticoid aldosterone to occupy the MR in vivo (27,28). Aldosterone is not metabolized by 11ßHSD2 because it forms a C11–C18 hemi-ketal group in aqueous solution. The MR is non-selective in vitro and cannot distinguish between the glucocorticoid cortisol and its natural ligand, aldosterone (29,30). Therefore, lack of protection of the receptor owing to the enzyme defect allows cortisol, which has higher circulating levels than aldosterone, to bind to the MR and to act as a mineralocorticoid. Clinical manifestations of AME mimic those of excessive mineralocorticoid activity, but no elevation of known mineralocorticoids is present in the AME patients. Three metabolite ratios are calculated, each reflecting a different aspect of enzyme function: (1) tetrahydrocortisol (THF) + allo-THF/ tetrahydrocortisone (THE) (global function of HSD) (31) ; (2) allo-THF/THF ratio (defect in 5ß-reductase activity) (32,33) ; (3) urinary free cortisol (UFF)/urinary free cortisone (UFE) (kidney HSD function)(34). Originally AME was described through the plasma half-life of [11-3H] cortisol (which when metabolized by 11ß-HSD yields tritiated water and cortisone), which may more accurately reflect renal 11ß-HSD2 activity (35).

AME usually presents in early life with low birth weight and postnatal failure to thrive, hypertension, and persistent polyuria and polydipsia. The disorder is characterized by hypokalemic alkalosis, hyporeninemia and undetectable serum concentrations of aldosterone. End-organ damage secondary to hypertension is common, even at a young age. Thirteen out of

fourteen AME patients demonstrated damage of one or more organs (kidney, heart, retina or central nervous system) at the time of diagnosis. In addition, most had hypercalcuria with nephrocalcinosis (36).

Treatment

The treatment of AME is primarily directed at the correction of hypokalemia and hypertension. Cortisol acts as the offending MC in AME, hence blockage of its binding to the MR reverses excess mineralocortocoidism. Spironolactone, an MR receptor antagonist, is the medication of choice: it binds competitively and protects the receptors against any MC in excess. The required dose of spironolactone in AME patients may go up to 3-5 mg/kg/day (or more than 400 mg per day in adults), to control blood pressure and to normalize renin. A reduction in dietary sodium and supplemental potassium are beneficial. Potassium supplement varies among patient to patient, range from 3-8 mEq/Kg/day. Patients with nephrocalcinosis require additional thiazide diuretic. In order to reduce urinary calcium and control blood pressure in these patients, either chlorothiazide at the dose of 20 mg/Kg/day or hydrochlorothiazide at the dose of 2 mg/Kg/day is recommended. Follow-up studies of AME patients treated with spironolactone revealed significant improvement in clinical symptoms. These outcomes demonstrate the importance of early diagnosis and adequate treatment (26,36). Another approach utilizing dexamethasone at the dose of 1.5-2.0 mg/day to suppress cortisol secretion demonstrated variable results. Normalization of BP occurred in approximately 60% of cases (37). Dexamethasone does not correct the hypokalemia and hypertension in all patients, and long-term therapy has excessive GC adverse effects. The low effectiveness of this treatment is not surprising based on theoretical grounds: in vitro data suggests that putative physiologic ligands to non-selective MR in the kidney include dexamethasone, as well as cortisol and other MCs (29). Therefore administering dexamethasone to suppress cortisol secretion, which is already lowered in AME, may supply an additional MR ligand to aggravate MC excess.

Additional antihypertensive medications, such as thiazides or amiloride, may be required during disease progression. Cure of AME was reported in one patient after kidney transplantation due to the normal 11β-HSD2 activity of the transplanted kidney (38,39). Advances in enhancing or inhibiting11βHSD2 activity by some medications may provide novel treatments for AME (40).

Although AME is very rare, mild or intermediate phenotypes of AME patients may be linked to common human disorders via alteration in cortisol-cortisone shuttle. These include several forms of hypertension, kidney failure, inflammatory processes (cirrhosis and cardiac fibrosis), low birth weight/ fetal programming of adult diseases and lately, carcinogenesis.

PRIMARY ALDOSTERONISM

Primary aldosteronism (PA) is a group of disorders, originally described by J.W.Conn in 1954 (41), in which there is a non-suppressible secretion of aldosterone. The major presentations are hypertension and hypokalemia. However, hypokalemia does not occur in the majority of patients with primary aldosteronism, with the prevalence ranging from 9 to 37% in adults (42). Various symptoms associated with hypokalemia can be found, including muscle weakness with various types of paresthesias, tiredness, thirst, polyuria and nocturia.

PA occurs in greater than 10% of hypertensive adult patients (43). Although it is considered rare in children, the high prevalence in the general adult population suggests that the disease

may develop in the pediatric population prior to its presentation of hypertension and vascular damage in adulthood [4]. Moderate to severe hypertension that does not respond to medication(s), spontaneous or diuretic induced hypokalemia and the presence of adrenal mass provide clues to diagnosis (43).

The major causes of PA are aldosterone-producing adenomas (often small tumors of less than 2 centimeters in diameter), bilateral or unilateral adrenal hyperplasia and rarely adrenal carcinoma. Plasma aldosterone-renin ratio (ARR) may be used as an initial screening test and should be repeated if the results are not conclusive or are difficult to interpret. Established ARR cut-offs in adults range between 20 to 40 (43). Further testing through suppressing aldosterone by oral sodium loading, saline infusion, and/or a challenge with either fludrocortisone or captopril can be used for diagnosis confirmation; however cut-off values and interpretation have only been established in adults. Adrenal computed tomography scan or an MRI image are used as the imaging study to identify the mass. The treatment options include unilateral adrenalectomy for unilateral diseases found on adrenal vein sampling and a MR antagonist such as spironolactone or eplerenone. (see details in Chapter 23 – Aldosterone Excess in ADRENAL PHYSIOLOGY AND DISEASES section)

PHEOCHROMOCYTOMA

Pheochromocytomas are reported to account for hypertension in 1 to 2% of children (44). They are catecholamine-producing tumors that arise from the chromaffin cells of the adrenal medulla and the sympathetic ganglia and they present with signs and symptoms that are related to the action of catecholamines. (See Chapter 34 in Adrenal Physiology and Disease section). Although the peak incidence is in the third to fourth decades, 10% to 20% occur in children, with increased frequency in boys, and a median age at presentation between 9.5 and 12.5 years (45). Certain symptoms are reported as occurring more commonly in children than adults. These include sweating, visual disturbances, nausea, vomiting, loss of weight, polyuria and polydipsia (46). In comparison with adults in whom the hypertension is often paroxysmal, it is sustained in 70 to 90% of children (47). However, hypertension is not invariable and can be absent in up to 20% of children (48). Furthermore, many pheochromocytomas, especially associated with MEN 2 and VHL disease, can be clinically silent.

OTHER CAUSES OF CHILDHOOD HYPERTENSION

Liddle’s syndrome is a rare autosomal dominant disease described by Liddle et al. in 1963 (49) causing arterial hypertension. Mutations in SCNN1B and SCNN1G, the genes that mapped to chromosome 16p12, have been described in Liddle’s syndrome patients (14). The clinical and biochemical findings other than elevated blood pressure are: chronic hypokalemia, increased urinary potassium excretion in conjunction with sodium retention, suppressed renin activity, aldosterone and angiotensin II. These presentations are similar to AME, but in contrast, Liddle’s syndrome is an autosomal dominant disorder that does not show a favorable response to spironolactone. (21)

Another rare cause is familial hyperaldosteronism type II (FHII), the first cases described by Gordon et al. in 1991 in three families with a familial variety of PA (50). It is distinguished from type I (GRA) by the failure of dexamethasone’s suppression of aldosterone and no hybrid gene mutation. FH-II is more common than FH-I, but their clinical presentations are indistinguishable from other forms of PA. Patients with FH II are older than those with FH I, perhaps owing to diagnosis of FH I at a younger age, made possible by genetic testing. No significance in age, sex, biochemical parameters, or aldosterone and renin levels was seen between patients with FH II and those with apparently sporadic PA. (21) It has been described both in families and in sporadic cases worldwide, with a range in age starting at 14 years and equal gender distribution (51). Although the inheritance in many families appears to be autosomal dominant, in sporadic cases it is still uncertain. Surgical treatment in the case of unilateral adrenal mass and medical treatment with MR antagonists can be effective (21).

Acknowledgement:

The author would like to express a special gratitude to C. Joan Riesland, M.Ed., BSN, RN for her editorial work on this article.

 

REFERENCES

  1. Corvol P, Persu A, Gimenez-Roqueplo AP, Jeunemaitre X Seven lessons from two candidate genes in human essential hypertension: Angiotensinogen and Epithelial Sodium Channel. Hypertension 1999; 33:1324-31
  2. The fourth report on the diagnosis, evaluation, and treatment of high blood pressure in children and adolescents. Pediatrics 2004;114(2 Suppl 4th Report):555-76.
  3. Zachmann M, Tassinari D, Prader A 1983 Clinical and biochemical variability of congenital adrenal hyperplasia due to 11beta-hydroxylase deficiency. J Endocrinol Metab 56:222-229
  4. Curnow KM, Slutsker L, Vitek J, et al. 1993 Mutations in the CYP11B1 gene causing congenital adrenal hyperplasia and hypertension cluster in exons 6, 7, and 8. Proc Natl Acad Sci (USA) 90:4552-6
  5. Khemiri M, Ridane H, Bou YO, Matoussi N, Khaldi F 2006 [11 beta hydroxylase deficiency: a clinical study of seven cases]. Tunis Med 84:106-13
  6. al-Jurayyan NA 1995 Congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency in Saudi Arabia: clinical and biochemical characteristics. Acta Paediatr 84:651-4
  7. Rosler A, Leiberman E, Sack J 1982 Clinical variability of congenital adrenal hyperplaisa due to 11B-hydroxylase deficiency. Hormone Research 16:133
  8. Mimouni M, Kaufman H, Roitman A, Morag C, Sadan N 1985 Hypertension in a neonate with 11 beta-hydroxylase deficiency. Eur J Pediatr 143:231-3
  9. Rosler A, Leiberman E, Sack J 1982 Clinical variability of congenital adrenal hyperplaisa due to 11B-hydroxylase deficiency. Hormone Research 16:133
  10. Ezquieta B, Luzuriaga C 2004 Neonatal salt-wasting and 11 beta-hydroxylase deficiency in a child carrying a homozygous deletion hybrid CYP11B2 (aldosterone synthase)- CYP11B1 (11 beta-hydroxylase). Clin Genet 66:229-35
  11. Hague WM, Honour JW 1983 Malignant hypertension in congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. Clin Endocrinol (Oxf) 18:505-10
  12. Chabre O, Portrat-Doyen S, Chaffanjon P, et al. 2000 Bilateral laparoscopic adrenalectomy for congenital adrenal hyperplasia with severe hypertension, resulting from two novel mutations in splice donor sites of CYP11B1. J Clin Endocrinol Metab 85:4060-8
  13. Quintos JB, Vogiatzi MG, Harbison MD, New MI 2001 Growth hormone therapy alone or in combination with gonadotropin-releasing hormone analog therapy to improve the height deficit in children with congenital adrenal hyperplasia. J Clin Endocrinol Metab 86:1511-7
  14. Stenson et al (2009), The Human Gene Mutation Database

(HGMD®): 2008 Update. Genome Med 1(1):13

 

  1. Geley S, Kapelari K, Johrer K, et al. 1996 CYP11B1 mutations causing congenital adrenal hyperplasia due to 11 beta-hydroxylase deficiency. J Clin Endocrinol Metab 81:2896-901
  2. Cerame BI, Newfield RS, Pascoe L, et al. 1999 Prenatal diagnosis and treatment of 11beta-hydroxylase deficiency congenital adrenal hyperplasia resulting in normal female genitalia. J Clin Endocrinol Metab 84:3129-34
  3. Nakajin S, Shinoda M, Haniu M, Shively JE, Hall PF. C21 steroid side chain cleavage enzyme from porcine adrenal microsomes. Purification and characterization of the 17 alpha-hydroxylase/C17,20-lyase cytochrome P-450. J Biol Chem 1984;259(6):3971-6.
  4. Fan YS, Sasi R, Lee C, Winter JS, Waterman MR, Lin CC. Localization of the human CYP17 gene (cytochrome P450(17 alpha)) to 10q24.3 by fluorescence in situ hybridization and simultaneous chromosome banding. Genomics 1992;14(4):1110-1.
  5. Sutherland D, Ruse J, Laidlaw J. Hypertension, increased aldosterone secretion and low plasma renin activity relieved by dexamethasone. Can Med Assoc J 1966;95(22): p1109-19.
  6. Dluhy R, Lifton R. Glucocorticoid-remediable aldosteronism. J Clin Endocrinol Metab 1999;84(12): p4341-4.
  7. New MI, Geller DS, Fallo F, Wilson RC. Monogenic low renin hypertension. Trends Endocrinol Metab 2005;16(3):92-7.
  8. Stowasser M, Bachmann AW, Huggard PR, Rossetti TR, Gordon RD. Treatment of familial hyperaldosteronism type I: only partial suppression of adrenocorticotropin required to correct hypertension. J Clin Endocrinol Metab 2000;85(9):3313-8.
  9. Stowasser M, Sharman J, Leano R, Gordon RD, Ward G, Cowley D, et al. Evidence for abnormal left ventricular structure and function in normotensive individuals with familial hyperaldosteronism type I. J Clin Endocrinol Metab 2005;90(9):5070-6.
  10. New MI, Levine LS, Biglieri EG, Pareira J, Ulick S (1977) Evidence for an unidentified steroid in a child with apparent mineralocorticoid hypertension. J Clin Endocrinol Metab 44: 924-933
  11. New MI (1994) The prismatic case of apparent mineralocorticoid excess. J Clin Endocrinol Metab 79: 1-3
  12. Wilson RC, Nimkarn S, New MI (2001) Apparent mineralocorticoid excess. Trends Endocrinol Metab 12: 104-111
  13. Edwards C, Stewart P, Burt D, et al. (1988) Localisation of 11 beta-hydroxysteroid dehydrogenase--tissue specific protector of the mineralocorticoid receptor. Lancet 2: p986-989
  14. Funder J, Pearce P, Smith R, Smith A (1988) Mineralocorticoid action: target tissue specificity is enzyme, not receptor, mediated. Science 242: p583-585
  15. Krozowski ZS, Funder JW (1983) Renal mineralocorticoid receptors and hippocampal corticosterone-binding species have identical intrinsic steroid specificity. Proc Natl Acad Sci (USA) 80: 6056-6060
  16. Arriza JL, Weinberger C, Cerelli G, et al. (1987) Cloning of human mineralocorticoid receptor complementary DNA: structural and functional kinship with the glucocorticoid receptor. Science 237: 268-275
  17. Palermo M, Quinkler M, Stewart PM (2004) Apparent mineralocorticoid excess syndrome: an overview. Arq Bras Endocrinol Metabol 48: 687-696
  18. Monder C, Shackleton CH, Bradlow HL, et al. (1986) The syndrome of apparent mineralocorticoid excess: its association with 11 beta-dehydrogenase and 5 beta- reductase deficiency and some consequences for corticosteroid metabolism. J Clin Endocrinol Metab 63: 550-557
  19. Shackleton CH, Rodriguez J, Arteaga E, Lopez JM, Winter JS (1985) Congenital 11 beta-hydroxysteroid dehydrogenase deficiency associated with juvenile hypertension: corticosteroid metabolite profiles of four patients and their families. Clin Endocrinol (Oxf) 22: 701-712
  20. Palermo M, Shackleton CH, Mantero F, Stewart PM (1996) Urinary free cortisone and the assessment of 11 beta-hydroxysteroid dehydrogenase activity in man. Clin Endocrinol (Oxf) 45: 605-611
  21. Ulick S, Levine LS, Gunczler P, et al. (1979) A syndrome of apparent mineralocorticoid excess associated with defects in the peripheral metabolism of cortisol. J Clin Endocrinol Metab 49: 757-764
  22. Dave-Sharma S, Wilson RC, Harbison MD, Newfield R, Razzaghy-Azar M, Krozowski Z, et al. Extensive Personal Experience: Examination of genotype and phenotype relationships in 14 patients with apparent mineralocorticoid excess. J Clin Endo Metab 1998;83:2244-2254.
  23. Cushing's disease of the kidney. Lancet 1988;2(8618):1002.
  24. Palermo M, Cossu M, Shackleton CH. Cure of apparent mineralocorticoid excess by 
kidney transplantation [letter]. N Engl J Med 1998;339(24):1787-8.
  25.  Palermo M, Delitala G, Sorba G, Cossu M, Satta R, Tedde R, et al. Does kidney transplantation normalise cortisol metabolism in apparent mineralocorticoid excess syndrome? J Endocrinol Invest 2000;23(7):457-62
  26. Riddle MC, McDaniel PA. Acute reduction of renal 11 beta-hydroxysteroid 
dehydrogenase activity by several antinatriuretic stimuli. Metabolism 1993;42(10):1370-
  27. Conn JW, Louis LH (1956) Primary aldosteronism, a new clinical entity. Ann Intern Med 
44: 1-15
  28. Mulatero P, Stowasser M, Loh KC, et al. (2004) Increased diagnosis of primary 
aldosteronism, including surgically correctable forms, in centers from five continents. J 
Clin Endocrinol Metab 89: 1045-1050
  29. Funder JW, Carey RM, Fardella C, et al. (2008) Case detection, diagnosis, and 
treatment of patients with primary aldosteronism: an endocrine society clinical practice guideline. J Clin Endocrinol Metab 93: 3266-3281
  30. Dubois R, Chappuis J. [Pheochromocytoma: pediatric features]. Arch Pediatr 1997;4(12): p1217-25.
  31. Mircescu H, Wilkin F, Paquette J, Oligny LL, Decaluwe H, Gaboury L, et al. Molecular characterization of a pediatric pheochromocytoma with suspected bilateral disease. J Pediatr 2001;138(2):269-73.
  32. Fonseca V, Bouloux P. Phaeochromocytoma and paraganglioma. Baillieres Clin Endocrinol Metab 1993;7(2): p509-44.
  33. Ross J. Pheochromocytoma. Special considerations in children. Urol Clin North Am 2000;27(3): p393-402.
  34. Khafagi FA, Shapiro B, Fischer M, Sisson JC, Hutchinson R, Beierwaltes WH. Phaeochromocytoma and functioning paraganglioma in childhood and adolescence: role of iodine 131 metaiodobenzylguanidine. Eur J Nucl Med 1991;18(3):191-8.
  35. Liddle GW, Bledsoe T, Coppage WS, Jr. A familial renal disorder simulating primary aldosteronism but with negligible aldosterone secretion. Trans. Assoc. Am. Phys. 1963;76:199-213.
  36. Gordon R, Stowasser M, Tunny T, Klemm S, Finn W, Krek A. Clinical and pathological diversity of primary aldosteronism, including a new familial variety. Clin Exp Pharmacol Physiol 1991;18(5): p283-6.
  37. Stowasser M, Gunasekera TG, Gordon RD. Familial varieties of primary aldosteronism. Clin Exp Pharmacol Physiol 2001;28(12):1087-90.

Assay of Thyroid Hormones and Related Substances

 ABSTRACT

This chapter reviews how improvements in sensitivity and specificity of thyroid function tests [total and free thyroid hormones, TSH, thyroid autoantibodies (TRAb, TPOAb and TgAb) and thyroglobulin (Tg)] have dramatically improved clinical strategies for detecting and treating thyroid disorders. The review discusses the strengths and limitations of the different methodologies currently used (RIA, IMA and LC-MS/MS) and their propensity for analyte-specific interferences caused by heterogeneity (TSH, TgAb and Tg) or analyte-specific autoantibodies (T4Ab, T3Ab, TSHAb and TgAb). In addition, non-analyte related interferences from heterophile antibodies, including human anti-mouse antibodies (HAMA) and Rheumatoid Factor (RF), and interferences related to the use of Biotin and Streptavidin reagents, are discussed. The review provides an update on collaborations between the International Federation of Clinical Chemistry (IFCC) committee for the standardization of thyroid function tests (C-STFT) and the in-vitro diagnostic (IVD) industry- the goal being to eliminate between-method biases. Although re-standardization of thyroid hormone tests against established reference measurement procedures, and harmonization of TSH tests to the all-method mean has proved effective, recalibration has yet to be implemented by the IVD.  Until between-method biases are eliminated, it is not feasible to propose universal reference ranges that would apply across methods. The review contains a comprehensive discussion of the clinical utility of Tg methodology (RIA, IMA or LC-MS/MS), used to monitor patients with differentiated thyroid cancer (DTC). Mechanisms for in-vitro and possible in-vivo TgAb interference with Tg testing are proposed. The methodologic and clinical strengths and weakness of each test are discussed relative to current guidelines.For complete coverage of this and related areas in Endocrinolofy, visit our free web-books, www.endotext.org and www.thyroidmanager.org.

 

INTRODUCTION

 

Over the past forty years, improvements in the sensitivity and specificity of thyroid testing methodologies have dramatically impacted clinical strategies for detecting and treating thyroid disorders. In the 1950s, only one thyroid test was available - an indirect estimate of the serum total (free + protein-bound) thyroxine (T4) concentration, using the protein bound iodine (PBI) technique (1). Since 1970, technological advances in radioimmunoassay (RIA) (2-6), immunometric assay (IMA) (7-11) and most recently liquid chromatography-tandem mass spectrometry (LC-MS/MS) (12-23) have progressively improved the specificity, reproducibility and sensitivity of thyroid tests (24,25). Currently, serum-based immunoassays and LC-MS/MS techniques are available for measuring total and free thyroid hormones, [Thyroxine (T4) and Triiodothyronine (T3)] (23,26-28), as well as the pituitary thyroid stimulator, Thyrotropin (Thyroid Stimulating Hormone, TSH) (8,29) and the thyroid hormone precursor protein, Thyroglobulin (Tg) (9,16,21,30-33). In addition, measurements can be made of the thyroid hormone binding proteins, Thyroxine Binding Globulin (TBG), Transthyretin (TTR)/Prealbumin (TBPA) and Albumin (34-36). Methods to detect the thyroid autoantibodies (24,37): TSH receptor antibodies (TRAb) (38-43), thyroid peroxidase antibodies (TPOAb) and thyroglobulin antibodies (TgAb) (31,44,45) have been developed in response to the recognition that autoimmunity is a major cause of thyroid dysfunction (46-48). Currently, most thyroid testing is performed on serum specimens using manual or automated immunoassays employing specific antibody reagents targeting these ligands (22,24).
Over the last ten years the International Federation of Clinical Chemistry (IFCC) committee for the standardization of thyroid function tests (C-STFT)* has been working with manufacturers to identify and reduce between-method variability for total and free thyroid hormones as well as TSH (49). Reference measurement procedures (RMP) for TT4 and TT3 using primary calibrators have been developed (12,13,26) and used to establish isotope-dilution liquid chromatography/tandem mass spectrometry (ID-LC-MS/MS) as the RMP for FT4 and FT3 after isolating free hormone by equilibrium dialysis (26,27,50-53) or ultrafiltration (14,23,27,28,54,55). Thyroglobulin has also been detected by LC-MS/MS after trypsinization (16,19-21). Despite technical improvements, sensitivity, specificity and standardization issues still result in substantial between-method variability for many thyroid analytes (8,9,16,32,44,49,53,56-58). The C-STFT studies have shown that recalibrating thyroid hormone methods to their RMPs (50-53) and harmonizing TSH methods to the all-method mean, derived by a robust factor analysis model, significantly reduces between-method biases (29,52,53,59,60). It is hoped that the industry will shortly recalibrate their thyroid tests to remove current biases thereby allowing establishment of universal reference ranges that could apply to all methods and improve the clinical utility of thyroid testing. This chapter is designed to give an overview of the current status and limitations of the thyroid testing methods most commonly used in clinical practice, as recommended by current guidelines (24,61-74). Table 1

 

Table 1-Reference ranges for Thyroid Function Tests Used in USC Clinical Laboratory

 

Test Method USC Reference Ranges *
Total Thyroxine (TT4) Roche Cobas 57-159 nmol/L ( 4.5-12.5 ug/dL)
Total Triiodothyronine (T3) Roche Cobas 1.2-2.8 nmol/L (80-180 ng/dL)
Thyroid Hormone Binding Ratio (THBR) Roche Cobas 0.72 - 1.24 (unitless)
Thyrotropin (TSH) Roche Cobas 0.3 - 4.0 mIU/L
Thyroxine Binding Globulin (TBG) Siemens Immulite 14.0-31.0 mg/L (14.0-31.0 μg/mL)
Thyroid Peroxidase Antibody (TPOAb) Kronus/RSR <1.0 kIU/L
Thyroglobulin (2G-Tg-IMA) Beckman Access 3-40 μg/L (3-40 ng/mL) #
Thyroglobulin RIA (Tg-RIA) USC LDM [31] 3-40 μg/L (3-40 ng/mL) #
Tg Autoantibody (TgAb) Kronus/RSR <0.4 kIU/L

*These ranges are only applicable to the method listed.  They were established for a non-pregnant <60 year-old euthyroid cohort recruited by USC.

# Tg range should be adjusted for thyroid mass and TSH status [see below].

 

 

 

* My sincere thanks to the C-STFT committee chair Professor Linda Thienpont for informative discussions and for providing some of the data contained in this chapter.

TOTAL THYROID HORMONE MEASUREMENTS (TT4 AND TT3)

 

Thyroxine (T4) circulates 99.97% bound to the plasma proteins, primarily TBG (60-75%) but also Transthyretin TTR/TBPA (15-30%) and Albumin (~10%)(Table 2) .  In contrast, approximately 99.7% of Triiodothyronine (T3) is protein-bound, primarily to TBG [34,35,75]. Total (free + protein-bound) concentrations of thyroid hormones (TT4 and TT3) circulate at nanomolar concentrations and are considerably easier to measure than the free hormone moieties (FT4 and FT3) that circulate in the picomolar range. Serum TT4 measurement has evolved over the past four decades from the protein-bound iodine and competitive protein binding tests [1,76] to non-isotopic immunometric assays [77] and LC-MS/MS methods [13,78-80].

 

Table 2: Conditions that Influence Thyroid Hormone Binding Proteins


Serum TT4 measurement has evolved over the past four decades from the protein-bound iodine and competitive protein binding tests [1,76] to non-isotopic immunometric assays [77] and LC-MS/MS methods [13,78-80]. Total hormone methods require the inclusion of inhibitors, such as 8-anilino-1-napthalene-sulphonic acid, to block hormone binding to serum proteins in order to facilitate binding to the antibody reagent [81]. Methodology for TT4 measurement has changed over the decades and been paralleled by changes in TT3 methodology. However TT3 measurement presents a greater sensitivity and precision challenge, because TT3 concentrations are ten-fold lower than TT4 [13,82-86]. Most laboratories currently measure TT4 and TT3 concentrations by non-competitive immunometric assays performed on automated platforms using enzymes, fluorescence or chemiluminescent molecules as signals [25,75,87]. A recent IFCC C-STFT study compared eleven TT4 and twelve TT3 immunoassays marketed by eight diagnostic companies [80]. TT4 and TT3 measurements were made in sera from healthy individuals using the various immunoassays and compared with values reported by isotope dilution tandem mass spectrometry (ID-LC-MS/MS) - the reference measurement procedure (RMP) based on using primary T4 and T3 standards for calibration [80,88]. Although most methods fell short of the optimal 5 percent goal established by the C-STFT, 4/11 TT4 assays agreed within 10 percent of the reference, whereas most TT3 assays exhibited a positive bias that would necessitate re-standardization [80, 88] (Figure 1). Thus, as would be expected, TT4 assays are more reliable than TT3 although assay variability persists, likely as a result of matrix differences between calibrators and patient sera, the efficiency of the blocking agent employed by different manufacturers and lot-to-lot variability [53,56,89,90].

 

Figure 1- Between-method TT4 and TT3 Variability

 

 

Figure 1. (A), (TT4); (D) (TT3): assay means (1-sided 95% CIs) vs the mean by the RMPs.The x axis gives the codes of the different assays, the dotted lines represent the mean of the RMP _10%. For the assays differing >10% from the mean of the RMP, the numerical value of the mean is listed. (B), (TT4); (E), (TT3): scatter plot (x = mean of the RMP, y = mean of singlicate results per assay) with indication of the line of equality (dotted) and the most extreme Deming regression lines/equations. The results for the most deviating assays are indicated by circles and triangles; all other assays are indicated with the same symbol, X. (C), (TT4); (F), (TT3): percent-difference plot with indication of the strongest negatively (circles) and positively (triangles) biased assays. Note that (B), (C), (E), and (F) are extended to show the complete range (10–221 nmol/L for TT4, 0.6 –1.9 nmol/L for TT3) [80].

Clinical Utility of TT4 and TT3 Measurements

 

The diagnostic accuracy of total hormone measurements would be equivalent to that of free hormone tests if all patients had similar binding protein concentrations [35,75]. In fact, a recent study has reported that a screening cord blood TT4 < 7.6 μg/dL (< 98 nmol/L) can be used as a screening test for congenital hypothyroidism [91]. Unfortunately, many conditions are associated with TBG abnormalities that distort the relationship between total and free thyroid hormones (Table 1). Additionally, some patients have abnormal thyroid hormone binding albumins (dysalbuminemias) [92-94], thyroid hormone autoantibodies [95-98], or are taking drugs [25,99-101] that render total hormone measurements diagnostically unreliable [Table 1]. Consequently, TT4 and TT3 measurements are rarely used as stand-alone tests, but are typically employed in conjunction with a direct TBG measurement or an estimate of binding proteins [i.e. a thyroid hormone binding ratio test, THBR, that can be used to calculate a free hormone index (FT4I or FT3I). This index approach effectively corrects for the most common thyroid hormone binding protein abnormalities that distort total hormone measurements [ [102-104]. Because free hormone immunoassays are more technically challenging than total hormone measurements [49,86] total hormone tests can useful confirmatory when a free hormone immunoassay result appears questionable, especially in pregnancy and critical illness where changes in binding protein concentrations and affinity for thyroid hormones can occur [22,104-106]. Suboptimal FT3 assay sensitivity limits reliable FT3 measurements to the high (hyperthyroid) range [86]. However, since T3 is typically only a 3rd-line test of thyroid status used for diagnosing unusual cases of hyperthyroidism, TT3 measurement can usually suffice in preference to FT3, especially when TT3 is used as a ratio with TT4 to eliminate binding protein effects [107]. In fact, in Graves' hyperthyroidism preferential thyroidal T3 secretion resulting from increased deiodinase activity secondary to thyroidal stimulation by TSH receptor antibodies (TRAb) [108] such that a high serum TT3/TT4 or FT3/FT4 ratio that can be used to differentiate Graves' from other causes of hyperthyroidism [107,109,110].

 

TT4 and TT3 Reference Ranges

 

Total T4 reference ranges have approximated 58 to 160 nmol/L (4.5-12.5 µg/dL) for more than four decades, although some between-method differences and sample-related variability remains [80, 104]. The IFCC C-STFT found that most TT4 methods report values within 10 percent of the ID-LC-MS/MS RMP (Figure 1) [80]. In euthyroid pregnant subjects the major influence on TT4 is the TBG concentration that rises approximately two-fold by mid-gestation. As a consequence, TT4 steadily increases from the first trimester to plateau at approximately 1.5-fold pre-pregnancy levels by mid-gestation [104,106,111-114]. Thus the non-pregnant TT4 reference range, adjusted by a factor of 1.5 can be used to assess thyroid status in the latter half of gestation [66,67,104,106,115,116].

 

TT3 reference ranges generally approximate 1.2 - 2.7 nmol/L (80 –180 ng/dL) [84]. However, TT3 methods display far more between-method variability than TT4, and most display more than a 10 percent bias relative to the reference method [79,80,86]. The IFCC C-STFT continues to work with manufacturers to the reduce variability and improve the calibration of TT3 methods against the RMP.

 

Free Thyroid Hormone Tests (FT4 and FT3)

 

In accord with the free hormone hypothesis, it is the free fraction of the thyroid hormones (0.02% of TT4 and 0.2% of TT3) that exerts biologic activity at the cellular level [117], whereas protein-bound hormone is considered as biologically inactive. Since binding-protein abnormalities are highly prevalent (Table 1) [35], free hormone measurement is considered preferable to total hormone testing [22,118]. However, free hormone measurement that is independent of thyroid hormone binding proteins remains challenging [22,118-120]. Free hormone methods fall into two categories – direct methods, that employ a physical separation of the free from protein-bound hormone, and estimate tests, that either calculate a free hormone “index” from a measurement of total hormone corrected for binding proteins with either a TBG measurement or a binding-protein estimate, or immunoassays that employing an antibody to sequester a small amount of the total hormone that is purportedly proportional to the free hormone concentration [22,75,118]. All free hormone tests are subject to limitations. Both index tests (FT4I and FT3I) and FT4 and FT3 immunoassays are typically protein-dependent to some extent, and may under- or overestimate free hormone, when binding proteins are abnormal [52,92,118-128]. Even direct methods that employ equilibrium dialysis or ultrafiltration to separate free from protein-bound hormone are not immune from technical problems relating to dilution, adsorption, membrane defects, temperature, the influence of endogenous binding protein inhibitors, fatty acid formation and sample-related effects [22,128-133]. The IFCC C-STFT has now established a reference measurement procedure (RMP) for free thyroid hormones that is based on equilibrium dialysis-dilution-mass spectrometry (ED-ID-MS) and primary calibrators [15,51,54,134]. An evaluation of current FT4 immunoassays has revealed major between-method variability and significant biases relative to the RMP that are far in excess of FT4 biological variation [50,53]. Recalibrating methods against the RMP was shown to significantly reduce biases that currently preclude implementing universal reference intervals that would apply across methods. The C-STFT is actively working with the in vitro diagnostic industry to re-standardize free hormone methods against the RMP to reduce current biases.

 

Direct FT4 and FT3 Methods

 

Direct free hormone methods have employed equilibrium dialysis [51,54,135-137], ultrafiltration [14,17,18,23,131,138-142] or gel filtration [143] to separate free hormone from the dominant protein-bound moiety. These separation techniques can be prone to inaccuracies causing under- or overestimate of free hormone due factors relating to dilution, adsorption, membrane defects, temperature, pH, the influence of endogenous binding protein inhibitors, fatty acid formation and sample-related effects [22,118,128,130-133,141,142,144-146]. The IFCC C-STFT has now established the RMP for FT4 as ED ID-LC-MS/MS. Specifically, equilibrium dialysis of serum is performed under defined conditions before FT4 is measured in the dialysate by isotope-dilution-liquid chromatography/tandem mass spectrometry [15,51,54]. Manufacturers are recommended to use this RMP to recalibrate their FT4 immunoassay tests  [52-54,134]. Because direct free hormone methods are technically demanding, inconvenient and expensive, they are typically only readily available in reference laboratories. Most FT4 and FT3 testing is made using estimate tests - either the two-test “index” approach or an immunoassay “sequestration” method [118]. However, all current FT4 and FT3 estimate tests are binding-protein dependent to some extent [118,147-150], and a direct free hormone test can be especially useful for evaluating thyroid status when immunoassay values appear discordant with the clinical presentation and/or the TSH measurement [22].

 

Equilibrium Dialysis

 

Early equilibrium dialysis methods used I-131 and later I-125 labeled T4 tracers to measure the free T4 fraction, that when multiplied by a total hormone measurement gave an estimate of the free hormone concentration [135]. Subsequently, symmetric dialysis in which serum was dialyzed without dilution (or employing a near-physiologic medium) was used to overcome dilution effects [132]. By the early 1970s higher affinity T4 antibodies (>1x1011 L/mol) and high specific activity T4-I125 tracers were used to develop sensitive RIA methods that could to directly measure FT4 and FT3 in dialyzates and ultrafiltrates [82,136-138,142,151-154]. Subsequent improvements have involved employing more physiologic buffer diluents and improving the dialysis cell design [132,137]. More recently, isotope-dilution liquid chromatography/tandem mass spectrometry (ID-LC-MS/MS) [155] has been used to measure FT4 in ultrafiltrates [14,156,157] and dialyzates [27,50,51,134]. The FT4 RMP recently established by the IFCC C-STFT is based on ED followed by ID-LC-MS/MS [15,51].

 

Figure 2. FT4 and FT3 Immunoassay Method Comparison

Figure 2. Between Assay Comparison of FT4 and FT3 Measurements in Healthy Euthyroid Subjects. A=FT4 and D=DT3: assay means versus the mean by the RMPs. Different assays are coded A-O on the x axis, manufacturer codes used to designate assays were different for FT4 and FT3 assays. The dotted lines represent mean +/- 10% of the RMP ED-ID-MS). B=FT4 and E=FT3: scatter plot (x=mean of the RMP vs. y= mean of 6 singlicate results per assay. Line of equality indicated by dotted line. The results for the most deviating assays are indicated by circles and triangles; all other assays are indicated with the same symbol, X.  C=FT4 and F=FT3: percent-difference plot indicating the strongest negatively (circles) and positively (triangles) biased assays [50].

 

 

Ultrafiltration Methods

 

A number of studies have used ultrafiltration to remove protein-bound T4 prior to LC-MS/MS measurement of FT4 in the ultrafiltrate [14,17,18,23,55,131,138-142]. Direct FT4 measurements employing ultrafiltration are sometimes higher than those made by equilibrium dialysis, because ultrafiltration avoids dilution effects [140]. Furthermore, ultrafiltration is not influenced by dialyzable inhibitors of T4-protein binding that can be present in conditions such as non-thyroidal illness (NTI) [130]. However, ultrafiltration can be prone to errors when there is a failure to completely exclude protein-bound hormone and/or adsorption of hormone onto the filters, glassware and tubing [127]. In addition, ultrafiltration is temperature sensitive and ultrafiltration performed at ambient temperature (25°C) will report FT4 results that are 67 percent lower than ultrafiltration performed at 37°C [133,158]. However, FT4 concentrations measured by ID-LC-MS/MS following either ultrafiltration at 37°C or equilibrium dialysis usually correlate [159].

 

Gel Absorption Methods.

 

Some early direct FT4 methods used Sephadex LH-20 columns to separate free from bound hormone before eluting the free T4 from the column for measurement by a sensitive RIA. However, because of a variety of technical issues, assays based on this methodologic approach are not currently used [75].

 

 

Indirect FT4 and FT3 Estimate Tests

 

Two-Test Index Methods (FT4I and FT3I)

Free hormone indexes (FT4I and FT3I) are unitless mathematical calculations made by correcting the total hormone test result for the binding protein, primarily TBG, concentration. These indexes require two separate tests and have been used to estimate free hormone concentrations for more than 40 years [118]. The first test involves the measurement of total hormone  (TT4 or TT3) ,whereas the second test assesses the binding protein concentration using either (i) a direct TBG immunoassay, (ii) a Thyroid Hormone Binding Ratio (THBR) or “Uptake” test or (iii) an isotopic determination of the free hormone fraction [118,160].

 

TBG Immunoassays

There is conflicting data concerning whether indexes employing THBR in preference to direct TBG are diagnostically superior [161]. Free hormone indexes calculated using direct TBG measurement (TT4/TBG) may offer improved diagnostic accuracy over THBR when the total hormone concentration is abnormally high (i.e. hyperthyroidism), or when drug therapies interfere with THBR tests [101,162-165]. Regardless, the TT4/TBG index is not totally independent of the TBG concentration, nor does it correct for Albumin or Transthyretin binding protein abnormalities (Table 1) [120].

 

Thyroid Hormone Binding Ratio (THBR) / "Uptake" Tests
The first "T3 uptake" tests developed in the 1950s employed the partitioning of T3-I131 tracer between the plasma proteins in the specimen and an inert scavenger (red cell membranes, talc, charcoal, ion-exchange resin or antibody) [119,166,167]. The "uptake" of T3 tracer onto the scavenger provided an indirect, reciprocal estimate of the TBG concentration of the specimen. Initially, T3 uptake tests were reported as percent uptakes (free/total tracer). Typically, sera with normal TBG concentrations had approximately 30 percent of the T3 tracer taken up by the scavenger. During the 1970s methods were refined by replacing I131-T3 tracers by I125-T3, calculating uptakes based on the ratio between absorbent and total minus absorbent counts, and expressing results expressed as a ratio with normal sera having an assigned value of 1.00 [160,167]. Historically, the use of T3 as opposed to T4 tracer was made for practical reasons relating to the ten-fold lower the affinity of TBG for T3 versus T4, facilitating a higher percentage of T3 tracer being taken up by the scavenger and allowing lower isotopic counting times.  Because current methods use non-isotopic proprietary T4 or T3 "analogs", counting time is no longer an issue and current tests may use a "T4 uptake" approach - which may be more appropriate for correcting for T4-binding protein effects. Differences between T3 and T4 "uptakes" have not been extensively studied [168]. Although all THBR tests are to some degree TBG dependent, the calculated FT4I and FT3I usually provides an adequate correction for mild TBG abnormalities (i.e. pregnancy and estrogen therapy) [104,122,169-171], although they may fail to correct for grossly abnormal binding proteins [94] in euthyroid patients with congenital TBG extremes [120,122,172], Familial Dysalbuminemic Hyperthyroxinemia (FDH) [75,92,173-176], thyroid hormone autoantibodies [95,97,177,178], non-thyroidal illness (NTI) [120,128,179,180] or medications that directly or indirectly influence thyroid hormone binding to plasma proteins [75,99,120,164,181,182].

 

Isotopic Index Methods

The first free hormone tests developed in the 1960s were indexes calculated from the product of the free hormone fraction, measured isotopically by dialysis, and TT4 measured by PBI and later RIA [135,183,184]. These early isotopic detection systems were technically demanding and included paper chromatography, electrophoresis, magnesium chloride precipitation and column chromatography [135,153,185-187]. The free fraction index approach was later extended to ultrafiltration and symmetric dialysis, the latter measuring the rate of transfer of isotopically-labeled hormone across a membrane separating two chambers containing the same undiluted specimen [92,138,140,184,188-190]. Ultrafiltration and symmetric dialysis had the advantage of eliminating dilution effects that influenced tracer dialysis values [129,191]. However, free hormone indexes calculated using an isotopic free fraction were not completely independent of the TBG concentration and furthermore were influenced by tracer purity and the buffer matrix employed [137,192].

 

Clinical Utility of Two-Test Index Methods (FT4I and FT3I

 

Some favored the two-test FT4I approach for evaluating the thyroid status of patients with abnormal binding protein states like pregnancy and NTI [104,193]. Continued use of the FT4I remains controversial [194]. However, until FT4 immunoassays are re-standardized to remove biases [50,52,53], FT4I remains a useful confirmatory test when binding proteins are abnormal and when diagnosing central hypothyroidism [195].

 

Free Thyroid Hormone Immunoassay Methods (FT4 and FT3)

 

Most free hormone testing is made using FT4 and FT3 immunoassays [87]. These immunoassays are based on "one-step",  "labeled antibody" or "two-step" principles, as described below [75,118,196]. For more than twenty years controversy has surrounded the standardization and diagnostic accuracy of these methods, especially in pathophysiologic conditions associated with the binding protein abnormalities such as pregnancy [22,104], or due to polymorphisms, drug interactions, high free fatty acid (FFA) levels or thyroid binding inhibitors such as those present in NTI [25,53,75,92,119,120, 126-128,130,147,150,196-200]. Studies showing correlations between FT4 immunoassay values and both TBG and albumin concentrations, as well as weak inverse FT4/TSH log/linear relationships [17,18,23,126], have emphasized the need to evaluate each method with clinical specimens containing abnormal binding proteins. Currently, most FT4 and FT3 immunoassays display significant negative or positive biases that exceed the intra-individual biological variability (Figure 2) [50,52,53]. The IFCC C-STFT is actively working with the IVD industry to recalibrate their free hormone immunoassays against the RMP [15,50,53,60]. However, although recalibration to the RMP has been shown to greatly reduce between-method biases [50,52,53], implementation of a global re-calibration effort has been delayed by practical, educational and regulatory complexity.

 

 

One-Step, FT4 and FT3 Methods

The “one-step” approach uses a proprietary labeled hormone analog, designed for minimal interaction with thyroid hormone binding proteins, that competes with hormone in the specimen for a solid-phase anti-hormone antibody in a classic competitive immunoassay format [22,75,118,119,201,202]. After washing away unbound constituents, the free hormone concentration should be inversely proportional to the labeled analog bound to the solid support. Although conceptually attractive, the diagnostic utility of the one-step approach has been shown to be critically dependent on the degree that the analog is "inert" with respect to binding protein abnormalities [17,18,23,118,119,147,180,200,203-208].

 

Labeled Antibody FT4 and FT3 Methods

Labeled antibody methods are "one-step" methods that use labeled-antibody in preference to a labeled hormone analog. The free hormone in the specimen competes with solid-phase hormone for the labeled antibody and is quantified as a function of the fractional occupancy of hormone-antibody binding sites in the reaction mixture [22,75,118,120,202,209]. The labeled antibody approach is used as the basis for a number of automated immunoassay platforms because it is easy to automate and considered less binding-protein dependent than the labeled analog approach, because the solid phase hormone does not compete with endogenous free hormone for hormone binding proteins [22,87,118,210,211].

 

Two-Step, Back-Titration FT4 and FT3 Methods

The two-step approach was first developed by Ekins and colleagues in the late 1970s [75,119,128,202]. Two-step methods typically employ immobilized T4 or T3 antibody (for FT4 and FT3 immunoassays, respectively) to sequester a small proportion of total hormone from a diluted serum specimen without disturbing the original free to protein-bound equilibrium [75,118]. After removing unbound serum constituents by washing, a labeled probe (125-I T4, or more recently a macromolecular T4 conjugate) is added to quantify unoccupied antibody-binding sites that are inversely related to the free hormone concentration - a procedure that has been referred to as "back-titration [118].

 

Clinical Utility of FT4 and FT3 Measurements

 

Most FT4 methods give diagnostically reliable results when binding proteins are near-normal, provided that a method-specific reference range is employed [53]. However, both TT3 and FT3 immunoassay methods tend to be inaccurate in the low range [86,212] and have no value for diagnosing or monitoring treatment for hypothyroidism [70,213], although T3 measurement can be useful for diagnosing or confirming unusual cases of hyperthyroidism.

 

Ambulatory Patients

 

Free hormone tests (FT4 or FT3) are used in preference to total hormone (TT4 or TT3) measurements in order to improve diagnostic accuracy for detecting hypo- and hyperthyroidism in patients with abnormal thyroid hormone binding proteins (Table 1). FT4 is typically employed as a second-line test for confirming primary thyroid dysfunction detected by an abnormal TSH ,but is the first-line test when thyroid status is unstable (early phase of treating hypo- or hyperthyroidism), in the presence of pituitary/hypothalamic disease when TSH is unreliable, or when patients are taking drugs such as dopamine or glucocorticoids that are known to affect TSH secretion [24,100,101,165,214-219].

 

Mild "subclinical" thyroid dysfunction is characterized by TSH/FT4 discordances (abnormal TSH/normal FT4). This reflects the intrinsic complex nature of the inverse log/linear TSH/FT4 relationship [24,220,226] - a relationship that is modified by age and gender [227,228]. Thus, small changes in FT4, even within normal limits, are expected to produce a mild degree of TSH abnormality - between 0.05 and 0.3 mIU/L (for subclinical hyperthyroidism) and 5 and 10 mIU/L (for subclinical hypothyroidism). An unexpected TSH/FT4 discordance, if confirmed, should prompt an investigation for interference with FT4, TSH or both tests [229,230]. FT4 interference can result from severe binding protein abnormalities such as congenital TBG excess or deficiency [75,94,122,159,231,232], dysalbuminemias [92,233-236], thyroid hormone autoantibodies [95,97,98,177,178,230,237] or drug interferences [75,99,120].

 

Hospitalized Patients with Nonthyroidal Illnesses (NTI)

 

The diagnostic performance of current FT4 methods has not been evaluated in hospitalized patients with NTI where binding protein inhibitors and drug therapies can negatively impact the reliability of both thyroid hormone and TSH testing [24,75,126,130,180,218,238,239]. Three categories of hospitalized patients deserve special attention: a) patients with NTI without known thyroid dysfunction who have a high or low T4 status; b) patients with primary hypothyroidism and concurrent NTI and, c) patients with hyperthyroidism and concurrent NTI [238,240,241]. Because the diagnostic reliability of FT4 testing is still questionable in sick hospitalized patients, a combination of both T4 (FT4 or TT4) and TSH may be needed to assess thyroid status in this setting [24,53,180,242]. In most clinical situations where FT4 and TSH results are discordant, the TSH test is the most diagnostically reliable, provided that the patient does not have pituitary failure or is receiving medications such as glucocorticoids and dopamine that directly inhibit TSH secretion [101,165,218]. Repetitive TSH testing may be helpful in resolving the cause of an abnormal FT4, because the TSH abnormalities of NTI are typically transient whereas the TSH abnormality will persist if due to underlying thyroid dysfunction [243-246]. It may be useful to test for TPOAb as a marker for underlying thyroid autoimmunity

 

 FT4 and FT3 reference ranges

 

Current reference ranges for FT4 and FT3 immunoassays are method-dependent because of calibration biases [50,52,53] (Figure 2). This calibration problem negatively impacts the clinical utility of FT3 and FT4 tests because it precludes establishing universal reference ranges that would apply across methods.

Pediatric FT4 and FT3 Reference Ranges

 

The determination of normal reference limits for pediatric age-groups is especially challenging, given the limited number of studies involving sufficient numbers of healthy children [247-249]. Most studies report that serum TSH peaks after birth and steadily declines throughout childhood to reach adult levels at puberty. Likewise, FT3 declines across the pediatric age groups during childhood and approaches the adult range at puberty, whereas FT4 levels for infants less than a year old are higher than for children 1 to 18 years old who have FT4 similar to that observed for adults [247-252].

 

Pregnancy FT4 Reference Ranges

 

As with non-pregnant patients, TSH is the first-line test to use for assessing thyroid status during pregnancy [253]. However, FT4 measurement is needed for monitoring anti-thyroid drug treatment of hyperthyroid pregnant patients who have undetectable TSH. The question whether an isolated low FT4 during pregnancy is a maternal or fetal risk factor, remains controversial [254-259]. However, a number of studies suggest that low FT4 may be a risk factor for gestational diabetes and fetal complications [260-264].  Non-pregnant FT4 reference ranges do not apply to pregnancy since FT4 progressively declines as gestation progresses, necessitating the use of trimester-specific reference ranges [104,113,265-271]. Currently it is not possible to propose universal trimester-specific FT4 reference ranges given current between-method differences [50,53,271] (Figure 2) compounded by differences related to the ethnicity [193,270,272-275], iodine intake [276-278], smoking [279] and BMI [269,270,280-283] between study cohorts. Establishing institution-specific trimester-specific reference ranges from the 2.5 to 97.5 percentiles of least 400 pregnant patients from each trimester [270] is not practical for most institutions. The feasibility of establishing universal trimester-specific reference ranges will improve after the proposed re-standardization of FT4 methods against the RMP [53]. However, binding protein effects will remain and population-specific factors will still have to be considered.

 

Interferences with Total and Free Thyroid Hormone Tests

 

Only the physician can suspect interference with a test result and request that the laboratory perform interference checks! This is because the hallmark of interference is discordance between the test result and the clinical presentation of the patient. Failure to recognize interferences can have adverse clinical consequences [229,284-289].

 

Laboratory checks for interference include showing discordance between different manufacturers methods [290-293], re-measurement of analyte after adding blocking agents [293-297] and performing linearity studies or precipitating immunoglobulin with polyethylene glycol (PEG) [229,290,291,293,294,298-300]. A change in analyte concentration in response to one of these maneuvers suggests interference, but a lack of effect does not rule out interference. Interferences can be classified as either (a) non-analyte-specific or (b) analyte-specific [301,302].

 

Non-Analyte-Specific Interferences

 

Protein Interferences

Immunoassays can be affected by interferences from both paraproteins [303-305] and abnormal immunoglobulins [306,307].

 

 Congenital TBG excess or deficiency.

Free hormone immunoassays and free T4 index tests may be susceptible to interference from grossly abnormal TBG concentrations, such as those seen in congenital TBG excess or deficiency states [75,94,122,159,231,232].

 

 Pregnancy.

 Estrogen stimulation causes TBG concentrations to progressively rise to plateau 2.5-fold higher than pre-pregnancy values by mid-gestation [193,308,309]. As a consequence, both TT4 and TT3 increase to approximately 1.5-fold of pre-pregnancy values by mid-gestation [113,310]. Despite the rise in total hormone, both FT4 and FT3 decline to a method-related degree during gestation [104,265-269]. It should be noted that lower FT4 levels would be expected during pregnancy from a consideration of the law of mass action as applied to T4-binding protein interactions [310]. However, the degree of FT4 decline during pregnancy is variable and method-dependent due to standardization differences (Figure 2) and in some cases method sensitivity to the declining albumin concentrations typical of late gestation [18,193,311].

 

Familial Dysalbuminemic and Transthyretin-Associated Hyperthyroxinemias.

Autosomal dominant mutations in the Albumin or Transthyretin (prealbumin) [312] gene can result in altered protein structures with enhanced affinity for thyroxine and/or triiodothyronine. These abnormal proteins can interfere with FT4 and/or FT3 measurements and result in inappropriately high FT4 and/or FT3 immunoassay values [92,173,237,312]. Familial Dysalbuminemic Hyperthyroxinemia (FDH) is a rare condition with a prevalence of ~1.8 % in the Hispanic population [313]. It arises from a number of genetic variants, with the R218H being the most common, some variants result in extremely high TT4, whereas other mutations (i.e. L66P) affect mainly T3 [233]. Affected individuals are euthyroid and have normal TSH and FT4 when measured by direct techniques such as equilibrium dialysis [92]. Unfortunately, most FT4 estimate tests (immunoassays and indexes) report falsely high values for FDH patients that may prompt inappropriate treatment for presumed hyperthyroidism if the condition is not recognized [92].

 

Heterophile Antibodies (HAbs)

Heterophile antibodies (HAb) are human poly-specific antibodies targeted against animal antigens, the most common being human anti-mouse antibodies (HAMA) [293,302,314,315]. Alternatively, HAb can target human antigens [302] such as rheumatoid factor (RF), an immunoglobulin commonly associated with autoimmune conditions that is widely considered a heterophile antibody [316]. RF has been shown to interfere with free and total thyroid hormone tests [87] as well as TSH [317] and Tg [318]. HAbs have a prevalence of 30-40 percent [319-321] and have the potential to interfere with a broad range of methods that use IMA principles [290,300,306,322]. In recent years assay manufacturers have increased the immunoglobulin blocker reagents added to their tests and this has reduced interference from 2 to 5 percent [290,297,323]. However, interference is still seen in approximately one percent of patients who have high enough HAb concentrations to overcome the assay blocker [296,298,322,324]. HAMA interference mostly affects non-competitive immunometric assays (IMA) that employ monoclonal antibodies of murine origin [325]. Assays based on the competitive format that employ high affinity anti-antigen polyclonal antibody reagents, are rarely affected [296,319]. HAb has the potential to interfere with both free [178,321,326-328] and total [178,326,327] thyroid hormone tests, as well as THBR [327], TSH [289,294,300,328-330] and Thyroglobulin (Tg) [295,296,323,324,331,332], TgAb [333] and calcitonin (CT)  [300,334-337] methods. Interference from HAb or HAMA typically causes falsely high results for one or more analytes. Less commonly falsely low test results may be seen [332]. The test marketed by one manufacturer can be severely affected, whereas the test from a different manufacturer may appear unaffected. This is why the first step for investigating for interference is re-measurement of the analyte in a different manufacturers method. It should be noted that patients receiving recent vaccines, blood transfusions or monoclonal antibodies (given for treatment or scintigraphy), as well as veterinarians and those coming into contact with animals, are especially prone to test interferences caused by induced HAb and HAMA [298,338].

 

Anti-Reagent Antibodies

Interference can be caused by antibodies against assay reagents. For example, there are a number of reports of anti-Rhuthenium antibodies interfering with TSH, FT4 and FT3 by  [339-343].  In Streptavidin-Biotin based assays interference can result from antibodies targeting either Streptavidin [344] or biotin reagents [345]. Alternatively, high dose biotin ingestion has been known to produce interference with thyroid and other tests in an analyte-specific, platform-specific manner [346-350].

 

Analyte-Specific Interferences

 

Analyte-specific interferences typically result from autoantibodies targeting the analyte. Depending on the analyte and test formulation, autoantibody interferences typically cause falsely-high test results, but can cause falsely-low test results, as in the case of Tg autoantibodies . It should be noted that transplacental passage both heterophile antibodies or anti-analyte autoantibodies (i.e. TSHAb or T4Ab) have the potential to interfere with neonatal screening tests [351-354]. Specifically, maternal TSH autoantibodies can cross the placenta and may cause a falsely high TSH screening test in the newborn mimicking congenital hypothyroidism, whereas maternal T4 autoantibodies could cause falsely high neonatal T4 masking the presence of congenital hypothyroidism [230,353].

 

T4 and T3 Autoantibodies (T4Ab/T3Ab)

T4 and T3 autoantibodies can falsely elevate total hormone, free hormone or THBR measurements depending on the method employed [95,97,98,177,178,230,237]. The prevalence of thyroid hormone autoantibodies approximates 2 percent in the general population but may be as much as 30 percent in patients with autoimmune thyroid disease or other autoimmune conditions [316,355-358]. However, despite their high prevalence, significant interference caused by thyroid autoantibodies is not common and depends on the qualitative characteristics of the autoantibody present (i.e. its affinity for the test reagents). Further, different methods exhibit such interferences to a greater or lesser degree [95,97]. Because autoantibody interference is difficult for the laboratory to detect proactively, it is the physician who should first suspect interference characterized by unexpected discordance between the clinical presentation of the patient and the test result(s) [96, 178].

 

SERUM TSH (THYROID STIMULATING HORMONE/THYROTROPIN) MEASUREMENT

 

Over the last four decades the dramatic improvements in TSH assay sensitivity and specificity have revolutionized thyroid testing and firmly established TSH as the first-line test for ambulatory patients not receiving drugs known to alter TSH secretion [24,70,71,120,216,218,359]. Serum TSH has become the therapeutic target for levothyroxine (L-T4) replacement therapy for hypothyroidism and suppression therapy for differentiated thyroid cancer [72]. The diagnostic superiority of TSH versus FT4 measurement arises from the inverse, predominantly log/linear, TSH/FT4 relationship, that is modified to some extent by factors such as age, sex, active smoking and TPOAb status [7,24,221-228].

 

TSH Assays

 

TSH assay "quality" has historically been defined by clinical sensitivity – the ability to discriminate between hyperthyroid and euthyroid TSH values [24,360-364]. The first generation of RIA methods had a detection limit approximating 1.0 mIU/L [365-367] that limited their clinical utility to diagnosing primary hypothyroidism [368-370] and necessitated the use of TRH stimulation to diagnose hyperthyroidism that was characterized by an absent TRH-stimulated TSH response [371-376]. With the advent of immunometric assay (IMA) methodology that uses a combination of poly- and/or monoclonal antibodies targeting different TSH epitope(s) in a "sandwich" format [377-379], a ten-fold improvement in TSH assay sensitivity (~ 0.1 mIU/L) was achieved when using isotopic (I125) signals [380]. This level of sensitivity facilitated the determination of the lower TSH reference limit (as 0.3-0.4 mIU/L), and the detection of overt hyperthyroidism without the need for TRH stimulation [7,374-376,380-386], but was still insufficient for distinguishing between differing degrees of hyperthyroidism (i.e. subclinical versus overt). Sensitization continued until a third-generation of TSH IMAs, using non-isotopic signals, were developed that could achieve a sensitivity of 0.01 mIU/L [7,8,374,387-389]. Initially different non-isotopic signals were used that gave rise to a lexicon of terminology to distinguish between assays: immunoenzymometric assays (IEMA) used enzyme signals; immunofluorometric assays (IFMA) used fluorophors as signals, immunochemiluminometric assays (ICMA) used chemiluminescent molecules as signals and immunobioluminometric assays (IBMA) used bioluminescent signal molecules [8,390]. Current TSH methods are automated ICMAs [87] that all achieve third-generation functional sensitivity (FS = ≤0.01 mIU/L) - a sensitivity the FS level that has subsequently become the standard of care [7,8,52,53,388,391-396].

 

Functional Sensitivity (FS) - determines the lowest reportable assay limit

 

During the period of active TSH assay improvement, different non-isotopic IMAs made competing claims for sensitivity. Methods were described as: "sensitive", "highly sensitive", "ultrasensitive" or "supersensitive" - marketing terms that had no scientific definition. This confusion led to a debate concerning what was the most clinically relevant parameter to use to determine the lowest reliable reportable TSH value for clinical practice [8,397-403]. Functional sensitivity (FS), defined as the lowest analyte concentration measured with 20 percent coefficient of variation [24] is now recognized as the parameter that best represents the between-run precision for measuring low analyte concentrations in clinical practice [24,395,404]. FS is used to define the lower clinical reporting limit for not only for TSH assays, but also Tg and TgAb measurements, for which assay sensitivity is critical [8,24,397,404,405]. Protocols used for establishing FS specify that precision be determined in human serum, not quality control materials based on artificial protein matrices, since immunoassays tend to be matrix-sensitive [406,407]. The time-span used for determining precision is also analyte-specific and should reflect the frequency of testing employed in clinical practice  - 6 to 8 weeks for TSH, but 6 to 12 months for the Tg and TgAb assays when used as tumor markers for monitoring differentiated thyroid cancer (DTC). This time-span is important because low-end, between-run assay precision erodes over time as a result of a myriad of variables, reagent lot-to-lot variability being a key variable [9,408-410].  Note that the FS parameter is more stringent than other biochemical sensitivity parameters such as limit of detection (LOD - a within-run parameter) and limit of quantitation (LOQ - a between-run parameter without stipulations regarding matrix and time-span for determining precision) [404,411]. A ten-fold difference in FS has been used to define each more sensitive "generation" of TSH [397] or Tg [32,404,412,413] method. Thus, TSH RIA methods with FS approximating 1.0 mIU/L were designated "first generation", TSH IMA methods with functional sensitivity approximating 0.1 mIU/L were designated "second generation", and TSH IMAs with FS approximating 0.01 mIU/L are designated "third generation" assays [8,57,395,397,405,414]. Analogous to TSH, Tg assays [Section 6A] with FS approximating 1 μg/L are designated "first generation", whereas Tg IMAs with FS approximating 0.10 μg/L meet the criteria for a "second generation" method [32,58,296,395,404,413,415,416].

 

TSH Biologic Variability

As compared with between-person variability, TSH intra-individual variability is relatively narrow (20-25 percent) in both non-pregnant and pregnant subjects, as compared with between-person variability [29,222,417,418]. In fact, the serum TSH of euthyroid volunteers was found to vary only ~0.5 mIU/L when tested every month over a span of one year [417]. Twin studies suggest that there are genetic factors that determine hypothalamic-pituitary-thyroid setpoints [419-421]. These studies report that the inheritable contribution to the serum TSH level approximates 65 percent [420,422]. This genetic influence appears, in part, to involve single nucleotide polymorphisms in thyroid hormone pathway genes such as the phosphodiesterase gene (PDE8B) [423-425], polymorphisms causing gain [426-433] or loss [434-436] of function TSH receptors [423,437,438] and the type II deiodinase enzyme polymorphisms  [423,439]. Undoubtedly, such polymorphisms account for some of the euthyroid outliers that skew TSH reference range calculations [423,434,440].

 

Figure 3. TSH Between-Method Variability

Figure 3. A. Geometric mean of the TSH results for the range 0.5– 6.6 mIU/L,  (x axis, different assays; dotted lines, overall mean and 10% error). In the plots, the 1-sided 95% CIs of the means are shown (note: the wide interval of assay O is due to results from only 2 runs with a high between-run variation and df = 1 by the Satterthwaite approximation). For the assays outside the 10% limit, the mean value is listed. B. Plot showing the %-difference between TSH methods. The most discrepant assays are shown by triangles and circles. Other assays are shown with the same symbol (x) [29,52].

 

The narrow TSH within-person variability and low (< 0.6) index of individuality (IoI) [222,417, 418,441-443] limits the clinical utility of using the TSH population-based reference range to detect thyroid dysfunction in an individual patient [222,418,443,444]. When evaluating patients with marginally (confirmed) low (0.1–0.4 mIU/L) or high (4–10 mIU/L) TSH abnormalities, it is more important to consider the degree of TSH abnormality relative to patient-specific risk factors for cardiovascular disease rather than the degree of the abnormality relative to the TSH reference range [69,445,446].

TSH Reference Ranges - General Considerations

 

IFCC C-STFT comparison studies (Figure 3) report significant biases between different TSH methods. Currently this prevents establishing universal population or trimester-specific TSH reference ranges that would apply across methods [52,447]. These method biases also impact the frequency of detecting subclinical hypothyroidism [61,448]. Since TSH is a complex glycoprotein, no reference measurement procedure (RMP) is available, or will likely be feasible in the future. However, a harmonization approach [59,60], where methods are recalibrated to the "all method mean", has been shown to have the potential to effectively eliminate current between-method TSH differences that are most pronounced at pathophysiologic levels [29,449]. The IFCC C-STFT is actively working with the IVD industry to encourage manufacturers to harmonize their methods. A reduction of between-method variability could eliminate the need to establish population and trimester-specific TSH reference ranges for each method - a practice that is costly and inconvenient given the large numbers of rigorously screened participants that are necessary to establish reliable 2.5th to 97.5th percentiles for a population [450]. However, even after harmonization minimizes inter-method differences, it remains to be determined to what extent universal ranges would be impacted by other factors such as age [451], ethnicity [396,452] and iodine intake [453]. It may be that a reference range established in one geographic location may not be representative of a different locale or population. After harmonization of TSH methods the advantages of consolidating data from different studies and establishing universal reference limits is clearly apparent.

 

The TSH Population Reference Range

The complex log/linear TSH/FT4 relationship [7,24,221-228] dictates that TSH will be the first abnormality to appear with the development of mild (subclinical) hypo- or hyperthyroidism.  It follows that the setting of the TSH reference limits critically influences the frequency of diagnosing subclinical thyroid disease [69,445,448,454].

 

Guidelines recommend that “TSH reference intervals should be established from the 95 percent confidence limits of the log-transformed values of at least 120 rigorously screened normal euthyroid volunteers who have: (a) no detectable thyroid autoantibodies, TPOAb or TgAb (measured by sensitive immunoassay); (b) no personal or family history of thyroid dysfunction; (c) no visible or palpable goiter and, (c) who are taking no medications except estrogen” [24,450].

 

Multiple factors influence population TSH reference limits, especially the upper (97.5th percentile) limit. Different methods report different ranges for the same population as a result of between-methods biases (Figure 3) [396,448,451,455]. A key factor affecting the upper limit is the stringency used for eliminating individuals with thyroid autoimmunity (thyroid autoantibody positive [456]) from the population [452,456-461]. Other factors relate to population demographics such as sex [452], ethnicity [452,462-464], iodine intake [465], BMI [466-477] and smoking status [462,478,479]. Age is a major factor the influences the TSH upper limit [460,463,480-482] leading to the suggestion that age-specific TSH reference limits should be used (Figure 4) [69,451,480]. However, the relationship between TSH and age is complex. Most studies in iodine sufficient populations have shown an increase in TSH with age [440,452,460,483], whereas other studies have reported no change or a decreased TSH with aging [457,484,485]. This conflicting data could merely represent population differences - with a rising TSH with age reflecting an increasing prevalence of thyroid autoimmunity in iodine-sufficient populations [452], whereas in iodine deficient populations, increasing autonomy of nodular goiter can result in decreased TSH with aging [486-488]. Some studies have reported that a mild TSH elevation in elderly individuals may convey a survival benefit [481,489-492], whereas other studies dispute this [493,494]. However, TSH is a labile hormone and studies cannot assume that a TSH abnormality found in a single determination is representative of thyroid status in the long-term [495,496].

 Figure 4. Suggested management algorithm from reference # 69 Initial management of persistent subclinical hypothyroidism in non-pregnant adults: persistent subclinical hypothyroidism describes patients with elevated serum TSH and within reference range serum FT 4 on two occasions separated by at least 3 months. This algorithm is meant as a guide and clinicians are expected to use their discretion and judgment in interpreting the age threshold around 70 years. * Depending on circumstances, individuals with goiter, dyslipidaemia, and diabetes may also be considered for treatment, along with those with planning pregnancy in the near future.

 

TSH is a heterogeneous glycoprotein [497,498], and TRH-mediated changes in TSH glycosylation [499] have the potential to influence immunoactivity [500,501]. A number of pathophysiologic circumstances are known to alter TSH glycosylation [498,500,502-504]. The demonstration that harmonization of TSH methods successfully mininizes between-method differences [52,53] suggests that under normal conditions current TSH IMAs appear to be "glycosylation blind", and detect different TSH glycoforms in an equimolar fashion [52,53,501]. However, future studies need to include sera from conditions where TRH dysregulation may lead to abnormal TSH glycosylation and bioactivity, such as pituitary dysfunction, NTI and aging [215,239,246,498,505-509].

Pediatric TSH Reference Ranges

 

The adult TSH population reference range does not apply to neonates or children. Serum TSH values are generally higher in neonates and then gradually decline until the adult range is reached after puberty [250-252, 485, 510-514]. This necessitates using age-specific TSH reference ranges for diagnosing thyroid dysfunction in these different pediatric age groups.

 

Subclinical Thyroid Dysfunction

 Subclinical Hyperthyroidism (SCHY).

 The lower (2.5th percentile) TSH reference limit approximates 0.3-0.4 mIU/L, and is fairly independent of the method used [445,452,484,485,515-520]. Subclinical hyperthyroidism (SCHY), is defined as a low but detectable TSH (0.01 –-0.3 mIU/L range) without a FT4 abnormality. The prevalence of endogenous SCHY is low (0.7%) in iodine-sufficient populations [452], but is higher in patients reporting thyroid disease as an iatrogenic consequence of L-T4 replacement therapy [521-523]. SCHY is a risk factor for osteoporosis and increased fracture risk [474,524-526] as well as atrial fibrillation and cardiovascular disease [445,474,527], especially in older patient patients.

Subclinical Hypothyroidism (SCHO).

 

Subclinical hypothyroidism is defined as a TSH above the upper (97.5th percentile) TSH reference limit without a FT4 abnormality [69,448,454,460,516,528-530]. However, since the setting of the TSH upper limit remains controversial, the prevalence of SCHO is highly variable - 4 to 8.5 % [452,521], rising to 15% in older populations [446,456]. In most cases, SCHO is associated with TPOAb positivity, indicative of an autoimmune etiology [452,456]. The clinical consequences of SCHO relate to the degree of TSH elevation [531]. Most guidelines recommend L-T4 treatment of SCHO when is TSH is above 10 mIU/L [68,69] but below 10 mIU/L recommend L-T4 treatment based on patient-specific risk factors (Figure 4) [69]. There is active debate concerning the efficacy of treating SCHO to prevent progression [532-535], or improve renal [536,537], cardiovascular [474,524,531,538-543], or lipid [544-546] abnormalities that can be associated with SCHO [69,547].

 

Thyroid Dysfunction and Pregnancy

 

It is well documented that overt hypo- or hyperthyroidism is associated with both maternal and fetal complications [548-550]. However, the impact of maternal subclinical thyroid dysfunction remains controversial [253]. No maternal or fetal complications appear associated with subclinical hyperthyroidism during pregnancy [258,551]. First trimester "gestational hyperthyroidism" is typically transient and hCG-related, as described above. In contrast, short-term and long-term outcome studies of maternal subclinical hypothyroidism [550] are complicated by heterogeneity among studies arising from a myriad of factors influencing TSH cutoffs, such as gestational stage, TSH method used, maternal TPOAb status, and current and pre-pregnancy iodine intake [277,454]. Using gestational age-specific reference intervals the frequency of SCHO in first trimester pregnancy approximates 2-5 percent [552-556]. A number of studies have reported that subclinical hypothyroidism is associated with increased frequency of maternal and fetal complications, especially when TPOAb is positive [557-559].  Maternal complications have included miscarriage [474,548,560-562], preeclamsia [548,563], placental abruption [552], preterm delivery [552,562,564] and post-partum thyroiditis [565]. Fetal complications have included intrauterine growth retardation and low birth weight [258,548,566-568] and possible impaired neuropsychological development [550,569,570]. It remains controversial whether L-T4 treatment of SCHO in early gestation decreases risk of complications [559,562,564,571].

 

 

Trimester-Specific TSH Reference Ranges.

As with non-pregnant patients, TSH is the first-line test used for assessing thyroid status during pregnancy when gestation-related TSH changes occur [66,67,253,254,555,556,572].  In the first trimester, there is a transient rise in FT4 caused by high hCG concentrations stimulating the TSH receptor - because hCG shares some homology with TSH [254,308,309,573,574]. The degree of TSH suppression is inversely related to the hCG concentration and can be quite profound in patients with hyperemesis who have especially high hCG [271,575-577]. As gestation progresses, TSH tends to return towards pre-pregnancy levels [271]. Recent studies from different geographic areas with diverse iodine intakes have using different TSH methods have reported higher trimester-specific TSH upper limits than recommended by previous guidelines [253,269,271,454,556,578-580]. In response, the American Thyroid Association has recently revised their pregnancy guidelines [66,74] to replace trimester-specific reference limits by a universal upper TSH limit of 4.0 mIU/L, when TPOAb is negative and local reference range data is not available. However, at this time between-method biases (Figure 3) clearly preclude proposing universal TSH or FT4 reference ranges that would apply to all methods and all populations [52,53,267,271,447]. It is critical that the IVD manufacturers respond to the urging of the IFCC C-STFT and harmonize their TSH methods to increase the feasibility of establishing TSH universal reference limits for pregnancy [52,53]. Requiring each institution to establish their own trimester-specific reference ranges for thyroid tests is impractical, given the costs, logistics and ethical considerations involved in recruiting the more than 400 disease-free pregnant women needed to establish reliable ranges for each trimester [270]. Only after methods are re-standardized (FT4) or harmonized (TSH), will it be feasible to propose trimester-specific reference ranges that would apply across methods.  However, such ranges would still be influenced by differences in ethnicity [280] and iodine intake, especially pre-pregnancy iodine intake that influences thyroidal iodine stores [277]. There is also a current need to reevaluate optimal TPOAb cutoffs needed to exclude those individuals with thyroid autoimmunity whose inclusion skews TSH upper limits [271,280,454,574,581,582].

 

 

 Clinical Utility of TSH Measurement

 

Ambulatory Patients

 

In the outpatient setting the reliability of TSH testing is not influenced by the time of day of the blood draw, because the diurnal TSH peak occurs between midnight and 0400 [583-586]. Third-generation TSH assays (FS ~0.01 mIU/L) have now become the standard of care because they can reliably detect the full spectrum of thyroid dysfunction from overt hyperthyroidism to overt hypothyroidism, provided that hypothalamic-pituitary function is intact and thyroid status is stable [24,57,216,242,359,414,587,588]. TSH is also used for optimizing L-T4 therapy - a drug with a very narrow therapeutic index [359,589,590]. Because TSH secretion is slow to respond to changes in thyroxine status there is no need to withhold the L-T4 dose on the day of the blood test [24]. In addition, targeting the degree of TSH suppression relative to recurrence risk plays a critical role in the management of thyroid cancer [72,591-593].

 

 

Hospitalized Patients with Nonthyroidal Ilnesses (NTI)

 

Routine thyroid testing in the hospital setting is not recommended because thyroid test abnormalities are frequently seen in euthyroid sick patients [238,594]. Non-thyroidal illness, sometimes called the "sick euthyroid syndrome" is associated with alterations in hypothalamic/pituitary function and thyroid hormone peripheral metabolism often exacerbated by drug influences [100,218,239,245,595]. T3 levels typically fall early in the illness followed by a fall in T4 as the severity of illness increases. [24,244,595-597]. As thyroid hormone levels fall TSH typically remains unchanged, or may be low early in the illness, especially in response to drug therapies such as dopamine or glucocorticoid [100,101,218]. During the recovery phase, TSH frequently rebounds above the reference range [243].  However, high TSH may also be seen associated with psychiatric illness [598]. It is important to distinguish the generally mild, transient TSH alterations typical of NTI from the more profound and persistent TSH changes associated with hyper- or hypothyroidism [24,238,244].

 

Misleading TSH Measurements

 

TSH can be diagnostically misleading either because of (a) biological or (b) technical factors. from heterophile antibodies (HAbs) or endogenous TSH autoantibodies are the most common causes of a falsely high TSH [299,329,599].

 

Biologic factors causing TSH diagnostic dilemmas

 

Unstable thyroid function

TSH can be misleading when there is unstable thyroid status - such as in the early phase of treating hyper- or hyperthyroidism or non-compliance with L-T4 therapy -when there is a lag in the resetting of pituitary TSH to reflect a new thyroid status [600]. During such periods of instability TSH will be misleading and FT4 will be the more diagnostically reliable test.

 

Pituitary/Hypothalamic Dysfunction

Pituitary dysfunction is rare in ambulatory patients [509]. TSH measurement is unreliable in cases of both central hypothyroidism and central hyperthyroidism caused by TSH-secreting adenomas [215,217,219,508].

 

Central Hypothyroidism (CH)

Central hypothyroidism (CH) is rare (1/1000 as prevalent as primary hypothyroidism, 1/160,000 detected by neonatal screening) [509, 601]. CH can arise from disease at either the pituitary or hypothalamic level, or both [509].  A major limitation of using a TSH-centered screening strategy is that this strategy will miss a diagnosis of CH, because the TSH isoforms secreted in CH are abnormally glycosylated and bio-inactive, yet will be detected as paradoxically normal TSH by current IMA methods despite the presence of clinical hypothyroidism [215, 217, 602]. The clinical diagnosis of CH can be confirmed biochemically as a low FT4/normal-low TSH discordance. Serum FT4 should be used to optimize L-T4 replacement therapy. In the absence of clinical suspicion, investigations for pituitary dysfunction should only be initiated after ruling-out technical interference.

 

TSH-secreting pituitary adenomas

TSHomas are characterized by near-normal TSH despite clinical hyperthyroidism [603]. Since this is a rare (0.7%) type of pituitary adenoma, technical interference causing paradoxically high TSH, such as a TSH autoantibody should be excluded before initiating inconvenient and unnecessary pituitary imaging or dynamic (T3 suppression or TRH stimulation) diagnostic testing. TSHomas are characterized by discordance between the clinical presentation and a paradoxically non-suppressed TSH despite high thyroid hormone levels and clinical hyperthyroidism [604]. This clinical/biochemical discordance reflects adenoma secretion of TSH isoforms with enhanced biologic activity that cannot be distinguished from bioactive TSH by IMA methods. Failure to diagnose the pituitary as the cause of the hyperthyroidism can lead to inappropriate thyroid ablation. The treatment of choice is surgery but in cases of surgical failure somatostatin analog treatment has been found effective [604]. Note that the biochemical profile (high thyroid hormones and non-suppressed TSH) is similar to that seen with thyroid hormone resistance syndromes [605]. When pituitary imaging is equivocal, genetic testing may be necessary to distinguish between these two conditions.

 

Resistance to Thyroid Hormone (RTH)

Resistance to thyroid hormone is biochemically characterized by high thyroid hormone (FT4 +/- T3) levels and a non-suppressed, sometimes slightly elevated TSH without signs and symptoms of thyroid hormone excess [606]. Early cases of resistance to thyroid hormone were shown to result from mutations in the thyroid hormone receptor B [607]. More recently the definition of RTH has been broadened to include other causes of thyroid hormone resistance - mutations in the thyroid hormone cell membrane transporter MCT8, and a range of genetic thyroid hormone metabolism defects (SBP2) [608]. These resistance syndromes display a spectrum of clinical and biochemical profiles may need to be identified by specialized genetic testing.

 

Activating or Inactivating TSH Receptor Mutations

Non-autoimmune hyperthyroidism resulting from an activating mutation of the TSH receptor (TSHR) is rare [426-433]. A spectrum of loss-of-function TSHR mutations (TSH resistance) causing clinical and subclinical hypothyroidism despite high thyroid hormone levels, have also been described [434-436]. Because TSHR mutations are a rare cause of TSH/FT4 discordances, technical interferences should first be excluded before considering a TSHR mutation as the cause of these discordant biochemical profiles.

 

Technical Factors causing TSH Diagnostic Dilemmas

 

Causes of technical interferences with TSH measurement are similar to those discussed for thyroid hormone tests.

 

Non Analyte -Specific Interferences

 

 Heterophile Antibodies (HAbs) can cause falsely high TSH IMA tests [289,294,300,328-330, 609].  The HAb in some patient's sera interfere strongly with some manufacturers tests but appear inert in others [609]. This is why re-measurement in a different manufacturers assay should be the first test for interference. A fall in TSH in response to blocker-tube treatment is typically used to confirm HAb interference

 

 Anti-Reagent Antibody Interferences.

  As discussed for free hormone tests,,,,,, some patients have antibodies that target test reagents (such Rhuthenium) that cause interference with TSH and/or free hormone tests. It should be noted that the anti-Rhuthenium antibodies of different patients may affect different analytes to different degrees [339-342].

 

Tests employing Streptavidin-Biotin

 reagents are prone to interferences from antibodies targeting either Streptavidin [344] or biotin reagents [345]. Alternatively, high dose biotin ingestion has been known to produce interference with thyroid and other tests in an analyte-specific, platform-specific manner [346-350].

 

Analyte-specific interferences typically result from autoantibodies targeting the analyte. Depending on the analyte and test formulation, autoantibody interferences typically cause falsely-high test results, but can cause falsely-low test results, as in the case of Tg autoantibodies. It should be noted that transplacental passage both heterophile antibodies or anti-analyte autoantibodies (i.e. TSHAb or T4Ab) have the potential to interfere with neonatal screening tests [351-354]. Specifically, maternal TSH autoantibodies can cross the placenta and may cause a falsely high TSH screening test in the newborn mimicking congenital hypothyroidism, whereas maternal T4 autoantibodies could cause falsely high neonatal T4 masking the presence of congenital hypothyroidism [230, 353].

 

TSH Autoantibodies (TSHAb)/"Macro TSH".

Analytically suspicious TSH measurements are not uncommon [290] and have been reported in up to five percent of specimens subjected to rigorous screening [294]. In recent years there have been a number of reports of TSHAb, often referred to as "macro" TSH, causing spuriously high TSH results in a range of different methods [610,611]. The prevalence of TSHAb approximates 0.8 percent, but can as high as ~1.6 percent in patients with subclinical hypothyroidism. Showing a lowering of TSH in response to a polyethylene glycol (PEG) precipitation of immunoglobulins is the most convenient test for TSHAb [599,611]. Alternatively, column chromatography can show TSH immunoactivity in a high molecular weight peak representing a bioinactive TSH-immunglobulin complex [599,611].

 

TSH Variants.

TSH variants are a rare cause of interference [612]. Nine different TSH beta variants have been identified to date [613]. These mutant TSH molecules may have altered immunoactivity and be detected by some TSH IMA methods but not others [612]. The bioactivity of these TSH mutants is variable and can range from normal to bio-inert [613], resulting in discordances between the TSH concentration and clinical status [612] and/or discordant TSH/FT4 relationships [613]. These TSH genetic variants are one of the causes of congenital hypothyroidism [614-616].

 

THYROID SPECIFIC AUTOANTIBODIES (TRAB , TPOAB AND TGAB)

 

Tests for antibodies targeting thyroid-specific antigens such as thyroid peroxidase (TPO), thyroglobulin (Tg) and TSH receptors (TSHR) are used as markers for autoimmune thyroid conditions [37,617]. Over the last four decades, thyroid antibody test methodologies have evolved from semi-quantitative agglutination, complement fixation techniques and whole animal bioassays to specific ligand assays using recombinant antigens or cell culture systems transfected with the human TSH receptor [37,618621]. Unfortunately, the diagnostic and prognostic value of these tests has been hampered by methodologic differences as well as difficulties with assay standardization [622]. Although most thyroid autoantibody testing is currently made on automated immunoassay platforms, methods vary in sensitivity, specificity and the numeric values they report because of standardization issues [44,582,620,623].  Thyroid autoantibody testing can be useful for diagnosing or monitoring treatment for a number of clinical conditions, however these tests should be selectively employed as adjunctive tests to other diagnostic testing procedures.

 

TSH Receptor Autoantibodies (TRAb)

 

The TSH receptor (TSHR) serves as a major autoantigen [624,625]. Thyroid gland stimulation occurs when TSH binds to TSHR on thyrocyte plasma membranes and activates the cAMP and phospholipase C signaling pathways [625]. The TSH receptor belongs to the G protein-coupled class of transmembrane receptors. It undergoes complex posttranslational processing in which the ectodomain of the receptor is cleaved to release a subunit into the circulation [624]. A TSH-like thyroid stimulator found uniquely in the serum of Graves’ disease patients was first described using a guinea pig bioassay system in 1956 [626]. Later, using a mouse thyroid bioassay system this serum factor displayed a prolonged stimulatory effect as compared to TSH and hence was termed to be a “long-acting thyroid stimulator” or LATS [627,628]. Much later, the LATS factor was recognized not to be a TSH-like protein but an antibody that was capable of stimulating the TSH receptor causing Graves’ hyperthyroidism [629]. TSH receptor antibodies have also become implicated in the pathogenesis of Graves’ opthalmopathy [629-632]. TRAbs are heterogeneous (polyclonal) and fall into two general classes both of which can be associated with autoimmune thyroid disorders – (a) thyroid stimulating autoantibodies (TSAb) that mimic that the actions of TSH and cause Graves’ hyperthyroidism and (b), blocking antibodies (TBAb) that block TSH binding to its receptor and can cause hypothyroidism [37,48,621,625,629,633,634]. Although TSH, TSAb and TBAb appear to bind to different sites on the TSH receptor ectoderm, TSAb and TBAb have similar affinities and often overlapping epitope specificities [635]. In some cases of Graves’ hyperthyroidism, TBAb have been detected in association with TSAb [636,637] and the dominance of one over the other can change over time in response to treatment [638]. Because both TSAb and TBAb can be present in the same patient, the relative concentrations and receptor binding characteristics of these two classes of TRAb may influence the severity of Graves’ hyperthyroidism and the response to antithyroid drug therapy or pregnancy [624,636,639-643]. For completeness, it should also be mentioned that a third class of “neutral” TRAb has also been described, of which the functional significance has yet to be determined [641,644].

 

Two different methodologic approaches have been used to quantify TSH receptor antibodies [40,620,633,645]: (i) TSH receptor tests (TRAb assays) also called TBII or TSH Binding Inhibition Immunoglobulin assays, and (ii) Bioassays that use whole cells transfected with human or chimeric TSH receptors that produce a biologic response (cAMP or bioreporter gene) when TSAb or TBAb are present in a serum specimen. In recent years automated immunometric assays using recombinant human TSHR constructs have been shown to have high sensitivity for reporting positive results in Graves' disease sera [620,646]. However, assay sensitivity varies among current receptor versus bioassay methods [43]

 

Bioassay methods (TSAb/TBAb)

 

The first TSH receptor assays used surgical human thyroid specimens, mouse or guinea pig thyroid cells, or rat FRTL-5 cell lines to detect TSH receptor antibodies. These methods typically required pre-extraction of immunoglobulins from the serum specimen [626,633,647-652]. Later, TRAb bioassays used cells with endogenously expressed or stably transfected human TSH receptors and could use unextracted serum specimens [653-655]. Current TRAb bioassays are functional assays that use intact (typically CHO) cells transfected with human or chimeric TSH receptors, which when exposed to serum containing TSH receptor antibodies use cAMP or a reporter gene (luciferase) as a biological marker for any stimulating or blocking activity in a serum [40,42,620,648,651,653,656]. Bioassays are more technically demanding than the more commonly used receptor assays because they use viable cells. However, these functional assays can be modified to detect TBAb that may coexist with TSAb in the same sera and make interpretation difficult [40,657]. The most recent development is for 2nd generation assays to use a chimeric human/rat LH TSHR to effectively eliminate the influence of blocking antibodies. This new approach has shown excellent sensitivity and specificity for diagnosing Graves' hyperthyroidism and clinical utility for monitoring the effects of anti-thyroid drug therapy [42].

 

 

TSH Receptor  (TRAb)/TSH Binding Inhibitory Immunoglobulin (TBII) Methods

 

TRAb methods detect serum immunoglobulins that bind TSHR but do not functionally discriminate stimulating from blocking antibodies. TRAb methods are based on standard competitive or noncompetitive principles. First generation methods were liquid-based whereby immunoglobulins in the serum inhibited the binding of 125I-labeled TSH or enzyme-labeled TSH to a TSH receptor preparation [40,658]. These methods used TSH receptors of human, guinea-pig or porcine origin [658]. After 1990, a second-generation of both isotopic and non-isotopic methods were developed that used and immobilized porcine or recombinant human TSH receptors [40,659-661]. These second-generation methods were shown to have significantly more sensitivity for detecting Graves' thyroid stimulating immunoglobulins than first-generation tests [620]. In 2003 a third-generation of non-isotopic methods were developed that were based on serum immunoglobulins competing for immobilized TSHR preparation (recombinant human or porcine TSHR) with a monoclonal antibody (M22) [37,40,42,620,648,656,660,662-666]. 3rd generation assays have also shown a good correlation and comparable overall diagnostic sensitivity with bioassay methods [620,636,648,667,669]. Current third-generation tests have now been automated on several immunoassay platforms [620]. However, between-method variability remains high and interassay precision often suboptimal (CVs > 10 %) despite the use of the same international reference preparation for calibration [622,670]. This fact makes it difficult to compare values using different methods and indicates that further efforts focused on additional assay improvements are needed [37,622,671].

 

 

Clinical Use of TRAb Tests

 

Over the last ten years automated IMA methods have dramatically lowered the cost and increased the availability of TRAb testing [43,646,672]. Automated TRAb IMAs are not functional tests and do not distinguish between stimulating and blocking TRAbs. However this distinction is usually unnecessary, since it is evident from clinical evidence of hyper- or hypothyroid features. Also, both TSHR stimulating and blocking antibodies may be detected simultaneously in the same patient and cause diagnostic confusion [42,673]. Because the sensitivity and specificity of current third-generation TRAb tests is over 98 percent, TRAb testing can be useful for determining the etiology of hyperthyroidism [620,672], as an independent risk factor for Graves’ opthalmopathy [632] and may be useful for monitor responses to therapy [620,674,675]. TRAb measured prior to radioiodine therapy for Graves' hyperthyroidism can also help predict the risk for exacerbating opthalmopathy [630,676-680]. There is conflicting data concerning the value of using TRAb to predict the response to antithyroid drug treatment or risk of relapse [42,637,661,667,681-685]. An important application of TRAb testing is to detect high TRAb concentrations in pregnant patients with a history of autoimmune thyroid disease or active or previously treated Graves’ hyperthyroidism, in whom transplacental passage of stimulating or blocking TRAb can cause neonatal hyper- or hypothyroidism, respectively [40,67,620,645,686-689]. Because the expression of thyroid dysfunction may be different in the mother and infant, automated IMA methods have the advantage of being able to detect both stimulating and blocking antibodies [690]. It is currently recommended [74] that TRAb be measured in the first trimester in all pregnant patients with active Graves’ hyperthyroidism or who have received prior ablative (radioiodine or surgery) therapy for Graves’ disease in whom TRAb can remain high even after patients have been rendered hypothyroid and are being maintained on L-T4 replacement therapy. When TRAb is high in the first trimester additional TRAb testing is recommended at weeks 18-22 and 30-34 [24,37,67,74,636,687,691].

 

 

Thyroid Peroxidase Autoantibodies (TPOAb)

 

TPO is a large, dimeric, membrane-associated, globular glycoprotein that is expressed on the apical surface of thyrocytes. TPO autoantibodies (TPOAb) found in sera typically have high affinities for an immunodominant region of the intact TPO molecule. When present, these autoantibodies vary in titre and IgG subclass and display complement-fixing properties [692]. Studies have shown that epitope fingerprints are genetically conserved suggesting a possible functional importance [693]. However, it is still unclear whether the epitope profile correlates with the presence of, or potential for, the development of thyroid dysfunction with which TPOAb presence is most commonly associated [692,694,697].

Prevalence of thyroid antibodies across TSH intervals in women (A) and men (B). The abscissa TSH values correspond to the upper and lower limits of the intervals spanning each set of bars. Asterisks denote a significant difference in prevalence from the TSH range with lowest antibody prevalence, 0.1 and 1.5 mIU/liter for women and 0.1 and 2.0 mIU/liter for men [456].

 

TPOAb antibodies were initially detected as antibodies against thyroid microsomes (antimicrosomal antibody, AMA) using semi-quantitative complement fixation and tanned erythrocyte hemaagglutination techniques [698-700]. Recent studies have identified the principal antigen in the AMA tests as the thyroid peroxidase (TPO) enzyme, a 100 kD glycosylated protein present in thyroid microsomes [701, 702]. Manual agglutination tests have now been replaced by automated, more specific TPOAb immunoassay or immunometric assay methods that use purified or recombinant TPO [24,37,619,703-710]. Despite calibration against the same International Reference Preparation (MRC 66/387), there is considerable inter-method variability of current TPOAb assays (correlation coefficients 0.65 and 0.87) that precludes the numeric comparison of serum TPOAb values reported by different tests [37,618,619,706,709,710]. It appears that both the methodologic principles of the test and the purity of the TPO reagent used may influence the sensitivity, specificity and reference range of the method [37,619]. The variability in sensitivity limits and the reference ranges of different methods has led to different interpretations regarding the normalcy of having a detectable TPOAb [37,582,710].

 

 TPOAb Clinical Significance

 

Estimates of TPOAb prevalence depend on the sensitivity and specificity of the method employed [582,710,711]. In addition, ethnic and/or geographic factors (such as iodine intake) influence the TPOAb prevalence in population studies [487]. For example, TPOAb prevalence is significantly higher (~11 percent) in dietary iodine-sufficient countries like the United States and Japan as compared with iodine deficient areas in Europe (~ 6 percent) [452,515,712]. The prevalence of TPOAb is higher in women of all age groups and ethnicities, presumably reflecting the higher propensity for autoimmunity as compared with men [452,712]. Approximately 70-80 % of patients with Graves' disease and virtually all patients with Hashimoto’s or post-partum thyroiditis have TPOAb detected [619,706,709,711,713]. TPOAb has, in fact, been implicated as a cytotoxic agent in the destructive thyroiditic process [697,714717]. However, TPOAb prevalence is also significantly higher in various non-thyroidal autoimmune disorders in which no apparent thyroid dysfunction is evident [718-720]. Aging is associated with an increasing prevalence of TPOAb that parallels the increasing prevalence of both subclinical (mild) and clinical hypothyroidism [452]. In fact, the NHANES III survey reported that TPOAb prevalence increases with age and approaches 15-20 percent in elderly females in the iodine-sufficient United States [452]. This same study found that the odds ratio for hypothyroidism was strongly associated with the presence of TPOAb but not TgAb, suggesting that only TPOAb has an autoimmune etiology [452]. Although the presence of TgAb alone did not appear to be associated with hypothyroidism or TSH elevations, the combination of TPOAb and TgAb versus TPOAb alone may be more pathologically significant (Figure 5), although further studies would be needed to confirm this [452,456,459,697]. It is now apparent that the presence of TPOAb in the serum of apparently euthyroid individuals (TSH within reference range) appears to be a risk factor for future development of overt hypothyroidism that subsequently becomes evident at the rate of approximately 2 percent per year in such populations [46,532,692,693].

 

In this context, it is reasonable to assume that TPOAb measurement may serve as a useful prognostic indicator for future thyroid dysfunction [46,721]. However, it is noteworthy that the detection of TPOAb does not always precede the development of thyroid dysfunction. A recent study suggests that a hypoechoic ultrasound pattern can be seen before a biochemical TPOAb abnormality is detected [458,487]. Further, some individuals with unequivocal TSH elevations, presumably resulting from autoimmune destructive disease of the thyroid, do not have TPOAb detected [456]. Presumably, this paradoxical absence of TPOAb in some patients with elevated TSH likely reflects the suboptimal sensitivity and/or specificity of current TPOAb tests or a non-autoimmune cause of thyroid failure (i.e. atrophic thyroiditis) [452,456,710,722].

 

Although changes in autoantibody concentrations often occur with treatment or reflect a change in disease activity, serial TPOAb measurements are not recommended for monitoring treatment for autoimmune thyroid diseases [359,619,723]. This is not surprising since treatment of these disorders addresses the consequence (thyroid dysfunction) and not the cause (autoimmunity) of the disease. However, where it may have an important clinical application is to employ the presence of serum TPOAb as a risk factor for developing thyroid dysfunction in patients receiving Amiodarone, Interferon-alpha, Interleukin-2 or Lithium therapies which all appear to act as triggers for initiating autoimmune thyroid dysfunction in susceptible (especially TPOAb-positive) individuals [24,101,724-730].

 

During pregnancy the presence of TPOAb has been linked to reproductive complications such as miscarriage, infertility, IVF failure, fetal death, pre-eclampisa, pre-term delivery and post-partum thyroiditis and depression [66,67,564,731-742]. However, if this association represents cause or effect has yet to be been resolved.

 

  Thyroglobulin Autoantibodies (TgAb)

 

Thyroglobulin autoantibodies predominantly belong to the immunoglobulin G (IgG) class, are not complement fixing and are generally conformational [743]. Serum TgAb were the first thyroid antibody to be detected in patients with autoimmune thyroid disorders using tanned red cell hemagglutination techniques [699]. Subsequently, methodologies for detecting TgAb have evolved in parallel with those for TPOAb measurement from semi-quantitative techniques, to more sensitive ELISA and RIA methods and most recently non-isotopic competitive or non-competitive immunoassays [10,37,44,706,710,713,744-747]. Unfortunately, the inter-method variability of these TgAb assays is even greater than that of TPOAb tests discussed above [10,37,44,745-747]. Additionally, high levels of thyroglobulin in the serum have the potential to influence TgAb measurements [747-750]. Between-method variability is influenced by the purity and the epitope specificity of the Tg reagent, as well as the patient-specific epitope specificity of the TgAb in the serum [751,752]. As with TPOAb methods, TgAb tests have highly variable sensitivity limits and cut-off values for "TgAb positivity", despite the use of the same International Reference Preparation (MRC 65/93) (Figure 6) [10, 44, 745-747, 753]. It should be noted that the manufacturer-recommended cutoffs are set for diagnosing thyroid autoimmunity and are too high for detecting levels of TgAb that interfere with Tg measurements - the much lower assay FS limit (Figure 6) is the recommended cutoff to define TgAb-positivity for DTC monitoring [24]. Although there are reports that low levels of TgAb may be present in normal euthyroid individuals, it is unclear whether this represents assay noise due to matrix effects or "natural" antibodies [744,754]. Further complicating this question are studies suggesting that there may be qualitative differences in TgAb epitope specificities expressed by normal individuals versus patients with either differentiated thyroid cancers (DTC) or autoimmune thyroid disorders [744,752,755]. These differences in test sensitivity and specificity negatively impact the reliability of determining the TgAb status (positive versus negative) of specimens prior to Tg testing.

Clinical Utility of TgAb Tests

 

Autoantibodies against Tg are encountered in autoimmune thyroid conditions, usually in association with TPOAb [46,452,746, 756]. However, the NHANES III survey found that only three percent of subjects with no risk factors for thyroid disease had serum TgAb present without detectable TPOAb (Figure 5) [452,456]. Further, in these subjects there was no association observed between the isolated presence of TgAb and TSH abnormalities [452,456]. This suggests that it may be unnecessary to measure both TPOAb and TgAb for a routine evaluation of thyroid autoimmunity [37,46,456]. In fact, when autoimmune thyroid disease is present, there is some evidence that assessing the combination of TPOAb and TgAb has greater diagnostic utility than the TPOAb measurement alone (Figure 5)  [46,456,459,757].

 

TgAb measurement is primarily used as an adjunctive test to serum Tg measurement when monitoring patients with differentiated thyroid cancers (DTC) [72,593]. The role of TgAb testing is two-fold: 1) to authenticate that a Tg measurement is not compromised by TgAb interference, 2) as an independent surrogate tumor-marker in the ~20 percent of patients with circulating TgAb. Current guidelines recommend that all sera be prescreened for TgAb by a sensitive immunoassay method prior to serum Tg testing, because there appears to be no threshold TgAb concentration that precludes TgAb interference with Tg measurements [9,10,24,44,593,713,746,758]. Immunoassay methods detect TgAb in approximately 25 percent of patients presenting with DTC [44,713,759-761]. The prevalence of TgAb is typically higher in patients with papillary versus follicular tumors and is frequently associated with the presence of lymph node metastases [746,759,761, 62]. Perhaps of even greater importance is the observation that serially determined TgAb concentrations may also serve as an independent parameter for detecting changes in tumor mass in patients with an established diagnosis of DTC [Figure 6Ad(ii)] [761-766]. For example, after TgAb-positive patients are rendered disease-free by surgery, TgAb concentrations typically progressively decline during the first few post-operative years and typically become undetectable after a median of three years of follow-up [761,762,766]. In contrast, a rise in, or de novo appearance of, TgAb is often the first indication of tumor recurrence [713,761,762]. However, when using serial TgAb measurements as a surrogate marker for changes in tumor burden it is essential to use the same TgAb method, because of the large between-method differences observed with this assay (Figure 6) [9,10,44,713,745,747,753].

Figure 6. TgAb Method Comparison. 143 DTC sera with TgAb above the MCO for the reference method (Kronus/RSR) and evidence of TgAb interference with Tg measurement (Tg-IMA/Tg-RIA ratios < 80 percent) were measured by three different TgAb methods. The red bars show the manufacturer recommended cutoff for TgAb-positivity for each method. The black bar denotes the functional sensitivity (FS) of each method [44].

 

THYROGLOBULIN (TG)

 

Thyroglobulin plays a central role in a wide variety of pathophysiologic thyroid conditions, including acting as an autoantigen for thyroid autoimmunity [617,743,767]. Serum Tg levels can serve as a marker for iodine status of a population [768-771], whereas dyshormongenesis resulting from genetic defects in Tg biosynthesis is a cause of congenital hypothyroidism [24,772-775]. Because Tg has a thyroid-tissue specific origin, a Tg measurement can aid in determining the etiology of congenital hypothyroidism (athyreosis versus dyshormonogenesis) [776,777]. Likewise, a paradoxically low serum Tg can be used to distinguish factitious hyperthyroidism from the high Tg expected with endogenous hyperthyroidism [778-780]. This chapter focuses on the primary clinical use of Tg measurement - a tumor-marker test for post-operative monitoring of patients with follicular-derived (differentiated) thyroid cancer (DTC) [32,72,404,781-788]. (Table 3)

 

Most Tg testing is currently made by rapid, automated immunometric assays (IMA) with second-generation functional sensitivity (2G-Tg-IMA, FS≤ 0.1 µg/L). Assays with this level of FS obviate the need for recombinant human TSH (rhTSH) stimulation [ [11,32,72,416,784,789-793]. The major limitation of IMA methodology is its propensity for TgAb interference causing falsely low/undetectable serum Tg-IMA that can mask disease [10,31,45,58,760,790,794-798]. Currently, most laboratories in the United States first establish the TgAb status of the specimen (negative or positive) in order to restrict Tg-IMA testing to TgAb-negative sera, whereas TgAb-positive specimens are reflexed for testing by Tg methodologies believed to be less prone to interferences, such as RIA [30,32,796] or LC-MS/MS   [31,799-801].

 

Technical Limitations of Tg Methods

Thyroglobulin measurement remains technically challenging [788]. Five methodologic problems impair the clinical utility of this test: (a) between-method biases; (b) suboptimal functional sensitivity; (c) suboptimal between-run precision over the typical clinical interval used to monitor DTC patients (6-12 months); (d) "hook" problems (some IMA methods), and interferences caused by both (e) Heterophile antibodies (HAb) and (f) Tg autoantibodies (TgAb).

 

Tg Assay Functional Sensitivity

 

As discussed for TSH, assay functional sensitivity (FS) represents the lowest analyte value that can be reliably detected under clinical practice conditions. For Tg assays FS is defined by the lowest Tg concentration that can be measured in human serum with 20 percent coefficient of variation (CV) in runs made over a 6-12 month period using at least two different lots of reagents and two instrument calibrations [24,58,72,404,802]. These stipulations are needed because assay precision erodes over time and the clinical interval for serum Tg monitoring of DTC patients is typically long (6-12 months) [9,408,803]. For Tg assays it is critical to use FS as the lowest reporting limit in preference to a LOQ calculation (20 percent CV), because LOQ does not stipulate a relevant time-span for assessing precision [24,405,407,804,805]. Another stipulation of the FS protocol [24] is to assess precision using the appropriate test matrix (human serum) in preference to a commercial QC preparation, because instruments and methods are matrix-sensitive [407]. Since Tg-IMA testing is typically restricted to TgAb-negative sera, precision estimates should be made in TgAb-negative human serum pools [407]. In contrast, Tg-RIA and Tg-LC-MS/MS testing is typically reserved for sera containing TgAb, necessitating precision estimation in TgAb-positive human serum pools.

 

As with TSH [220,397], there has been a progressive improvement in the FS of Tg methods that has led to the adoption of a generational approach to Tg assay nomenclature. Currently, some Tg-IMAs, all Tg-RIAs and all Tg-LC-MS/MS methodologies still only have first-generation functional sensitivity (FS = 0.5-1.0 µg/L) [4,10,32,33,58]. Over the last ten years second-generation immunometric assays (2G-Tg-IMA), characterized by an order of magnitude greater functional sensitivity (FS 0.05-0.10 µg/L), have become available. 2G-Tg-IMA testing is now considered the standard of care in the absence of TgAb [31-33,58,72,296,783,806-808]. When disease is absent the basal serum 2G-Tg-IMA is typically below 0.5 µg/L, even without RAI treatment [809,810]. It follows that the inferior FS (~1 µg/L) of first-generation assays can barely distinguish subnormal values from the Tg levels seen when an intact thyroid gland is present (~2-40 µg/L), and are clearly too insensitive to detect recurrences in thyroidectomized patients unless recombinant human TSH (rhTSH) stimulation is employed   [296,593,758,782,811,812]. Now that 2G-IMA-Tg testing has become the standard of care [72], there is no longer a need for routine rhTSH stimulation to boost the Tg level to values detectable by first-generation tests, because basal (TSH suppressed) Tg correlates with rhTSH-stimulated Tg measured by 2G-Tg-IMA   [10,11,32,58,72,296,413, 416,789,791-793,806,807,813-816]. Studies have shown that a basal 2G-Tg-IMA below 0.1 µg/L predicts a negative rhTSH test (rhTSH-stimulated Tg <2.0 µg/L) with a high degree of confidence [72,296,791,792,817]. Even so, the use of a 2nd generation Tg assay does not eliminate the need for periodic ultrasound examinations, because many histologically confirmed lymph nodes metastases are inefficient Tg secretors and may be associated with an undetectable serum Tg, even when measured by 2G-Tg-IMA [807,818-821].

Figure 7. Panel A shows the comparison of serum Tg values reported for 37 TgAb-negative DTC patients with persistent/recurrent DTC measured by a 2G-Tg-IMA (Beckman), Tg-LC-MS/MS (Mayo Medical Labs) and the USC Tg-RIA method. Sera with Tg values below the FS limit of the method are shown in the shaded areas, Although each method was standardized against CRM-457, the sera marked in red displayed  > 30% difference in Tg values that reflected different method specificities for detecting tumor-derived Tg molecules - differences with the potential to disrupt clinical management following a change in Tg method. Panel B shows the method comparison for 52 TgAb-positive DTC patients with structural disease. Sera with unequivocally undetectable Tg-LC-MS/MS values (no peak) are shown by solid red squares, whereas sera with marginally detectable Tg-LC-MS/MS values in the 0.3 to 0.5 µg/L range are shown by open red squares [31].

 

Between-Method Biases

 

Although most Tg methods claim to be standardized against the Certified Reference Preparation CRM-457 [9,822,823] there can be significant differences between the Tg values reported for the same serum measured by different methods, even in the absence of TgAb (Figure 7A) [10,19,24,32,58,799,824]. Between-method Tg variability is higher than the biologic variability (~16 percent) in euthyroid subjects [442, 803]. In fact, studies have shown that there can be a two-fold difference in Tg values reported for the same serum measured by different methods [32]. Although this reflects standardization and matrix differences to some extent [299,797], for the most part this between-method variability reflects differences in method specificities for detecting heterogeneous serum Tg isoforms [10,825-827]. It should be noted that because IMA methodology uses monoclonal antibody reagents, IMAs have narrower specificities for detecting Tg heterogeneity than RIA methods that use polyclonal antibodies [9,10,826-829]. Because Tg-IMAs differ in their sensitivity to TgAb interference, between-method Tg variability can also result from using an insensitive TgAb test that reports false-negative TgAb values (Figure 6)  [19,44,830].

 

When TgAb is absent and a 2G-Tg-IMA method is used consistently, the between-run precision across a 6-12 month timespan (the typical interval for monitoring DTC patients) is less than 10%, yet the between-method variability seen for some TgAb-negative patients (shown in Figure 7A by red lines) can be greater than 30 percent [31]. These differences likely reflect different method specificities for detecting heterogeneous serum Tg isoforms. Clearly this magnitude of between-method difference has the potential to disrupt serial Tg monitoring and could negatively impact clinical management should a change in Tg method be made without re-baselining the Tg level [10,24,58,72,805]. In recognition of the differences between Tg methods, current guidelines stress the critical importance of using the same Tg method (and preferably the same laboratory) to monitor the serum Tg trend in DTC patients [72].

 

High-Dose Hook Effect

 

Tumor marker tests employing IMA methods can be prone to so-called "high-dose hook effects", whereby very high antigen concentrations can overwhelm the binding capacity of the monoclonal antibody reagents leading to a falsely normal/low value [9,831-834]. This phenomenon reduces the ability of the endogenous analyte to form a bridge between the capture and signal monoclonals resulting in an inappropriately low signal [9,831,835,836]. Manufacturers have largely overcome hook problems by adopting a two-step procedure, whereby a wash step is used to remove unbound antigen after the first incubation of specimen with the capture monoclonal antibody before introducing the labeled monoclonal followed by a second incubation when signal binds captured antigen [790,832]. When using any particular IMA method, it is primarily the laboratory’s responsibility to determine whether a hook effect is likely to generate falsely normal or low values.

 

Approaches for detecting and overcoming hook effects occurring with IMA methods are:

  • Routinely run each specimen at two dilutions. For example, the value obtained with a 1/5 or 1/10 dilution of the test serum would, if a hook effect were present, be higher than that obtained with an undiluted sample.
  • To carry out appropriate dilution studies to rule out a possible hook effect when an unexpectedly low serum Tg value is encountered for a patient with known metastatic disease. In such cases, consultation with the physician may provide valuable information regarding this issue.
  • To perform a Tg recovery test. If there is a hook effect present, the recovery of added antigen (Tg) will produce an inappropriately low result.

 

 

Interferences with Tg Measurement

 

Heterophile Antibody (HAb) Interferences

As discussed for FT4 and TSH, HAb, including human anti-mouse antibodies (HAMA) and Rheumatoid Factor (RF), interferes selectively with IMA but not RIA or Tg-LC-MS/MS methodologies [295,296,318,323,324,331,332,761,837]. HAb interferences are thought to reflect the binding of human immunoglobulins in the serum specimen to the murine-derived monoclonal antibody IMA reagents.  The rabbit polyclonal antibodies (PAb) used for Tg-RIA methods are not susceptible to this problem. In most cases HAb interferences are characterized by a false-positive Tg-IMA result [323,324,331,784], although falsely-low Tg-IMA results have also been reported [332].

 

Tg Autoantibody (TgAb) Interferences

TgAb interference with Tg measurement remains the major problem that limits the clinical utility of Tg testing. TgAb has the potential to undermine the clinical reliability of Tg measurements by both in-vitro mechanisms (epitope masking/low recoveries) [10,760,796,838, 839] and/or in-vivo mechanisms (enhanced TgAb-mediated Tg clearance) [677,840-842], irrespective of the Tg methodology used. There appears to be no threshold TgAb concentration that precludes TgAb interference [9,10,24,31,44,72,746,796,830]. High TgAb concentrations do not necessarily interfere, whereas low TgAb may profoundly interfere [9,31,44,761,795,796,830,839,843-846]. The Tg recovery approach is not reliable for detecting TgAb interference [10,752,839].

 

 

In-vitro Mechanisms of TgAb Interference.

TgAb interferes with Tg testing in a qualitative, quantitative and method-dependent manner [44,761,796,838,847,848]. The potential for in vitro interference is multifactorial and depends not only on the assay methodology (IMA, RIA or LC-MS/MS), but also the concentration and epitope specificity of the patient's TgAb [10,761,844]. RIA methodology appears to quantify total Tg (free Tg + TgAb-bound Tg) whereas IMA primarily detects only the free Tg moiety - Tg molecules whose epitopes are not masked by TgAb complexing. Steric masking of Tg epitopes is the reason why TgAb interference with IMA methodology is always unidirectional (underestimation), and why a low Tg-IMA/Tg-RIA ratio has been used to indicate TgAb interference [31,44,713,797,849,850]. The new Tg-LC-MS/MS methodology uses trypsin digestion of Tg-TgAb complexes to liberate a Tg proteotypic peptide. This conceptually attractive approach was primarily developed to overcome TgAb interference with IMA and thereby eliminate falsely low/undetectable Tg-IMA results that can mask disease. However, recent studies have reported a high percentage (>40%) of TgAb-positive DTC patients with structural disease who have paradoxically undetectable Tg-LC-MS/MS [31,799-801]. The reason why LC-MS/MS fails to detect Tg despite disease when TgAb is present needs further study. Possibilities to investigate include, tumor Tg polymorphisms that prevent the production of the Tg-specific tryptic peptide [21], suboptimal trypsinization of Tg-TgAb complexes, or Tg levels that are truly below detection because of increased clearance of Tg-TgAb complexes by the hepatic asialoglycoprotein receptor [677,840-842].

 

In-vivo Mechanisms of TgAb Interference.

A number of studies over past decades have suggested that the presence of TgAb enhances Tg metabolic clearance. In the 1967 Weigle et al showed enhanced clearance of endogenously 131I-labeled Tg in rabbits, after inducing TgAb by immunizing the animals with an immunogenic Tg preparation (840). Human studies of Tg and TgAb acute responses to sub-total thyroidectomy have also suggested that TgAb may increase Tg metabolic clearance (851). Changes (rise or fall) in TgAb versus Tg-RIA concentrations are typically concordant and appropriate for clinical status, whereas the direction of change of Tg-IMA is typically discordant with not only TgAb but also Tg-RIA and clinical status (31,32,44,713,798). In general, the change in TgAb concentrations tends to be steeper than for Tg-RIA (713), as would be consistent with TgAb-mediated Tg clearance. It may be that some TgAbs act as "sweeper" antibodies that facilitate clearance of antigen (842,852-854).

Figure 9 Serial TgAb, Tg-RIA and Tg-IMA concentrations in two DTC patients who underwent a change in TgAb status (panel A, negative to positive) or (panel B, positive to negative) before death from structural DTC. Panel A: When TgAb appeared de novo 2.5 years after initial treatment (thyroidectomy, Tx + RAI) for PTC a progressive fall in Tg-IMA to undetectable levels occurred together with an approximate 90 percent fall in Tg-RIA. Thereafter as disease exacerbated, TgAb remained elevated and Tg-IMA rose to parallel Tg-RIA but at an 80 percent lower concentration. Panel B. This patient was TgAb-positive at the time of initial Tx+RAI treatment at which time Tg-RIA was detectable and Tg-IMA was undetectable. Despite extensive disease, TgAb became undetectable 5 years after initial treatment. This change in TgAb status was associated with a rapid rise in Tg-IMA to parallel a steep increase in Tg-RIA with a doubling time <1 year before demise.

Figure 9 provides insights on the influence of TgAb on Tg measurements. These two DTC patients who eventually died of structural disease, illustrate how changes in TgAb status (Panel A-TgAb-negative to TgAb-positive versus Panel B- TgAb-positive to TgAb-negative) can produce Tg method discordances. These patients also serve to illustrate how disparate TgAb versus Tg responses can be associated with a poor prognosis and emphasize why a Tg measurement cannot be interpreted without knowing the TgAb status of the specimen (72). The de novo appearance of TgAb in the patient shown in Figure 9A either reflects a change in tumor-derived heterogeneity (secretion of a more immunogenic Tg molecule), or immune system recognition of tumor-derived Tg. In the patient shown in Figure 9B, TgAb was lost despite exacerbation of disease. This TgAb loss could be a response to the decrease in normally iodinated Tg antigen as normal remnant tissue was destroyed by RAI, at the same time as poorly iodinated (less immunogenic) tumor-derived Tg was rising with exacerbation of disease.

TgAb interference with Tg-RIA.

Tg-RIA methodology is based on Tg antigen (from serum or added 125I-Tg tracer) competing for a low concentration of polyclonal (rabbit) Tg antibody (PAb). After incubation, the Tg-PAb complex is precipitated and the serum antigen concentration quantified as an inverse relationship to the 125I-Tg in the precipitate. The first Tg-RIAs developed in the 1970s were very insensitive (~2 µg/L) (4,855). Over subsequent decades some Tg-RIAs have achieved first-generation functional sensitivity (FS = 0.5 µg/L) by using a 48-hour pre-incubation before adding a high specific activity 125I-Tg tracer (856,857). The use of a high affinity PAb (858) coupled with a species-specific second antibody minimizes TgAb interference. Resistance to TgAb interference is evidenced by appropriately normal Tg-RIA values for TgAb-positive euthyroid controls (10) and detectable Tg-RIA for TgAb-positive DTC patients with structural disease (Figures 7B and 8) (31).  The clinical performance of this Tg-RIA contrasts with IMA methods that report paradoxically undetectable serum Tg for some TgAb-positive normal euthyroid subjects (10) as well as TgAb-positive Graves' hyperthyroid patients (794) TgAb-positive patients with structural disease (Figures 7B and 8) (10). It should be noted that the propensity of TgAb to interfere with Tg-RIA determinations and cause underestimation (859) or overestimation (847,860) depends on not only the assay formulation but also patient-specific interactions between the endogenous Tg and TgAb in the specimen and the exogenous RIA reagents (848).

TgAb interference with Tg-IMA.

Non-competitive IMA methodology is based on a two-site reaction that involves antigen capture by a solid-phase monoclonal antibody (MAb) followed by addition of a labeled MAb that targets different epitopes of the captured antigen (377). TgAb interferes with IMA methodology by steric inhibition. Specifically, when the Tg epitope(s) necessary for binding to the IMA monoclonals are blocked by TgAb complexing, the 2-site reaction cannot take place and the test antigen is reported as falsely low or undetectable. This mechanism involving epitope masking is supported by recovery studies (data not shown). Clinically, the Tg-IMA underestimation caused by TgAb interference is evident from paradoxically low/undetectable Tg-IMA seen for TgAb-positive normal controls (10), patients with Graves' hyperthyroidism (794) and DTC patients with active disease (Figures 7B and 8) (9,10,44,45,752,755,846,861-863). High Tg concentrations can overwhelm the TgAb binding capacity rendering Tg-IMA concentrations detectable and lessening the degree of interference (31,44). It follows that as Tg concentrations rise, more Tg is free, the influence of TgAb lessens and the discordance between Tg-IMA and Tg-RIA disappears (Figure 9) (31,44). Although some IMA methods have claimed to overcome TgAb interference by using monoclonal antibodies directed against specific epitopes not involved in thyroid autoimmunity (790,864), this approach does not overcome TgAb interferences in clinical practice, possibly because less restricted TgAb epitopes are more often associated with thyroid carcinoma than with autoimmune thyroid conditions (746,752,755,862,865).

 

 

TgAb Interference with Tg LC-MS/MS.

The new LC-MS/MS methods measure Tg as a Tg-specific peptide(s) generated after trypsinization of serum containing Tg-TgAb complexes (16,21,790,866). Currently LC-MS/MS methods only have first-generation functional sensitivity (FS ~ 0.5 µg/L) (19,20,799). Tg-LC-MS/MS methodology has been shown free from HAb/HAMA interferences (837), and is being promoted as being free from TgAb interference (19,20,799). However, the reliability of using LC-MS/MS to detect Tg in the presence of TgAb is currently questionable. A number of studies have reported that over 40 percent of TgAb-positive patients with structural disease have paradoxically undetectable Tg-LC-MS/MS values (31,799-801). The most recent study concluded that Tg-LC-MS/MS offers no diagnostic advantage over 2G-Tg-IMA when TgAb is present (801). This study also confirmed earlier observations (867) that the higher the TgAb concentration, the more likely that Tg-LC-MS/MS would be undetectable despite disease (801). An inverse relationship between TgAb concentration and Tg-LC-MS/MS detectability would be expected if the presence of TgAb enhanced Tg clearance in vivo (see above).

 

Use of the TgAb Trend as a Surrogate DTC Tumor-Marker (Table 2)

IIt is now generally recognized that the serum TgAb concentration can be used as a surrogate tumor-marker for TgAb-positive DTC patients in whom the reliability of Tg testing is compromised by TgAb interference [Figures 9 and 10] (24,32,45,72,743,761-764,766,796,868-874). Following successful surgery (± RAI treatment), TgAb concentrations typically decline progressively over subsequent months, and may become undetectable during the first few post-operative years as a result of reduced Tg antigen stimulation of the immune system (32,44,72,762-766,870,875). The time needed for TgAb to become undetectable is inversely related to TgAb concentration around the time of initial treatment (32).  It should be noted that in the early post-operative period a significant percentage (~5%) of TgAb-negative patients develop transient de novo TgAb-positivity, presumably a response to Tg antigen released by surgical trauma (876). Such TgAb-negative to TgAb-positive conversions is one reason why Guidelines mandate that TgAb be measured with every Tg test  (45,72). Transient rises in TgAb may be seen in response to the acute release of Tg following thyroid surgery (877,878), fine needle aspiration biopsy (879,880) or more chronically (months) in response to radiolytic damage following RAI treatment (759,761,881-884). Patients exhibiting a TgAb decline of more than 50 percent by the end of the first post-operative year have been shown to have a low recurrence risk (762,874,876,885,886). In contrast, patients with persistent/recurrent disease may exhibit only a marginal TgAb decline, or have stable or rising TgAb (760,762,764,796,868,874). In fact, a rise, or de novo appearance of, TgAb, is an indication of persistent/recurrent disease (Figure 9A) (9,10,32,44,745,747,753,762-764,796,850,873,887). Because TgAb tests differ in sensitivity and specificity (44,45,745,753,888,889) (Figure 6), it is essential that serum TgAb concentrations be measured using the same manufacturers method and preferably the same laboratory (10,44,45,72,710,745,747,753,796,888,890,891).

Table 3--Clinical Significance of Changes in TgAb Concentrations:

1.    Approximately 25 percent of DTC patients have TgAb detected before or within three months of surgery [713,760]. TgAb prevalence in DTC patients is double that of the general population [452,713].

2.    Pre-operative TgAb-positivity is a risk factor for PTC in nodules with indeterminate cytology [892-895].

3.    The post-operative trend in TgAb (measured with the same method and preferably by the same laboratory) can be a useful surrogate tumor marker. A declining TgAb trend is a good prognostic sign, whereas a stable or rising TgAb may indicate persistent/recurrent disease [24,32,45,72,743,762-764,766,796,868-870,872-874].

4.    After successful treatment for DTC, TgAb (and Tg-RIA) concentrations typically fall more than 50% in first post-operative year and continue to fall in subsequent months-years, often becoming undetectable within a median time of four years [32,760,761,876].

5.    With successful treatment of disease, serum Tg-RIA typically becomes undetectable (< 0.5 µg/L) before TgAb [32,896].

6.    The time needed for a TgAb-positive patient to become TgAb-negative in response to successful treatment is proportional to the initial TgAb concentration, perhaps

7.    Approximately 10 percent of TgAb-negative DTC patients develop TgAb-positivity during post-operative monitoring [850], necessitating TgAb measurement with every Tg test [45,72].

8.    Most (75 %) TgAb-negative to TgAb-positive conversions are transient (months) and occur in response to the release of Tg antigen by surgical trauma [677,877], fine-needle biopsy [880] or RAI treatment [759,761,881-884].

9.    Approximately 3 percent of TgAb-negative DTC patients exhibit a de novo TgAb appearance more than one year following thyroidectomy without an initiating factor (surgery, biopsy or RAI treatment). Such TgAb-negative to TgAb-positive conversions are often associated with the presence representing the long-lived memory of plasma cells [32,896,897]. of metastatic disease, such as illustrated in Figure 9A [763,887].

10.  The de novo appearance of TgAb is typically associated with a rapid fall in Tg-IMA, often to undetectability, as a result of TgAb interference (Figure 9A). TgAb interference is less apparent when Tg-IMA is high before a TgAb appearance, because a high Tg concentration can saturate TgAb binding sites and reduce interference  [31,44].

11.  When serum Tg (RIA or IMA) persists after TgAb disappearance (~3% of cases) risk for disease remains (Figure 9B).

 

 

 

The Use of Serum Tg for Monitoring Patients with DTC

 

Over the past decade, the incidence of DTC has substantially risen partly as a result of detecting small thyroid nodules and micropapillary cancers (72,898-900) by ultrasound and other anatomic imaging modalities used for nonthyroidal purposes (901-904). Although most DTC patients are rendered disease-free by their initial surgery, overall approximately 15 percent of patients experience recurrences and approximately 5 percent die from disease-related complications (790,905-908). A risk-stratified approach to diagnosis and treatment is now recommended by current guidelines (72,785,787,908). In most cases, persistent/recurrent disease is detected within the first five post-operative years, although recurrences can occur decades after initial surgery, necessitating life-long monitoring for recurrence (906,907). Since most patients have a low pre-test probability for disease, protocols for follow-up need a high negative predictive value (NPV) to eliminate unnecessary testing, as well as a high positive predictive value (PPV) for identifying patients with persistent/recurrent disease. Serum Tg testing is generally recognized more sensitive for detecting disease than diagnostic 131I whole body scanning (909-912). It is recommended that biochemical testing (serum Tg+TgAb) be used in conjunction with periodic ultrasound (72,787,912,913). The persistent technical limitations of Tg and TgAb measurements necessitate close physician-laboratory cooperation.

 

The majority (~75%) of DTC patients have no Tg antibodies detected (713). In the absence of TgAb, four factors primarily influence the interpretation of serum Tg concentrations: (1) the mass of thyroid tissue present (normal tissue + tumor); (2) The intrinsic ability the tumor to secrete Tg; (3) the presence of any inflammation of, or injury to, thyroid tissue, such as following fine needle aspiration biopsy, surgery, RAI therapy or thyroiditis; and (4) the degree of TSH receptor stimulation by TSH, hCG or TSAb (24). In the presence of TgAb, interference with Tg measurement remains a problem necessitating a shift in focus from monitoring serum Tg as the primary tumor-marker, to monitoring the serum TgAb concentration as a surrogate tumor-marker.

 

Serum Tg Reference Ranges

 

The serum Tg reference range in adults approximates 3-40 µg/L (24,914). Serum Tg is higher in newborn infants but falls to the adult range after two years of age (915,916). However, because most Tg testing is made following surgery (thyroidectomy or lobectomy) for DTC, the Tg reference range is only relevant in the preoperative period. Tg methods can report up to 2-fold differences in numeric values for the same serum specimen (32). Between-method variability reflects differences in assay standardization and specificity for recognizing different serum Tg isoforms (10,58,825-827). When evaluating a thyroidectomized patient, the reference range of the assay should be adjusted for thyroid mass (thyroidectomy versus lobectomy) as well as the TSH status of the patient (24,882).

 

When using a 2G-Tg-IMA method standardized directly against the International Reference Preparation CRM-457, Tg should be detectable in all sera from TgAb-negative normal euthyroid subjects. Although the intra-individual serum Tg variability is relatively narrow (CV ~15%) (442,825), the Tg population reference range for TgAb-negative euthyroid subjects is broad, (~ 3 to 40 µg/L) (30,58,817,914). It follows that 1 gram of normal thyroid tissue results in ~1.0 µg/L Tg in the circulation under euthyroid TSH conditions (24,917,918). Following a lobectomy, euthyroid patients should be evaluated using a mass-adjusted reference range (1.5 - 20 µg/L). The range should be lowered a further 50 percent (0.75 - 10 µg/L) during TSH-suppression (24,882). After thyroidectomy, the typical 1-2 gram thyroid remnant (790,919) would be expected to produce a serum Tg below 2 µg/L (with a non-elevated TSH) (809,810). By this same reasoning, truly athyreotic patients would be expected to have no Tg detected irrespective of their TSH status (24).


Pre-operative Tg Measurement

 

An elevated Tg is a non-specific indicator of thyroid pathology and cannot be used to diagnose malignancy. However, a number of studies have reported that a Tg elevation, detected decades before a DTC diagnosis, is a risk factor for thyroid malignancy (920-926). This suggests that most thyroid cancers secrete Tg protein to an equal or greater degree than normal thyroid tissue, underscoring the importance of Tg as a DTC tumor marker (927). Approximately 50 percent of DTC patients have an elevated preoperative serum Tg, the highest serum Tg concentrations are seen in Follicular > Hurthle > Papillary (927). Up to one-third of tumors may be poor Tg secretors relative to tumor mass, especially tumors containing the BRAF mutation associated with reduced expression of Tg protein (928). Although current guidelines do not recommend routine pre-operative serum Tg measurement (72,782), some believe that a preoperative serum Tg (drawn before or more than two weeks after FNA) can provide information regarding the tumor’s intrinsic ability to secrete Tg and thus aid with the interpretation of postoperative Tg changes (929,930). For example, knowing that a tumor is an inefficient Tg secretor could prompt a physician to focus more on anatomic imaging and less on postoperative Tg monitoring (928,931).

 

Post-operative Tg measurement - First Post-Operative Year

 

Because TSH exerts such a strong influence on serum Tg concentrations it is important to promptly initiate thyroid hormone therapy after surgery to establish a stable post-operative Tg baseline to begin biochemical monitoring (882). When surgery is followed by RAI treatment it may take time (months) to establish a stable Tg baseline because the Tg rises in response to TSH-stimulation may be augmented by Tg release from radiolytic damage. Short-term rhTSH stimulation is expected to produce an approximate 10-fold serum Tg elevation (412), whereas chronic endogenous TSH stimulation following thyroid hormone withdrawal results in an approximate 20-fold serum Tg rise (811). Serum Tg measurements performed as early as 6 to 8 weeks after thyroidectomy have been shown to have prognostic value - the higher the serum Tg the greater the risk of persistent/recurrent disease (813,895,932-940). Since the half-life of Tg in the circulation approximates 3 days (941), the acute Tg release resulting from the surgical injury and healing of surgical margins should largely resolve within the first six months, provided that post-operative thyroid hormone therapy prevents TSH from rising. Patients who receive RAI for remnant ablation may exhibit a slow Tg decline over subsequent years, presumably reflecting the long-term radiolytic destruction of remnant tissue (942,943).

 

The Tg secretion expected from the ~1 gram of normal remnant tissue left after thyroidectomy (790,919), is expected to result in a serum Tg concentration ~1.0 µg/L under non-elevated TSH conditions (24). It should be noted that many thyroidectomized patients have a low serum Tg (0.10 – 0.99 µg/L) detected by 2G-Tg-IMA. A recent study found that in the first six months following thyroidectomy (without RAI treatment) disease-free PTC patients had a serum Tg nadir < 0.5 µg/L when TSH was maintained below 0.5 mIU/L (32,809,810). This is consistent with earlier studies using receiver operator curve (ROC) analysis that found a 6-week serum Tg of <1.0 µg/L, when measured during TSH suppression had a 98 percent negative predictive value (NPV) (although the positive predictive value (PPV) was only 43 percent) (940).

 

 Long-term Tg monitoring (without TSH stimulation)

 

The higher the post-operative serum Tg measured without TSH stimulation, the greater the risk for persistent/recurrent disease (813,932-940). If a stable TSH is maintained (≤0.5 mIU/L) (32,810), changes in the serum Tg will reflect changes in tumor mass. Under these conditions a rising Tg would be suspicious for tumor recurrence whereas a declining Tg levels suggests the absence or regression of disease. Now that sensitive 2G-Tg-IMA methods have become the standard of care, the trend in serum Tg, measured without TSH stimulation, is a more reliable indicator for disease status than using a fixed Tg cutoff value to assess disease (32,72,413,785,806,911,940,944-949). It is the degree of Tg elevation, not merely a "detectable" Tg, that is the risk factor for disease, since Tg “detectability” is merely determined by the assay FS (58,783,807,810,816). As with other tumor-markers such as Calcitonin, the Tg doubling time, measured without TSH stimulation, can be used as a prognostic marker that has an inverse relationship to mortality (809,949-956).  However, between-method variability necessitates that the serum Tg trend be established using the same method, and preferably the same laboratory. One approach used to mitigate between-run imprecision and improve the reliability of establishing the Tg trend has been to measure the current specimen concurrently (in the same run) with an archived specimen from the patient, thereby eliminating run-to-run variability and increasing the confidence for detecting small changes in serum Tg (9,804).

 

Serum Tg responses to TSH Stimulation

 

The degree of tumor differentiation determines the presence and density of TSH receptors that in large part determine the magnitude of the serum Tg response to TSH stimulation (928,931,957,958). The serum Tg rise in response to endogenous TSH (thyroid hormone withdrawal) is twice that seen with short-term rhTSH stimulation (~20-fold versus ~10-fold, respectively) (593,758,811,819,959). RhTSH administration was adopted as a standardized approach for stimulating serum Tg into the measureable range of the insensitive first-generation tests (296,593,758,782,811,812). A consensus rhTSH-stimulated serum Tg cut-off of ≥2.0 µg/L, measured 72 hours after the second dose of rhTSH, was found to be a risk factor for disease (758,811). A "positive" rhTSH response had a higher NPV (>95 percent) than basal (unstimulated) Tg measured by an insensitive first-generation test (813,818,819,911,937,940,945,946,948,960,961). However, a negative rhTSH test did not guarantee the absence of tumor (811,819,960). Furthermore, the reliability of adopting a fixed numeric rhTSH-Tg cut-off value for a positive response is problematic, given that different methods can report different numeric Tg values for the same specimen (Figure 7) (10,58). Other variables include differences in the dose of rhTSH delivered relative to absorption from the injection site as well as the surface area and age of the patient (962-965). One critical variable is the TSH sensitivity of tumor tissues, with poorly differentiated tumors having blunted TSH-mediated Tg responses (928,958,966). When using a more sensitive 2G-Tg-IMA, an undetectable basal Tg (<0.10 µg/L) had a comparable NPV to rhTSH stimulation, and was rarely associated with a "positive" rhTSH-stimulated response (>2.0 µg/L) (58,296,416,792,806,807,814,816,967,968). This relationship would be expected, given the strong relationship between basal Tg and rhTSH-stimulated Tg values (296,816). As 2G-Tg-IMA methods have become the standard of care, it became apparent the rhTSH-stimulated Tg value provides no additional information over and above a basal Tg measured by 2nd generation assay (58,72,296,416,792,807,814,816,967).

 

One important use of rhTSH-stimulated Tg testing remains - that as a test for HAb/HAMA/RF interferences. Specifically when the Tg-IMA value appears clinically inappropriate (usually high), an absent rhTSH-stimulated Tg response suggests interference, and a blocker tube test is indicated (296). An alternative reason for an absent/blunted rhTSH-stimulated response is the presence of TgAb (816). A blunted rhTSH-stimulated Tg response might be expected if TgAb enhanced the clearance of Tg-TgAb complexes (794,840,842,851).

Tg Measurement in FNA Needle Washouts (FNA-Tg)

 

Because Tg protein is tissue-specific, the detection of Tg in non-thyroidal tissues or fluids (such as pleural fluid) indicates the presence of metastatic thyroid cancer (779). Struma ovarii is the only (rare) condition in which the Tg in the circulation does not originate from the thyroid (969,970). Cystic thyroid nodules are commonly encountered in clinical practice, the large majority arising from follicular epithelium and the minority from parathyroid epithelium. A high concentration of Tg or parathyroid hormone (PTH) measured in the cyst fluid provides a reliable indicator of the tissue origin of the cyst (thyroid versus parathyroid, respectively), information critical for surgical decision-making (779,971). Lymph node metastases are found in up to 50 percent of patients with papillary cancers but only 20 percent of follicular cancers (972-975). High-resolution ultrasound has now become an important component of the protocols used for postoperative surveillance for recurrence (72,593,758). Although ultrasound characteristics are helpful for distinguishing benign reactive lymph nodes from those suspicious for malignancy, the finding of Tg in the needle washout of a lymph node biopsy has higher diagnostic accuracy than the ultrasound appearance (976-988). An FNA needle washout is now widely accepted as a useful adjunctive test for improving the diagnostic sensitivity of the cytological evaluation of a suspicious lymph node or thyroid mass (976-981,983,986,987,989). The current protocol for obtaining FNA-Tg samples recommends rinsing the biopsy needle in 1.0 mL of saline and sending this specimen to the laboratory for Tg analysis. In thyroidectomized patients a common cutoff value for a "positive" FNA-Tg result is 1.0 µg/L (980,987,990), however this cutoff can vary by assay and Institution (986,991). For investigations of suspicious lymph nodes in patients with an intact thyroid, a higher FNA-Tg cutoff value (~35-40 µg/L) is recommended (978,982). There is still controversy whether TgAb interferes with FNA-Tg analyses (979,992,993). It should be noted that when the serum TgAb concentration is very high and there is serum contamination of the FNA wash, the expected ~40-fold dilution in the wash fluid may be insufficient to lower TgAb below detection, and there is potential for TgAb to interfere with the FNA-Tg IMA test causing a falsely low/undetectable FNA-Tg result. The FNA needle wash-out procedure can also be used to detect Calcitonin in neck masses of patients with primary and metastatic medullary thyroid cancer (971,994-996), and FNA-PTH determinations may be useful for identifying lymph nodes arising from parathyroid tissue (971).

 

 THYROID SPECIFIC MRNAS USED AS THYROID TUMOR MARKERS

 

Reverse transcription-polymerase chain reaction (RT-PCR) has been used to detect thyroid specific mRNAs (Tg, TSHR, TPO and NIS) in the peripheral blood of patients with DTC (918,997-999). Initial studies suggested that circulating Tg mRNA might be employed as a useful tumor marker for thyroid cancer, especially in TgAb-positive patients in whom Tg measurements were subject to assay interference (1000,1001). More recently, this approach has been applied to the detection of NIS, TPO and TSH receptor (TSHR) mRNA (1001-1005). Although some studies have suggested that thyroid specific mRNA measurements could be useful for cancer diagnosis and detecting recurrent disease, most studies have concluded that they offer no advantages over sensitive serum Tg measurements (918,1001,1006,1007). Further, the recent report of false positive Tg mRNA results in patients with congenital athyreosis (1008) suggests that Tg mRNA can arise as an assay artifact originating from non-thyroid tissues, or illegitimate transcription (1009,1010). Conversely, false negative Tg mRNA results have also been observed in patients with documented metastatic disease (1011-1013). Although Tg, TSHR, NIS and TPO are generally considered “thyroid specific” proteins, mRNAs for these antigens have been detected in a number of non-thyroidal tissues such as lymphocytes, leukocytes, kidney, hepatocytes, brown fat and skin (625,1014-1019). Additional sources of variability in mRNA analyses relate to the use of primers that detect splice variants, sample-handling techniques that introduce variability, and difficulties in quantifying the mRNA detected (1006,1011). There is now a general consensus is that thyroid specific mRNA measurements presently lack the optimal specificity and practicality to be useful tumor markers (918,1001,1006). Finally, the growing number of reports of functional TSH receptors and Tg mRNA present in non-thyroidal tissues further suggests that these mRNA measurements will have limited clinical utility in the management of DTC in the future (625,1017-1019).

 

 

 

REFERENCES

  1. Benotti J, Benotti N. Protein-bound iodine, total iodine and butanol extractable iodine by partial automation. Clin Chem 1963; 9:408-416
  2. Chopra IJ. A radioimmunoassay for measurement of thyroxine in unextracted serum. J Clin Endocrinol Metab 1972; 34:938-947
  3. Chopra IJ, Ho RS, Lam R. An improved radioimmunoassay of triiodothyronine in serum: Its application to clinical and physiological studies. J Lab Clin Med 1972; 80:729-?
  4. Van Herle AJ, Uller RP, Matthews NL, Brown J. Radioimmunoassay for measurement of thyroglobulin in human serum. J Clin Invest 1973; 52:1320-1327
  5. Nicoloff JT, Spencer CA. Clinical review 12: The use and misuse of the sensitive thyrotropin assays. J Clin Endocrinol Metab 1990; 71:553-558
  6. Ekins R. Measurement of free hormones in blood. Endocrine Rev 1990; 11:5-46
  7. Spencer CA, LoPresti JS, Patel A, Guttler RB, Eigen A, Shen D, Gray D, Nicoloff JT. Applications of a new chemiluminometric thyrotropin assay to subnormal measurement. J Clin Endocrinol Metab 1990; 70:453-460
  8. Spencer CA, Takeuchi M, Kazarosyan M. Current status and performance goals for serum thyrotropin (TSH) assays. Clinical Chemistry 1996; 42:141-145
  9. Spencer CA, Takeuchi M, Kazarosyan M. Current Status and Performance Goals for Serum Thyroglobulin Assays. Clin Chem 1996; 42:164-173
  10. Spencer CA, Bergoglio LM, Kazarosyan M, Fatemi S, Lopresti JS. Clinical Impact of Thyroglobulin (Tg) and Tg autoantibody Method Differences on the Management of patients with Differentiated Thyroid Carcinomas. J Clin Endocrinol Metab 2005; 90:5566-5575
  11. Giovanella L, Treglia G, Sadeghi R, Trimboli P, Ceriani L, Verburg FA. Unstimulated high-sensitive thyroglobulin in follow-up of differentiated thyroid cancer patients: a meta-analysis. J Clin Endocrinol Metab 2014; 99:440-447
  12. Thienpont LM, De Brabandere VI, Stockl D, De Leenheer AP. Development of a new method for the determination of thyroxine in serum based on isotope dilution gas chromatography mass spectrometry. Biological mass spectrometry 1994; 23:475-482
  13. Thienpont LM, Fierens C, De Leenheer AP, Przywara L. Isotope dilution-gas chromatography/mass spectrometry and liquid chromatography/electrospray ionization-tandem mass spectrometry for the determination of triiodo-L-thyronine in serum. Rapid communications in mass spectrometry : RCM 1999; 13:1924-1931
  14. Kahric-Janicic N, Soldin SJ, Soldin OP, West T, Gu J, Jonklaas J. Tandem mass spectrometry improves the accuracy of free thyroxine measurements during pregnancy. Thyroid 2007; 17:303-311
  15. Thienpont LM, Beastall G, Christofides ND, Faix JD, Ieiri T, Jarrige V, Miller WG, Miller R, Nelson JC, Ronin C, Ross HA, Rottmann M, Thijssen JH, Toussaint B. Proposal of a candidate international conventional reference measurement procedure for free thyroxine in serum. Clin Chem Lab Med 2007; 45:934-936
  16. Hoofnagle AN, Becker JO, Wener MH, Heinecke JW. Quantification of thyroglobulin, a low-abundance serum protein, by immunoaffinity peptide enrichment and tandem mass spectrometry. Clin Chem 2008; 54:1796-1804
  17. Jonklaas J, Kahric-Janicic N, Soldin OP, Soldin SJ. Correlations of free thyroid hormones measured by tandem mass spectrometry and immunoassay with thyroid-stimulating hormone across 4 patient populations. Clin Chem 2009; 55:1380-1388
  18. van Deventer HE, Mendu DR, Remaley AT, Soldin SJ. Inverse log-linear relationship between thyroid-stimulating hormone and free thyroxine measured by direct analog immunoassay and tandem mass spectrometry. Clin Chem 2011; 57:122-127
  19. Clarke NJ, Zhang Y, Reitz RE. A novel mass spectrometry-based assay for the accurate measurement of thyroglobulin from patient samples containing antithyroglobulin autoantibodies. J Investig Med 2012; 60:1157-1163
  20. Kushnir MM, Rockwood AL, Roberts WL, Abraham D, Hoofnagle AN, Meikle AW. Measurement of thyroglobulin by liquid chromatography-tandem mass spectrometry in serum and plasma in the presence of antithyroglobulin autoantibodies. Clin Chem 2013; 59:982-990
  21. Hoofnagle AN, Roth MY. Clinical review: improving the measurement of serum thyroglobulin with mass spectrometry. J Clin Endocrinol Metab 2013; 98:1343-1352
  22. Thienpont LM, Van Uytfanghe K, Poppe K, Velkeniers B. Determination of free thyroid hormones. Best Pract Res Clin Endocrinol Metab 2013; 27:689-700
  23. Gounden V, Jonklaas J, Soldin SJ. A pilot study: subclinical hypothyroidism and free thyroid hormone measurement by immunoassay and mass spectrometry. Clin Chim Acta 2014; 430:121-124
  24. Baloch Z, Carayon P, Conte-Devolx B, Demers LM, Feldt-Rasmussen U, Henry JF, LiVosli VA, Niccoli-Sire P, John R, Ruf J, Smyth PP, Spencer CA, Stockigt JR. Laboratory medicine practice guidelines. Laboratory support for the diagnosis and monitoring of thyroid disease. Thyroid 2003; 13:3-126
  25. Dufour DR. Laboratory tests of thyroid function: uses and limitations. Endocr Metab Clin North Am 2007; 36:155-169
  26. Thienpont LM, Van Uytfanghe K, Marriot J, Stokes P, Siekmann L, Kessler A, Bunk D, Tai S. Metrologic traceability of total thyroxine measurements in human serum: efforts to establish a network of reference measurement laboratories. Clin Chem 2005; 51:161-168
  27. Yue B, Rockwood AL, Sandrock T, La'ulu SL, Kushnir MM, Meikle AW. Free thyroid hormones in serum by direct equilibrium dialysis and online solid-phase extraction--liquid chromatography/tandem mass spectrometry. Clin Chem 2008; 54:642-651
  28. Jonklaas J, Sathasivam A, Wang H, Gu J, Burman KD, Soldin SJ. Total and free thyroxine and triiodothyronine: measurement discrepancies, particularly in inpatients. Clin Biochem 2014; 47:1272-1278
  29. Thienpont LM, Van Uytfanghe K, Beastall G, Faix JD, Ieiri T, Miller WG, Nelson JC, Ronin C, Ross HA, Thijssen JH, Toussaint B. Report of the IFCC Working Group for Standardization of Thyroid Function Tests; part 1: thyroid-stimulating hormone. Clin Chem 2010; 56:902-911
  30. Spencer CA, Bergoglio LM, Kazarosyan M, Fatemi S, LoPresti JS. Clinical impact of thyroglobulin (Tg) and Tg autoantibody method differences on the management of patients with differentiated thyroid carcinomas. J Clin Endocrinol Metab 2005; 90:5566-5575
  31. Spencer C, Petrovic I, Fatemi S, LoPresti J. Serum thyroglobulin (Tg) monitoring of patients with differentiated thyroid cancer using sensitive (second-generation) immunometric assays can be disrupted by false-negative and false-positive serum thyroglobulin autoantibody misclassifications. J Clin Endocrinol Metab 2014; 99:4589-4599
  32. Spencer C, LoPresti J, Fatemi S. How sensitive (second-generation) thyroglobulin measurement is changing paradigms for monitoring patients with differentiated thyroid cancer, in the absence or presence of thyroglobulin autoantibodies. Curr Opin Endocrinol Diabetes Obes 2014; 21:394-404
  33. Giovanella L, Clark P, Chiovato L, Duntas LH, Elisei R, Feldt-Rasmussen U, Leenhardt L, Luster M, Schalin-Jantti C, Schott M, Seregni E, Rimmele H, Smit JW, Verburg FA. Thyroglobulin measurement using highly sensitive assays in patients with differentiated thyroid cancer: a clinical position paper. Eur J Endocrinol 2014; 171:R33-46
  34. Robbins J, Cheng SY, Gershengorn MC, Glinoer D, Cahnmann HJ, Edelnoch H. Thyroxine transport proteins of plasma. Molecular properties and biosynthesis. Recent Prog Horm Res 1978; 34:477-519
  35. Bartalena L, Robbins J. Thyroid hormone transport proteins. Clin Lab Med 1993; 13:583-598
  36. Schussler GC. The thyroxine-binding proteins. Thyroid 2000; 10:141-149
  37. Feldt-Rasmussen U. Analytical and clinical performance goals for testing autoantibodies to thyroperoxidase, thyroglobulin and thyrotropin receptor. Clin Chem 1996; 42:160-163
  38. Kamijo K. TSH-receptor antibody measurement in patients with various thyrotoxicosis and Hashimoto's thyroiditis: a comparison of two two-step assays, coated plate ELISA using porcine TSH-receptor and coated tube radioassay using human recombinant TSH-receptor. Endocr J 2003; 50:113-116
  39. Kamijo K. TSH-receptor antibodies determined by the first, second and third generation assays and thyroid-stimulating antibody in pregnant patients with Graves' disease. Endocr J 2007; 54:619-624
  40. Ajjan RA, Weetman AP. Techniques to quantify TSH receptor antibodies. Nat Clin Pract Endocrinol Metab 2008; 4:461-468
  41. Kamijo K, Murayama H, Uzu T, Togashi K, Olivo PD, Kahaly GJ. Similar clinical performance of a novel chimeric thyroid-stimulating hormone receptor bioassay and an automated thyroid-stimulating hormone receptor binding assay in Graves' disease. Thyroid 2011; 21:1295-1299
  42. Giuliani C, Cerrone D, Harii N, Thornton M, Kohn LD, Dagia NM, Bucci I, Carpentieri M, Di Nenno B, Di Blasio A, Vitti P, Monaco F, Napolitano G. A TSHR-LH/CGR chimera that measures functional thyroid-stimulating autoantibodies (TSAb) can predict remission or recurrence in Graves' patients undergoing antithyroid drug (ATD) treatment. J Clin Endocrinol Metab 2012; 97:1080-1087
  43. Diana T, Wuster C, Kanitz M, Kahaly GJ. Highly variable sensitivity of five binding and two bio-assays for TSH-receptor antibodies. J Endocrinol Invest 2016; 39:1159-1165
  44. Spencer C, Petrovic I, Fatemi S. Current thyroglobulin autoantibody (TgAb) assays often fail to detect interfering TgAb that can result in the reporting of falsely low/undetectable serum Tg IMA values for patients with differentiated thyroid cancer. J Clin Endocrinol Metab 2011; 96:1283-1291
  45. Verburg FA, Luster M, Cupini C, Chiovato L, Duntas L, Elisei R, Feldt-Rasmussen U, Rimmele H, Seregni E, Smit JW, Theimer C, Giovanella L. Implications of thyroglobulin antibody positivity in patients with differentiated thyroid cancer: a clinical position statement. Thyroid 2013; 23:1211-1225
  46. Hutfless S, Matos P, Talor MV, Caturegli P, Rose NR. Significance of prediagnostic thyroid antibodies in women with autoimmune thyroid disease. J Clin Endocrinol Metab 2011; 96:E1466-1471
  47. Balucan FS, Morshed SA, Davies TF. Thyroid autoantibodies in pregnancy: their role, regulation and clinical relevance. J Thyroid Res 2013; 2013:182472
  48. Morshed SA, Davies TF. Graves' Disease Mechanisms: The Role of Stimulating, Blocking, and Cleavage Region TSH Receptor Antibodies. Horm Metab Res 2015; 47:727-734
  49. Thienpont LM, Van Uytfanghe K, Van Houcke S. Standardization activities in the field of thyroid function tests: a status report. Clin Chem Lab Med 2010; 48:1577-1583
  50. Thienpont LM, Van Uytfanghe K, Beastall G, Faix JD, Ieiri T, Miller WG, Nelson JC, Ronin C, Ross HA, Thijssen JH, Toussaint B. Report of the IFCC Working Group for Standardization of Thyroid Function Tests; part 2: free thyroxine and free triiodothyronine. Clin Chem 2010; 56:912-920
  51. Van Houcke SK, Van Uytfanghe K, Shimizu E, Tani W, Umemoto M, Thienpont LM. IFCC international conventional reference procedure for the measurement of free thyroxine in serum: International Federation of Clinical Chemistry and Laboratory Medicine (IFCC) Working Group for Standardization of Thyroid Function Tests (WG-STFT)(1). Clin Chem Lab Med 2011; 49:1275-1281
  52. Thienpont LM, Van Uytfanghe K, Van Houcke S, Das B, Faix JD, MacKenzie F, Quinn FA, Rottmann M, Van den Bruel A. A Progress Report of the IFCC Committee for Standardization of Thyroid Function Tests. Eur Thyroid J 2014; 3:109-116
  53. Faix JD, Miller WG. Progress in standardizing and harmonizing thyroid function tests. Am J Clin Nutr 2016; 104 Suppl 3:913s-917s
  54. Thienpont LM, Beastall G, Christofides ND, Faix JD, Ieiri T, Miller WG, Miller R, Nelson JC, Ross HA, Ronin C, Rottmann M, Thijssen JH, Toussaint B. Measurement of free thyroxine in laboratory medicine--proposal of measurand definition. Clin Chem Lab Med 2007; 45:563-564
  55. Soldin OP, Soldin SJ. Thyroid hormone testing by tandem mass spectrometry. Clin Biochem 2011; 44:89-94
  56. Steele BW, Wang E, Klee GG, Thienpont LM, Soldin SJ, Sokoll LJ, Winter WE, Fuhrman SA, Elin RJ. Analytic bias of thyroid function tests: analysis of a College of American Pathologists fresh frozen serum pool by 3900 clinical laboratories. Arch Pathol Lab Med 2005; 129:310-317
  57. Beckett G, MacKenzie F. Thyroid guidelines - are thyroid-stimulating hormone assays fit for purpose? Ann Clin Biochem 2007; 44:203-208
  58. Schlumberger M, Hitzel A, Toubert ME, Corone C, Troalen F, Schlageter MH, Claustrat F, Koscielny S, Taieb D, Toubeau M, Bonichon F, Borson-Chazot F, Leenhardt L, Schvartz C, Dejax C, Brenot-Rossi I, Torlontano M, Tenenbaum F, Bardet S, Bussiere F, Girard JJ, Morel O, Schneegans O, Schlienger JL, Prost A, So D, Archambeaud F, Ricard M, Benhamou E. Comparison of seven serum thyroglobulin assays in the follow-up of papillary and follicular thyroid cancer patients. J Clin Endocrinol Metab 2007; 92:2487-2495
  59. Stockl D, Van Uytfanghe K, Van Aelst S, Thienpont LM. A statistical basis for harmonization of thyroid stimulating hormone immunoassays using a robust factor analysis model. Clin Chem Lab Med 2014; 52:965-972
  60. Vesper HW, Myers GL, Miller WG. Current practices and challenges in the standardization and harmonization of clinical laboratory tests. Am J Clin Nutr 2016; 104 Suppl 3:907s-912s
  61. Gharib H, Tuttle RM, Baskin HJ, Fish LH, Singer PA, McDermott MT. Subclinical thyroid dysfunction: A joint statement on management from the American Association of Clinical Endocrinologiss, the American Thyroid Association and the Endocrine Society. J Clin Endocrinol Metab 2005; 90
  62. Pacini F, Schlumberger M, Dralle H, Elisei R, Smit JW, Wiersinga W. European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur J Endocrinol 2006; 154:787-803
  63. Negro R, Beck-Peccoz P, Chiovato L, Garofalo P, Guglielmi R, Papini E, Tonacchera M, Vermiglio F, Vitti P, Zini M, Pinchera A. Hyperthyroidism and pregnancy. An Italian Thyroid Association (AIT) and Italian Association of Clinical Endocrinologists (AME) joint statement for clinical practice. J Endocrinol Invest 2011; 34:225-231
  64. Bahn RS, Burch HB, Cooper DS, Garber JR, Greenlee MC, Klein I, Laurberg P, McDougall IR, Montori VM, Rivkees SA, Ross DS, Sosa JA, Stan MN. Hyperthyroidism and other causes of thyrotoxicosis: management guidelines of the American Thyroid Association and American Association of Clinical Endocrinologists. Thyroid 2011; 21:593-646
  65. Takami H, Ito Y, Okamoto T, Yoshida A. Therapeutic strategy for differentiated thyroid carcinoma in Japan based on a newly established guideline managed by Japanese Society of Thyroid Surgeons and Japanese Association of Endocrine Surgeons. World J Surg 2011; 35:111-121
  66. Stagnaro-Green A, Abalovich M, Alexander E, Azizi F, Mestman J, Negro R, Nixon A, Pearce EN, Soldin OP, Sullivan S, Wiersinga W. Guidelines of the American Thyroid Association for the diagnosis and management of thyroid disease during pregnancy and postpartum. Thyroid 2011; 21:1081-1125
  67. De Groot L, Abalovich M, Alexander EK, Amino N, Barbour L, Cobin RH, Eastman CJ, Lazarus JH, Luton D, Mandel SJ, Mestman J, Rovet J, Sullivan S. Management of thyroid dysfunction during pregnancy and postpartum: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2012; 97:2543-2565
  68. Garber JR, Cobin RH, Gharib H, Hennessey JV, Klein I, Mechanick JI, Pessah-Pollack R, Singer PA, Woeber KA. Clinical practice guidelines for hypothyroidism in adults: cosponsored by the American Association of Clinical Endocrinologists and the American Thyroid Association. Thyroid 2012; 22:1200-1235
  69. Pearce S.H.S, Brabant G, Duntas L.H, Monzani F, Peeters R.P, Razvi S, Wemeau J. 2013 ETA Guideline: Management of Subclinical Hypothyroidism. Eur Thyroid J 2013; 2:215-228
  70. Jonklaas J, Bianco AC, Bauer AJ, Burman KD, Cappola AR, Celi FS, Cooper DS, Kim BW, Peeters RP, Rosenthal MS, Sawka AM. Guidelines for the treatment of hypothyroidism: prepared by the american thyroid association task force on thyroid hormone replacement. Thyroid 2014; 24:1670-1751
  71. Ross DS, Burch HB, Cooper DS, Greenlee MC, Laurberg P, Maia AL, Rivkees S, Samuels M, Sosa JA, Stan MN, Walter M. 2016 American Thyroid Association Guidelines for Diagnosis and Management of Hyperthyroidism and other causes of Thyrotoxicosis. Thyroid 2016;
  72. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, Schuff KG, Sherman SI, Sosa JA, Steward DL, Tuttle RM, Wartofsky L. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 2016; 26:1-133
  73. Gharib H, Papini E, Garber JR, Duick DS, Harrell RM, Hegedus L, Paschke R, Valcavi R, Vitti P. AMERICAN ASSOCIATION OF CLINICAL ENDOCRINOLOGISTS, AMERICAN COLLEGE OF ENDOCRINOLOGY, AND ASSOCIAZIONE MEDICI ENDOCRINOLOGI MEDICAL GUIDELINES FOR CLINICAL PRACTICE FOR THE DIAGNOSIS AND MANAGEMENT OF THYROID NODULES--2016 UPDATE. Endocr Pract 2016; 22:622-639
  74. Alexander EK, Pearce EN, Brent GA, Brown RS, Chen H, Dosiou C, Grobman W, Laurberg P, Lazarus JH, Mandel SJ, Peeters R, Sullivan S. 2016 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease during Pregnancy and the Postpartum. Thyroid 2017;
  75. Stockigt JR. Free thyroid hormone measurement. A critical appraisal. Endocrinol Metab Clin North Am 2001; 30:265-289
  76. Murphy BE, Pattee CJ, Gold A. Clinical evaluation of a new method for the determination of serum thyroxine. J Clin Endocrinol Metab 1966; 26:247-256
  77. Islam KN, Ihara M, Dong J, Kasagi N, Mori T, Ueda H. Micro open-sandwich ELISA to rapidly evaluate thyroid hormone concentration from serum samples. Bioanalysis 2010; 2:1683-1687
  78. Soldin OP, Tractenberg RE, Soldin SJ. Differences between measurements of T4 and T3 in pregnant and nonpregnant women using isotope dilution tandem mass spectrometry and immunoassays: are there clinical implications? Clin Chim Acta 2004; 347:61-69
  79. Soukhova N, Soldin OP, Soldin SJ. Isotope dilution tandem mass spectrometric method for T4/T3. Clin Chim Acta 2004; 343:185-190
  80. Thienpont LM, Van Uytfanghe K, Beastall G, Faix JD, Ieiri T, Miller WG, Nelson JC, Ronin C, Ross HA, Thijssen JH, Toussaint B. Report of the IFCC Working Group for Standardization of Thyroid Function Tests; part 3: total thyroxine and total triiodothyronine. Clin Chem 2010; 56:921-929
  81. Matsuda M, Sakata S, Komaki T, Nakamura s, Kojima N, Takuno H, Miura K. Effect of 8-anilino-1-napthalene sulfonic acid (ANS) on the interaction between thyroid hormone and anti-thyroid hormone antibodies. Clin Chim Acta 1989; 185:139-146
  82. Chopra IJ, Taing P, Mikus L. Direct determination of free triiodothyronine (T3) in undiluted serum by equilibrium dialysis/radioimmunoassay (RIA). Thyroid 1996; 6:255-259
  83. Zucchelli GC, Pilo A, Chiesa MR, Piro MA. Progress report on a national quality-control survey of triiodothyronine and thyroxin assay. Clin Chem 1984; 30:395-398
  84. Klee GG. Clinical usage recommendations and analytic performance goals for total and free triiodothyronine measurements. Clin Chem 1996; 42:155-159
  85. Karapitta CD, Sotiroudis TG, Papadimitriou A, Xenakis A. Homogeneous enzyme immunoassay for triiodothyronine in serum. Clin Chem 2001; 47:569-574
  86. Welsh KJ, Soldin SJ. DIAGNOSIS OF ENDOCRINE DISEASE: How reliable are free thyroid and total T3 hormone assays? Eur J Endocrinol 2016; 175:R255-r263
  87. Martel J, Despres N, Ahnadi CE, Lachance JF, Monticello JE, Fink G, Ardemagni A, Banfi G, Tovey J, Dykes P, John R, Jeffery J, Grant AM. Comparative multicentre study of a panel of thyroid tests using different automated immunoassay platforms and specimens at high risk of antibody interference. Clin Chem Lab Med 2000; 38:785-793
  88. Thienpont LM, Van Uytfanghe K, Marriott J, Stokes P, Siekmann L, Kessler A, Bunk D, Tai S. Feasibility study of the use of frozen human sera in split-sample comparison of immunoassays with candidate reference measurement procedures for total thyroxine and total triiodothyronine measurements. Clin Chem 2005; 51:2303-2311
  89. Zhou Q, Li S, Li X, Wang W, Wang Z. Comparability of five analytical systems for the determination of triiodothyronine, thyroxine and thyroid-stimulating hormone. Clin Chem Lab Med 2006; 44:1363-1366
  90. Van Houcke SK, Stepman HC, Thienpont LM, Fiers T, Stove V, Couck P, Anckaert E, Gorus F. Long-term stability of laboratory tests and practical implications for quality management. Clin Chem Lab Med 2013; 51:1227-1231
  91. Abduljabbar M, Al Shahri A, Afifi A. Is umbilical cord blood total thyroxin measurement effective in newborn screening for hypothyroidism? J Med Screen 2009; 16:119-123
  92. Cartwright D, O'Shea P, Rajanayagam O, Agostini M, Barker P, Moran C, Macchia E, Pinchera A, John R, Agha A, Ross HA, Chatterjee VK, Halsall DJ. Familial dysalbuminemic hyperthyroxinemia: a persistent diagnostic challenge. Clin Chem 2009; 55:1044-1046
  93. Ross HA, de Rijke YB, Sweep FC. Spuriously high free thyroxine values in familial dysalbuminemic hyperthyroxinemia. Clin Chem 2011; 57:524-525
  94. Pappa T, Ferrara AM, Refetoff S. Inherited defects of thyroxine-binding proteins. Best Pract Res Clin Endocrinol Metab 2015; 29:735-747
  95. Zouwail SA, O'Toole AM, Clark PM, Begley JP. Influence of thyroid hormone autoantibodies on 7 thyroid hormone assays. Clin Chem 2008; 54:927-928
  96. Giovanella L, Dorizzi RM, Keller F. A hypothyroid patient with increased free thyroid hormones. Clin Chem Lab Med 2008; 46:1650-1651
  97. Massart C, Elbadii S, Gibassier J, Coignard V, Rasandratana A. Anti-thyroxine and anti-triiodothyronine antibody interferences in one-step free triiodothyronine and free thyroxine immunoassays. Clin Chim Acta 2009; 401:175-176
  98. Loh TP, Leong SM, Loke KY, Deepak DS. Spuriously elevated free thyroxine associated with autoantibodies, a result of laboratory methodology: case report and literature review. Endocr Pract 2014; 20:e134-139
  99. Surks MI, Sievert R. Drugs and thyroid function. N Engl J Med 1995; 333:1688-1694
  100. Stockigt JR, Lim CF. Medications that distort in vitro tests of thyroid function, with particular reference to estimates of serum free thyroxine. Best Pract Res Clin Endocrinol Metab 2009; 23:753-767
  101. Kundra P, Burman KD. The effect of medications on thyroid function tests. The Medical clinics of North America 2012; 96:283-295
  102. Burr WA, Evans SE, Lee J, Prince HP, Ramsden DB. The ratio of thyroxine to thyroxine-binding globulin in the assessment of thyroid function. Clin Endocrinol (Oxf) 1979; 11:333-342
  103. Glinoer D, Fernandez-Deville M, Ermans AM. Use of direct thyroxine-binding globulin measurement in the evaluation of thyroid function. J Endocrinol Invest 1978; 1:329-335
  104. Lee RH, Spencer CA, Mestman JH, Miller EA, Petrovic I, Braverman LE, Goodwin TM. Free T4 immunoassays are flawed during pregnancy. Am J Obstet Gynecol 2009; 200:e1-6
  105. Van den Berghe G. Endocrine changes in critically ill patients. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society 1999; 9 Suppl A:77-81
  106. Wilson KL, Casey BM, McIntire DD, Cunningham FG. Is total thyroxine better than free thyroxine during pregnancy? Am J Obstet Gynecol 2014; 211:132.e131-136
  107. Sriphrapradang C, Bhasipol A. Differentiating Graves' disease from subacute thyroiditis using ratio of serum free triiodothyronine to free thyroxine. Annals of medicine and surgery (2012) 2016; 10:69-72
  108. Ito M, Toyoda N, Nomura E, Takamura Y, Amino N, Iwasaka T, Takamatsu J, Miyauchi A, Nishikawa M. Type 1 and type 2 iodothyronine deiodinases in the thyroid gland of patients with 3,5,3'-triiodothyronine-predominant Graves' disease. Eur J Endocrinol 2011; 164:95-100
  109. Yoshimura Noh J, Momotani N, Fukada S, Ito K, Miyauchi A, Amino N. Ratio of serum free triiodothyronine to free thyroxine in Graves' hyperthyroidism and thyrotoxicosis caused by painless thyroiditis. Endocr J 2005; 52:537-542
  110. Tagami T, Hagiwara H, Kimura T, Usui T, Shimatsu A, Naruse M. The incidence of gestational hyperthyroidism and postpartum thyroiditis in treated patients with Graves' disease. Thyroid 2007; 17:767-772
  111. Whitworth AS, Midgley JE, Wilkins TA. A comparison of free T4 and the ratio of total T4 to T4-binding globulin in serum through pregnancy. Clin Endocrinol (Oxf) 1982; 17:307-313
  112. Weeke J DL, Granlie K, Eskjaer Jensen S, Kjaerulff E, Laurberg P, Magnusson B,. A longitudinal study of serum TSH, and total and free iodothyronines during normal pregnancy. Acta Endocrinol (Copenh) 1982; 101:531-537
  113. Zhang X, Li C, Mao J, Wang W, Xie X, Peng S, Wang Z, Han C, Zhang X, Wang D, Fan C, Shan Z, Teng W. Gestation-Specific Changes in Maternal Thyroglobulin during Pregnancy and Lactation in an Iodine-Sufficient Region in China: A Longitudinal Study. Clin Endocrinol (Oxf) 2016;
  114. Korevaar TI, Chaker L, Medici M, de Rijke YB, Jaddoe VW, Steegers EA, Tiemeier H, Visser TJ, Peeters RP. Maternal total T4 during the first half of pregnancy: physiologic aspects and the risk of adverse outcomes in comparison with free T4. Clin Endocrinol (Oxf) 2016; 85:757-763
  115. Pedersen KM, Laurberg P, Iversen E, Knudsen PR, Gregersen HE, Rasmussen OS, Larsen KR, Eriksen GM, Johannesen PL. Amelioration of some pregnancy-associated variations in thyroid function by iodine supplementation. J Clin Endocrinol Metab 1993; 77:1078-1083
  116. Mandel SJ, Spencer CA, Hollowell JG. Are detection and treatment of thyroid insufficiency in pregnancy feasible? Thyroid 2005; 15:44-53
  117. Ekins R. Measurement of free hormones in blood. Endocr Rev 1990; 11:5-46
  118. Faix JD. Principles and pitfalls of free hormone measurements. Best Pract Res Clin Endocrinol Metab 2013; 27:631-645
  119. Midgley JE. Direct and indirect free thyroxine assay methods: theory and practice. Clin Chem 2001; 47:1353-1363
  120. Stockigt JR, Lim CF. Medications that distort in vitro tests of thyroid function, with particular reference to estimates of serum free thyroxine. Best Pract Res Clin Endocrinol Metab 2009; 23:753-767
  121. Sarne DH, Refetoff S, Nelson JC, Linarelli LG. A new inherited abnormality of thyroxine-binding globulin (TBG-San Diego) with decreased affinity for thyroxine and triiodothyronine. J Clin Endocrinol Metab 1989; 68:114-119
  122. Nelson JC, Tomei RT. Dependence of the thyroxin/thyroxin-binding globulin (TBG) ratio and the free thyroxin index on TBG concentrations. Clin Chem 1989; 35:541-544
  123. Nelson JC, Wilcox BR, Pandian MR. Dependence of free thyroxine estimates obtained with equilibrium tracer dialysis on the concentration of thyroxine-binding globulin. Clin Chem 1992; 38:1294-1300
  124. Nelson JC, Nayak SS, Wilcox RB. Variable underestimates by serum free thyroxine (T4) immunoassays of free T4 concentrations in simple solutions. J Clin Endocrinol Metab 1994; 79:1373-1375
  125. Nelson JC, Wilcox RB. Analytical performance of free and total thyroxine assays. Clin Chem 1996; 42:146-154
  126. Toldy E, Locsei Z, Szabolcs I, Bezzegh A, Kovács GL. Protein interference in thyroid assays: an in vitro study with in vivo consequences. Clin Chim Acta 2005; 353:93-104
  127. Fritz KS, Wilcox RB, Nelson JC. Quantifying spurious free T4 results attributable to thyroxine-binding proteins in serum dialysates and ultrafiltrates. Clin Chem 2007; 53:985-988
  128. Fillée C, Cumps J, Ketelslegers JM. Comparison of three free T4 (FT4) and free T3 (FT3) immunoassays in healthy subjects and patients with thyroid diseases and severe non-thyroidal illnesses. Clin Lab 2012; 58:725-736
  129. Uchimura H, Nagataki S, Tabuchi T, Mizuno M, Ingbar SH. Measurements of free thyroxine: comparison of per cent of free thyroxine in diluted and undiluted sera. J Clin Endocrinol Metab 1976; 42:561-566
  130. Iitaka M, Kawasaki S, Sakurai S, Hara Y, Kuriyama R, Yamanaka K, Kitahama s, Miura S, Kawakami Y, Katayama S. Serum substances that interfere with thyroid hormone assays in patients with chronic renal failure. Clin Endocrinol 1998; 48:739-746
  131. Holm SS, Andreasen L, Hansen SH, Faber J, Staun-Olsen P. Influence of adsorption and deproteination on potential free thyroxine reference methods. Clin Chem 2002; 48:108-114
  132. Nelson JC, Weiss RM. The effect of serum dilution on free thyroxine (T4) concentration in the low T4 syndrome of nonthyroidal illness. J Clin Endocrinol Metab 1985; 61:239-246
  133. Christofides ND, Midgley JE. Inaccuracies in free thyroid hormone measurement by ultrafiltration and tandem mass spectrometry. Clin Chem 2009; 55:2228-2229; author reply 2229-2230
  134. Van Uytfanghe K, Stockl D, Ross HA, Thienpont LM. Use of frozen sera for FT4 standardization: investigation by equilibrium dialysis combined with isotope dilution-mass spectrometry and immunoassay. Clin Chem 2006; 52:1817-1821
  135. Sterling K, Brenner MA. Free thyroxine in human serum : simplified measurement with the aid of magnesium precipitation. J Clin Invest 1966; 45:153-163
  136. Ellis SM, Ekins R. Direct measurement by radioimmunoassay of the free thyroid hormone concentrations in serum. Acta Endocrinol (Suppl) 1973; 177:106-
  137. Nelson JC, Tomei RT. Direct determination of free thyroxin in undiluted serum by equilibrium dialysis/radioimmunoassay. Clin Chem 1988; 34:1737-1744
  138. Wang YS, Hershman JM, Pekary AE. Improved ultrafiltration method for simultaneous measurement of free thyroxin and free triiodothyronine in serum. Clin Chem 1985; 31:517-522
  139. Weeke J, Boye N, Orskov H. Ultrafiltration method for direct radioimmunoassay measurement of free thyroxine and free tri-iodothyronine in serum. Scand J Clin Lab Invest 1986; 46:381-389
  140. Tikanoja SH, Liewendahl BK. New ultrafiltration method for free thyroxin compared with equilibrium dialysis in patients with thyroid dysfunction and nonthyroidal illness. Clin Chem 1990; 36:800-804
  141. Soldin SJ, Soukhova N, Janicic N, Jonklaas J, Soldin OP. The measurement of free thyroxine by isotope dilution tandem mass spectrometry. Clin Chim Acta 2005; 358:113-118
  142. Fritz KS, Wilcox RB, Nelson JC. A direct free thyroxine (T4) immunoassay with the characteristics of a total T4 immunoassay. Clin Chem 2007; 53:911-915
  143. Romelli PB, Pennisi F, Vancheri L. Measurement of free thyroid hormones in serum by column adsorption chromatography and radioimmunoassay. J Endocrinol Invest 1979; 2:25-40
  144. Ross HA, Benraad TJ. Is free thyroxine accurately measurable at room temperature? Clin Chem 1992; 38:880-886
  145. Wilcox RB, Nelson JC, Tomei RT. Heterogeneity in affinities of serum proteins for thyroxine among patients with non-thyroidal illness as indicated by the serum free thyroxine response to serum dilution. Eur J Endocrinol 1994; 131:9-13
  146. Wang R, Nelson JC, Weiss RM, Wilcox RB. Accuracy of free thyroxine measurements across natural ranges of thyroxine binding to serum proteins. Thyroid 2000; 10:31-39
  147. Csako G, Zweig MH, Benson C, Ruddel M. On the albumin-dependence of measurements of free thyroxin. II. Patients with non-thyroidal illness. Clin Chem 1987; 33:87-92
  148. Witherspoon LR, el Shami AS, Shuler SE, Neely H, Sonnemaker R, Gilbert SS, Alyea K. Chemically blocked analog assays for free thyronines. II. Use of equilibrium dialysis to optimize the displacement by chemical blockers of T4 analog and T3 analog from albumin while avoiding displacement of T4 and T3 from thyroxin-binding globulin. Clin Chem 1988; 34:17-23
  149. Witherspoon LR, el Shami AS, Shuler SE, Neely H, Sonnemaker R, Gilbert SS, Alyea K. Chemically blocked analog assays for free thyronines. I. The effect of chemical blockers on T4 analog and T4 binding by albumin and by thyroxin-binding globulin. Clin Chem 1988; 34:9-16
  150. Csako G, Zweig MH, Glickman J, Ruddel M, Kestner J. Direct and indirect techniques for free thyroxin compared in patients with nonthyroidal illness. II. Effect of prealbumin, albumin, and thyroxin-binding globulin. Clin Chem 1989; 35:1655-1662
  151. Weeke J, Orskov H. Ultrasensitive radioimmunoassay for direct determination of free triiodothyronine concentration in serum. Scand J Clin Lab Invest 1975; 35:237-244
  152. Ekins RP. Ligand assays: from electrophoresis to miniaturized microarrays. Clin Chem 1998; 44:2015-2030
  153. Levinson SS, Rieder SV. Parameters affecting a rapid method in which Sephadex is used to determine the percentage of free thyroxine in serum. Clin Chem 1974; 20:1568-1572
  154. Holm SS, Hansen SH, Faber J, Staun-Olsen P. Reference methods for the measurement of free thyroid hormones in blood: evaluation of potential reference methods for free thyroxine. Clin Biochem 2004; 37:85-93
  155. Hopley CJ, Stokes P, Webb KS, Baynham M. The analysis of thyroxine in human serum by an 'exact matching' isotope dilution method with liquid chromatography/tandem mass spectrometry. Rapid communications in mass spectrometry : RCM 2004; 18:1033-1038
  156. Jonklaas J, Kahric-Janicic N, Soldin OP, Soldin SJ. Correlations of free thyroid hormones measured by tandem mass spectrometry and immunoassay with thyroid-stimulating hormone across 4 patient populations. Clin Chem 2009; 55:1380-1388
  157. Soldin OP, Soldin SJ. Thyroid hormone testing by tandem mass spectrometry. Clin Biochem 2011; 44:89-94
  158. Soldin OP, Jang M, Guo T, Soldin SJ. Pediatric reference intervals for free thyroxine and free triiodothyronine. Thyroid 2009; 19:699-702
  159. Tractenberg RE, Jonklaas J, Soldin SJ. Agreement of immunoassay and tandem mass spectrometry in the analysis of cortisol and free t4: interpretation and implications for clinicians. Int J Anal Chem 2010; 2010
  160. Larsen PR, Alexander NM, Chopra IJ, Hay ID, Hershman JM, Kaplan MM, Mariash CN, Nicoloff JT, Oppenheimer JH, Solomon DH, Surks MI. Revised nomenclature for tests of thyroid hormones and thyroid-related proteins in serum. (Committee on Nomenclature of the American Thyroid Association). Clin Chem 1987; 33:2114-2119
  161. Litherland PGH, Bromage NR, Hall RA. Thyroxine binding globulin (TBG) and thyroxine binding prealbumin (TBPA) measurement, compared with the conventional T3 uptake in the diagnosis of thyroid disease. Clin Chim Acta 1982; 122:345-352
  162. Burr WA, Ramsden DB, Evans SE, Hogan T, Hoffenberg R. Concentration of thyroxine-binding globulin: value of direct assay. Br Med J 1977; 1:485-488
  163. Surks MI, DeFesi CR. Normal serum free thyroid hormone concentrations in patients treated with phenytoin or carbamazepine. A paradox resolved. Jama 1996; 275:1495-1498
  164. Stevenson HP, Archbold GP, Johnston P, Young IS, Sheridan B. Misleading serum free thyroxine results during low molecular weight heparin treatment. Clin Chem 1998; 44:1002-1007
  165. Berberoğlu M. Drugs and thyroid interaction. Pediatr Endocrinol Rev 2003; 1:251-256
  166. Witherspoon LR, Shuler SE, Garcia MM. The triiodothyronine uptake test: an assessment of methods. Clin Chem 1981; 27:1272-1276
  167. Witherspoon LR, Shuler SE, Gilbert S. Estimation of thyroxin, triiodothyronine, thyrotropin, free thyroxin, and triiodothyronine uptake by use of magnetic-particle solid phases. Clin Chem 1985; 31:413-419
  168. Harpen MD, Lee WNP, Siegel JA, Greenfield MA. Serum binding of triiodothyronine: theoretical and practical implications for in vitro triiodothyronine uptake. Endocrinol 1982; 110:1732-1739
  169. Wilke TJ. Free thyroid hormone index, thyroid hormone/thyroxin-binding globulin ratio, triiodothyronine uptake, and thyroxin-binding globulin compared for diagnostic value regarding thyroid function. Clin Chem 1983; 29:74-79
  170. Faix JD, Rosen HN, Velazquez FR. Indirect estimation of thyroid hormone-binding proteins to calculate free thyroxine index: comparison of nonisotopic methods that use labeled thyroxine ("T-uptake"). Clin Chem 1995; 41:41-47
  171. Roberts RF, La'ulu SL, Roberts WL. Performance characteristics of seven automated thyroxine and T-uptake methods. Clin Chim Acta 2007; 377:248-255
  172. Burr WA, Ramsden DB, Hoffenberg R. Hereditary abnormalities of thyroxine-binding globulin concentration. A study of 19 kindreds with inherited increase or decrease of thyroxine-binding globulin. The Quarterly journal of medicine 1980; 49:295-313
  173. Jensen IW, Faber J. Familial dysalbuminemic hyperthyroxinemia: a review. J Royal Soc Med 1988; 81:34-37
  174. Tokmakjian S, Haines M, Edmonds M. Free thyroxine measured by the Ciba Corning ACS-180 on samples from patients with familial dysalbuminemic hyperthyroxinemia. Clin Chem 1993; 39:1748-1749
  175. Pannain S, Feldman M, Eiholzer U, Weiss RE, Scherberg NH, Refetoff S. Familial dysalbuminemic hyperthyroxinemia in a Swiss family caused by a mutant albumin (R218P) shows an apparent discrepancy between serum concentration and affinity for thyroxine. J Clin Endocrinol Metab 2000; 85:2786-2792
  176. Hoshikawa S, Mori K, Kaise N, Nakagawa Y, Ito S, Yoshida K. Artifactually elevated serum-free thyroxine levels measured by equilibrium dialysis in a pregnant woman with familial dysalbuminemic hyperthyroxinemia. Thyroid 2004; 14:155-160
  177. Pietras SM, Safer JD. Diagnostic confusion attributable to spurious elevation of both total thyroid hormone and thyroid hormone uptake measurements in the setting of autoantibodies: case report and review of related literature. Endocr Pract 2008; 14:738-742
  178. van der Watt G, Haarburger D, Berman P. Euthyroid patient with elevated serum free thyroxine. Clin Chem 2008; 54:1239-1241
  179. Surks MI, Hupart KH, Pan C, Shapiro LE. Normal free thyroxine in critical nonthyroidal illnesses measured by ultrafiltration of undiluted serum and equilibrium dialysis. J Clin Endocrinol Metab 1988; 67:1031-1038
  180. Sapin R, Schlienger JL, Gasser F, Noel E, Lioure B, Grunenberger F, Goichot B, Grucker D. Intermethod discordant free thyroxine measurements in bone marrow-transplanted patients. Clin Chem 2000; 46:418-422
  181. Mendel CM, Frost PH, Kunitake ST, Cavalieri RR. Mechanism of the heparin-induced increase in the concentration of free thyroxine in plasma. J Clin Endocrinol Metab 1987; 65:1259-
  182. Hawkins RC. Furosemide interference in newer free thyroxine assays. Clin Chem 1998; 44:2550-2551
  183. Schussler GC, Plager JE. Effect of preliminary purification of 131-Thyroxine on the determination of free thyroxine in serum. J Clin Endocrinol 1967; 27:242-250
  184. Sophianopoulos J, Jerkunica I, Lee CN, Sgoutas D. An improved ultrafiltration method for free thyroxine and triiodothyronine in serum. Clin Chem 1980; 26:159-162
  185. Sterling K, Hededus A. Free thyroxine in human serum. J Clin Invest 1962; 41:1031-1040
  186. Oppenheimer JH, Squef R, Surks MI, Hauer H. Binding of thyroxine by serum proteins evaluated by equilibrium dialysis and electrophoresis techniques. J Clin Invest 1963; 42:1769-?
  187. Snyder SM, Cavalieri RR, Ingbar SH. Simultaneous measurement of percentage free thyroxine and triiodothyronine : comparison of equilibrium dialysis and sephadex chromatography. J Nucl Med 1976; 17:660-664
  188. Ross HA. A dialysis rate method for the measurement of free triiodothyronine and steroid hormones in blood. Experimentia 1978; 34:538-539
  189. Ross HA, Visser JW, der Kinderen PJ, Tertoolen JF, Thijssen JH. A comparative study of free thyroxine estimations. Ann Clin Biochem 1982; 19:108-113
  190. Swinkels LM, Ross HA, Benraad TJ. A symmetric dialysis method for the determination of free testosterone in human plasma. Clin Chim Acta 1987; 165:341-349
  191. Nelson JC, Weiss RM. The effects of serum dilution on free thyroxine (T4) concentration in the low T4 syndrome of nonthyroidal illness. J Clin Endocrinol Metab 1985; 61:239-246
  192. Witherspoon LR, Shuler SE, Garcia MM, Zollinger LA. Effects of contaminant radioactivity on results of 125I-radioligand assay. Clin Chem 1979; 25:1975-1977
  193. Bourcigaux N, Lepoutre-Lussey C, Guéchot J, Donadille B, Faugeron I, Ouzounian S, Christin-Maître S, Bouchard P, Duron F. Thyroid function at the third trimester of pregnancy in a Northern French population. Ann Endocrinol (Paris) 2010; 71:519-524
  194. Midgley JE, Hoermann R. Measurement of total rather than free thyroxine in pregnancy: the diagnostic implications. Thyroid 2013; 23:259-261
  195. Pantalone KM, Hatipoglu B, Gupta MK, Kennedy L, Hamrahian AH. Measurement of Serum Free Thyroxine Index May Provide Additional Case Detection Compared to Free Thyroxine in the Diagnosis of Central Hypothyroidism. Case reports in endocrinology 2015; 2015:965191
  196. Ekins R. Free hormone assays. Nucl Med Commun 1993; 14:676-688
  197. Bayer MF. Free thyroxine results are affected by albumin concentration and nonthyroidal illness. Clin Chim Acta 1983; 130:391-396
  198. Liewendahl K, Tikanoja S, Helenius T, Valimaki M. Discrepancies between serum free triiodothyronine and free thyroxin as measured by equilibrium dialysis and analog radioimmunoassay in nonthyroidal illnesses. Clin Chem 1984; 30:760-762
  199. Midgley JEM, Sheehan CP, Christofides ND, Fry JE, Browning D, Mandel R. Concentrations of free thyroxin and albumin in serum in nonthyroidal illness : assay artefacts and physiological influences. Clin Chem 1990; 36:765-771
  200. Wilcox RB, Nelson JC. Counterpoint: legitimate and illegitimate tests of free-analyte assay function: we need to identify the factors that influence free-analyte assay results. Clin Chem 2009; 55:442-444
  201. Ekins R. Validity of analog free thyroxin immunoassays. Clin Chem 1987; 33:2137-2152
  202. Ekins R. Analytical measurements of free thyroxine. Clinics Lab Med 1993; 13:599-630
  203. Bayer MF. Free thyroxine results are affected by albumin concentration and nonthyroidal illness. Clin Chim Acta 1983; 130:391-396
  204. Ekins R, Edwards P, Jackson T, Geiscler D. Interpretation of labeled-analog free hormone assay. Clin Chem 1984; 30:491-493
  205. Meinhold H, Wenzel KW. Comparative methodological studies with six commercial FT4 kits. NucCompact 1985; 16:317-320
  206. Midgley JEM, Moon CR, Wilkins TA. Validity of analog free thyroxin immunoassays. Part II. Clin Chem 1987; 33:2145-2152
  207. Zacharopoulou AD, Christofidis I, Kakabakos SE, Koupparis MA. Free thyroxine solid-analog immunoassays. investigation of the albumin effect on the antibody binding to immobilized thyroxine-protein conjugates. J Immunoassay Immunochem 2002; 23:95-105
  208. Midgley JE, Christofides ND. Point: legitimate and illegitimate tests of free-analyte assay function. Clin Chem 2009; 55:439-441
  209. Piketty ML, d'Herbomez M, Le Guillouzic D, Lebtahi R, Cosson E, Dumont A, Dilouya A, Helal BO. Clinical comparison of three labeled-antibody immunoassays of free triiodothyronine. Clin Chem 1996; 42:933-941
  210. Sheehan CP, Christofides ND. One-step, labeled-antibody assay for measuring free thyroxin. II. Performance in a multicenter trial. Clin Chem 1992; 38:19-25
  211. Sapin R, d'Herbomez M. Free thyroxine measured by equilibrium dialysis and nine immunoassays in sera with various serum thyroxine-binding capacities. Clin Chem Lab Med 2003; 49:1531-1535
  212. Masika LS, Zhao Z, Soldin SJ. Is measurement of TT3 by immunoassay reliable at low concentrations? A comparison of the Roche Cobas 6000 vs. LC-MSMS. Clin Biochem 2016; 49:846-849
  213. Livingston M, Birch K, Guy M, Kane J, Heald AH. No role for tri-iodothyronine (T3) testing in the assessment of levothyroxine (T4) over-replacement in hypothyroid patients. British journal of biomedical science 2015; 72:160-163
  214. Persani L, Asteria C, Beck-Peccoz P. Dissociation between immunological and biological activities of circulating TSH. Exp Clin Endocrinol 1994; 102:38-48
  215. Persani L, Ferretti E, Borgato S, Faglia G, Beck-Peccoz P. Circulating thyrotropin bioactivity in sporadic central hypothyroidism. J Clin Endocrinol Metab 2000; 85:3631-3635
  216. Baskin HJ, Cobin RH, Duick DS, Gharib H, Guttler RB, Kaplan MM, Segal RL. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the evaluation and treatment of hyperthyroidism and hypothyroidism. Endocr Pract 2002; 8:457-469
  217. Lania A, Persani L, Beck-Peccoz P. Central hypothyroidism. Pituitary 2008; 11:181-186
  218. Haugen BR. Drugs that suppress TSH or cause central hypothyroidism. Best Pract Res Clin Endocrinol Metab 2009; 23:793-800
  219. Roelfsema F, Kok S, Kok P, Pereira AM, Biermasz NR, Smit JW, Frolich M, Keenan DM, Veldhuis JD, Romijn JA. Pituitary-hormone secretion by thyrotropinomas. Pituitary 2009; 12:200-210
  220. Spencer CA, LoPresti JS, Patel A, Guttler RB, Eigen A, Shen D, Nicoloff JT. Applications of a new chemiluninometric thyrotropin assay to subnormal measurement. J Clin Endocrinol Metab 1990; 70:453-460
  221. Meier CA, Maisey MN, Lowry A, Müller J, Smith MA. Interindividual differences in the pituitary-thyroid axis influence the interpretation of thyroid function tests. CLIN Endocrinol (Oxf) 1993; 39:101-107
  222. Benhadi N, Fliers E, Visser TJ, Reitsma JB, Wiersinga WM. Pilot study on the assessment of the setpoint of the hypothalamus-pituitary-thyroid axis in healthy volunteers. Eur J Endocrinol 2010; 162:323-329
  223. Hoermann R, Eckl W, Hoermann C, Larisch R. Complex relationship between free thyroxine and TSH in the regulation of thyroid function. Eur J Endocrinol 2010; 162:1123-1129
  224. Clark PM, Holder RL, Haque SM, Hobbs FD, Roberts LM, Franklyn JA. The relationship between serum TSH and free T4 in older people. J Clin Pathol 2012; 65:463-465
  225. De Grande LA, Van Uytfanghe K, Thienpont LM. A Fresh Look at the Relationship between TSH and Free Thyroxine in Cross-Sectional Data. Eur Thyroid J 2015; 4:69-70
  226. Brown SJ, Bremner AP, Hadlow NC, Feddema P, Leedman PJ, O'Leary PC, Walsh JP. The log TSH-free T4 relationship in a community-based cohort is nonlinear and is influenced by age, smoking and thyroid peroxidase antibody status. Clin Endocrinol (Oxf) 2016;
  227. Hadlow NC, Rothacker KM, Wardrop R, Brown SJ, Lim EM, Walsh JP. The relationship between TSH and free T(4) in a large population is complex and nonlinear and differs by age and sex. J Clin Endocrinol Metab 2013; 98:2936-2943
  228. Chaker L, Korevaar TI, Medici M, Uitterlinden AG, Hofman A, Dehghan A, Franco OH, Peeters RP. Thyroid Function Characteristics and Determinants: The Rotterdam Study. Thyroid 2016; 26:1195-1204
  229. Jones AM, Honour JW. Unusual results from immunoassays and the role of the clinical endocrinologist. Clin Endocrinol (Oxf) 2006; 64:234-244
  230. Teti C, Nazzari E, Galletti MR, Mandolfino MG, Pupo F, Pesce G, Lillo F, Bagnasco M, Benvenga S. Unexpected Elevated Free Thyroid Hormones in Pregnancy. Thyroid 2016; 26:1640-1644
  231. Soheilipour F, Fazilaty H, Jesmi F, Gahl WA, Behnam B. First report of inherited thyroxine-binding globulin deficiency in Iran caused by a known de novo mutation in SERPINA7. Molecular genetics and metabolism reports 2016; 8:13-16
  232. Jin HY. Thyroxine binding globulin excess detected by neonatal screening. Annals of pediatric endocrinology & metabolism 2016; 21:105-108
  233. Greenberg SM, Ferrara AM, Nicholas ES, Dumitrescu AM, Cody V, Weiss RE, Refetoff S. A novel mutation in the Albumin gene (R218S) causing familial dysalbuminemic hyperthyroxinemia in a family of Bangladeshi extraction. Thyroid 2014; 24:945-950
  234. Osaki Y, Hayashi Y, Nakagawa Y, Yoshida K, Ozaki H, Fukazawa H. Familial Dysalbuminemic Hyperthyroxinemia in a Japanese Man Caused by a Point Albumin Gene Mutation (R218P). Japanese clinical medicine 2016; 7:9-13
  235. Kragh-Hansen U, Minchiotti L, Coletta A, Bienk K, Galliano M, Schiott B, Iwao Y, Ishima Y, Otagiri M. Mutants and molecular dockings reveal that the primary L-thyroxine binding site in human serum albumin is not the one which can cause familial dysalbuminemic hyperthyroxinemia. Biochim Biophys Acta 2016; 1860:648-660
  236. Cho YY, Song JS, Park HD, Kim YN, Kim HI, Kim TH, Chung JH, Ki CS, Kim SW. First Report of Familial Dysalbuminemic Hyperthyroxinemia With an ALB Variant. Annals of laboratory medicine 2017; 37:63-65
  237. Sapin R, Gasser F, Schlienger JL. Familial dysalbuminemic hyperthyroxinemia and thyroid hormone autoantibodies: interference in current free thyroid hormone assays. Horm Res 1996; 45:139-141
  238. Stockigt JR. Guidelines for diagnosis and monitoring of thyroid disease: nonthyroidal illness. Clin Chem 1996; 42:188-192
  239. Van den Berghe G. Non-thyroidal illness in the ICU: a syndrome with different faces. Thyroid 2014; 24:1456-1465
  240. Kaptein EM, Macintyre SS, Weiner JM, Spencer CA. Free thyroxine estimates in nonthyroidal illness: comparison of eight methods. J Clin Endocrinol Metab 1981; 52:1073-1077
  241. Drinka PJ, Nolten WE, Voeks S, Langer E, Carlson IH. Misleading elevation of the free thyroxine index in nursing home residents. Arch Pathol Lab Med 1991; 115:1208-1211
  242. Becker DV, Bigos ST, Gaitan E, Morris JC, Rallison ML, Spencer CA, Sugawara M, Middlesworth LV, Wartofsky L. Optimal use of blood tests for assessment of thyroid function. JAMA 1993; 269:2736
  243. Hamblin PS, Dyer SA, Mohr VS, Le Grand BA, Lim CF, Tuxen DV, Topliss DJ, Stockigt JR. Relationship between thyrotropin and thyroxine changes during recovery from severe hypothyroxinemia of critical illness. J Clin Endocrinol Metab 1986; 62:717-722
  244. Spencer CA, Eigen A, Shen D, Duda M, Qualls S, Weiss S, Nicoloff JT. Specificity of sensitive assays of thyrotropin (TSH) used to screen for thyroid disease in hospitalized patients. Clin Chem 1987; 33:1391-1396
  245. de Vries EM, Fliers E, Boelen A. The molecular basis of the non-thyroidal illness syndrome. J Endocrinol 2015; 225:R67-81
  246. Moura Neto A, Zantut-Wittmann DE. Abnormalities of Thyroid Hormone Metabolism during Systemic Illness: The Low T3 Syndrome in Different Clinical Settings. International journal of endocrinology 2016; 2016:2157583
  247. Lem AJ, de Rijke YB, van Toor H, de Ridder MA, Visser TJ, Hokken-Koelega AC. Serum thyroid hormone levels in healthy children from birth to adulthood and in short children born small for gestational age. J Clin Endocrinol Metab 2012; 97:3170-3178
  248. Chaler EA, Fiorenzano R, Chilelli C, Llinares V, Areny G, Herzovich V, Maceiras M, Lazzati JM, Mendioroz M, Rivarola MA, Belgorosky A. Age-specific thyroid hormone and thyrotropin reference intervals for a pediatric and adolescent population. Clin Chem Lab Med 2012; 50:885-890
  249. La'ulu SL, Rasmussen KJ, Straseski JA. Pediatric Reference Intervals for Free Thyroxine and Free Triiodothyronine by Equilibrium Dialysis-Liquid Chromatography-Tandem Mass Spectrometry. Journal of clinical research in pediatric endocrinology 2016; 8:26-31
  250. Soldin SJ, Cheng LL, Lam LY, Werner A, Le AD, Soldin OP. Comparison of FT4 with log TSH on the Abbott Architect ci8200: Pediatric reference intervals for free thyroxine and thyroid-stimulating hormone. Clin Chim Acta 2010; 411:250-252
  251. Verburg FA, Kirchgässner C, Hebestreit H, Steigerwald U, Lentjes EG, Ergezinger K, Grelle I, Reiners C, Luster M. Reference ranges for analytes of thyroid function in children. Horm Metab Res 2011; 43:422-426
  252. Loh TP, Sethi SK, Metz MP. Paediatric reference interval and biological variation trends of thyrotropin (TSH) and free thyroxine (T4) in an Asian population. J Clin Pathol 2015; 68:642-647
  253. Lazarus J, Brown RS, Daumerie C, Hubalewska-Dydejczyk A, Negro R, Vaidya B. 2014 European thyroid association guidelines for the management of subclinical hypothyroidism in pregnancy and in children. Eur Thyroid J 2014; 3:76-94
  254. Chan S, Boelaert K. Optimal management of hypothyroidism, hypothyroxinaemia and euthyroid TPO antibody positivity preconception and in pregnancy. Clin Endocrinol (Oxf) 2015; 82:313-326
  255. Furnica RM, Lazarus JH, Gruson D, Daumerie C. Update on a new controversy in endocrinology: isolated maternal hypothyroxinemia. J Endocrinol Invest 2015; 38:117-123
  256. Noten AM, Loomans EM, Vrijkotte TG, van de Ven PM, van Trotsenburg AS, Rotteveel J, van Eijsden M, Finken MJ. Maternal hypothyroxinaemia in early pregnancy and school performance in 5-year-old offspring. Eur J Endocrinol 2015; 173:563-571
  257. Min H, Dong J, Wang Y, Wang Y, Teng W, Xi Q, Chen J. Maternal Hypothyroxinemia-Induced Neurodevelopmental Impairments in the Progeny. Molecular neurobiology 2016; 53:1613-1624
  258. Tong Z, Xiaowen Z, Baomin C, Aihua L, Yingying Z, Weiping T, Zhongyan S. The Effect of Subclinical Maternal Thyroid Dysfunction and Autoimmunity on Intrauterine Growth Restriction: A Systematic Review and Meta-Analysis. Medicine (Baltimore) 2016; 95:e3677
  259. Dosiou C, Medici M. MANAGEMENT OF ENDOCRINE DISEASE: Isolated maternal hypothyroxinemia during pregnancy: Knowns and unknowns. Eur J Endocrinol 2016;
  260. Agarwal MM, Dhatt GS, Punnose J, Bishawi B, Zayed R. Thyroid function abnormalities and antithyroid antibody prevalence in pregnant women at high risk for gestational diabetes mellitus. Gynecol Endocrinol 2006; 22:261-266
  261. Cleary-Goldman J, Malone FD, Lambert-Messerlian G, Sullivan L, Canick J, Porter TF, Luthy D, Gross S, Bianchi DW, D'Alton ME. Maternal thyroid hypofunction and pregnancy outcome. Obstet Gynecol 2008; 112:85-92
  262. Oguz A, Tuzun D, Sahin M, Usluogullari AC, Usluogullari B, Celik A, Gul K. Frequency of isolated maternal hypothyroxinemia in women with gestational diabetes mellitus in a moderately iodine-deficient area. Gynecol Endocrinol 2015; 31:792-795
  263. Haddow JE, Craig WY, Neveux LM, Palomaki GE, Lambert-Messerlian G, Malone FD, D'Alton ME. Free Thyroxine During Early Pregnancy and Risk for Gestational Diabetes. PLoS One 2016; 11:e0149065
  264. Yang S, Shi FT, Leung PC, Huang HF, Fan J. Low Thyroid Hormone in Early Pregnancy Is Associated with an Increased Risk of Gestational Diabetes Mellitus. J Clin Endocrinol Metab 2016:jc20161506
  265. Panesar NS, Li CY, Rogers MS. Reference intervals for thyroid hormones in pregnant Chinese women. Ann Clin Biochem 2001; 38:329-332
  266. Sapin R, D'Herbomez M, Schlienger JL. Free thyroxine measured with equilibrium dialysis and nine immunoassays decreases in late pregnancy. Clin Lab 2004; 50:581-584
  267. Berta E, Samson L, Lenkey A, Erdei A, Cseke B, Jenei K, Major T, Jakab A, Jenei Z, Paragh G, Nagy EV, Bodor M. Evaluation of the thyroid function of healthy pregnant women by five different hormone assays. Pharmazie 2010; 65:436-439
  268. Anckaert E, Poppe K, Van Uytfanghe K, Schiettecatte J, Foulon W, Thienpont LM. FT4 immunoassays may display a pattern during pregnancy similar to the equilibrium dialysis ID-LC/tandem MS candidate reference measurement procedure in spite of susceptibility towards binding protein alterations. Clin Chim Acta 2010; 411:1348-1353
  269. Männistö T, Surcel HM, Ruokonen A, Vääräsmäki M, Pouta A, Bloigu A, Järvelin MR, Hartikainen AL, Suvanto E. Early pregnancy reference intervals of thyroid hormone concentrations in a thyroid antibody-negative pregnant population. Thyroid 2011; 21:291-298
  270. Medici M, Korevaar TI, Visser WE, Visser TJ, Peeters RP. Thyroid function in pregnancy: what is normal? Clin Chem 2015; 61:704-713
  271. Laurberg P, Andersen SL, Hindersson P, Nohr EA, Olsen J. Dynamics and Predictors of Serum TSH and fT4 Reference Limits in Early Pregnancy: A Study Within the Danish National Birth Cohort. J Clin Endocrinol Metab 2016; 101:2484-2492
  272. Price A, Obel O, Cresswell J, Catch I, Rutter S, Barik S, Heller SR, Weetman AP. Comparison of thyroid function in pregnant and non-pregnant Asian and western Caucasian women. Clin Chim Acta 2001; 308:91-98
  273. Dhatt GS, Jayasundaram R, Wareth LA, Nagelkerke N, Jayasundaram K, Darwish EA, Lewis A. Thyrotrophin and free thyroxine trimester-specific reference intervals in a mixed ethnic pregnant population in the United Arab Emirates. Clin Chim Acta 2006; 370:147-151
  274. La'ulu SL, Roberts WL. Ethnic differences in first-trimester thyroid reference intervals. Clin Chem 2011; 57:913-915
  275. Korevaar TI, Medici M, de Rijke YB, Visser W, de Muinck Keizer-Schrama SM, Jaddoe VW, Hofman A, Ross HA, Visser WE, Hooijkaas H, Steegers EA, Tiemeier H, Bongers-Schokking JJ, Visser TJ, Peeters RP. Ethnic differences in maternal thyroid parameters during pregnancy: the Generation R study. J Clin Endocrinol Metab 2013; 98:3678-3686
  276. Antonangeli L, Maccherini D, Cavaliere R, Di Giulio C, Reinhardt B, Pinchera A, Aghini-Lombardi F. Comparison of two different doses of iodide in the prevention of gestational goiter in marginal iodine deficiency: a longitudinal study. Eur J Endocrinol 2002; 147:29-34
  277. Moleti M, Di Bella B, Giorgianni G, Mancuso A, De Vivo A, Alibrandi A, Trimarchi F, Vermiglio F. Maternal thyroid function in different conditions of iodine nutrition in pregnant women exposed to mild-moderate iodine deficiency: an observational study. Clin Endocrinol 2011; 74:762-768
  278. Shi X, Han C, Li C, Mao J, Wang W, Xie X, Li C, Xu B, Meng T, Du J, Zhang S, Gao Z, Zhang X, Fan C, Shan Z, Teng W. Optimal and safe upper limits of iodine intake for early pregnancy in iodine-sufficient regions: a cross-sectional study of 7190 pregnant women in China. J Clin Endocrinol Metab 2015; 100:1630-1638
  279. Mannisto T, Hartikainen AL, Vaarasmaki M, Bloigu A, Surcel HM, Pouta A, Jarvelin MR, Ruokonen A, Suvanto E. Smoking and early pregnancy thyroid hormone and anti-thyroid antibody levels in euthyroid mothers of the Northern Finland Birth Cohort 1986. Thyroid 2012; 22:944-950
  280. Ashoor G, Kametas NA, Akolekar R, Guisado J, Nicolaides KH. Maternal thyroid function at 11-13 weeks of gestation. Fetal Diagn Ther 2010; 27:156-163
  281. Pop VJ, Biondi B, Wijnen HA, Kuppens SM, Lvader H. Maternal thyroid parameters, body mass index and subsequent weight gain during pregnancy in healthy euthyroid women. Clin Endocrinol (Oxf) 2013; 79:577-583
  282. Bestwick JP, John R, Maina A, Guaraldo V, Joomun M, Wald NJ, Lazarus JH. Thyroid stimulating hormone and free thyroxine in pregnancy: expressing concentrations as multiples of the median (MoMs). Clin Chim Acta 2014; 430:33-37
  283. Han C, Li C, Mao J, Wang W, Xie X, Zhou W, Li C, Xu B, Bi L, Meng T, Du J, Zhang S, Gao Z, Zhang X, Yang L, Fan C, Teng W, Shan Z. High Body Mass Index Is an Indicator of Maternal Hypothyroidism, Hypothyroxinemia, and Thyroid-Peroxidase Antibody Positivity during Early Pregnancy. BioMed research international 2015; 2015:351831
  284. Rotmensch S, Cole LA. False diagnosis and needless therapy of presumed malignant disease in women with false-positive human chorionic gonadotropin concentrations. Lancet 2000; 355:712-715
  285. Ballieux BE, Weijl NI, Gelderblom H, van Pelt J, Osanto S. False-positive serum human chorionic gonadotropin (HCG) in a male patient with a malignant germ cell tumor of the testis: a case report and review of the literature. The oncologist 2008; 13:1149-1154
  286. Henry N, Sebe P, Cussenot O. Inappropriate treatment of prostate cancer caused by heterophilic antibody interference. Nature clinical practice Urology 2009; 6:164-167
  287. Georges A, Charrie A, Raynaud S, Lombard C, Corcuff JB. Thyroxin overdose due to rheumatoid factor interferences in thyroid-stimulating hormone assays. Clin Chem Lab Med 2011; 49:873-875
  288. Bjerner J, Bolstad N, Piehler A. Belief is only half the truth--or why screening for heterophilic antibody interference in certain assays makes double sense. Ann Clin Biochem 2012; 49:381-386
  289. Pishdad GR, Pishdad P, Pishdad R. The effect of glucocorticoid therapy on a falsely raised thyrotropin due to heterophilic antibodies. Thyroid 2013; 23:1657-1658
  290. Marks V. False-positive immunoassay results: a multicenter survey of erroneous immunoassay results from assays of 74 analytes in 10 donors from 66 laboratories in seven countries. Clin Chem 2002; 48:2008-2016
  291. Ellis MJ, Livesey JH. Techniques for identifying heterophile antibody interference are assay specific: study of seven analytes on two automated immunoassay analyzers. Clin Chem 2005; 51:639-641
  292. Lewandowski KC, Dabrowska K, Lewinski A. Case report: When measured free T4 and free T3 may be misleading. Interference with free thyroid hormones measurements on Roche(R) and Siemens(R) platforms. Thyroid research 2012; 5:11
  293. Bolstad N, Warren DJ, Nustad K. Heterophilic antibody interference in immunometric assays. Best Pract Res Clin Endocrinol Metab 2013; 27:647-661
  294. Emerson JF, Ngo G, Emerson SS. Screening for interference in immunoassays. Clin Chem 2003; 49:1163-1169
  295. Massart C, Corcuff JB, Bordenave L. False-positive results corrected by the use of heterophilic antibody-blocking reagent in thyroglobulin immunoassays. Clin Chim Acta 2008; 388:211-213
  296. Spencer CA, Fatemi S, Singer P, Nicoloff JT, LoPresti JS. Serum Basal Thyroglobulin Measured by A 2nd Generation Assay Correlates with the Recombinant Human TSH-Stimulated Thyroglobulin Response in Patients Treated for Differentiated Thyroid Cancer Thyroid 2010; 20:587-595
  297. Koshida S, Asanuma K, Kuribayashi K, Goto M, Tsuji N, Kobayashi D, Tanaka M, Watanabe N. Prevalence of human anti-mouse antibodies (HAMAs) in routine examinations. Clin Chim Acta 2010; 411:391-394
  298. Ismail AA. On detecting interference from endogenous antibodies in immunoassays by doubling dilutions test. Clin Chem Lab Med 2007; 45:851-854
  299. Ross HA, Menheere PP, Thomas CM, Mudde AH, Kouwenberg M, Wolffenbuttel BH. Interference from heterophilic antibodies in seven current TSH assays. Ann Clin Biochem 2008; 45:616-618
  300. Gulbahar O, Konca Degertekin C, Akturk M, Yalcin MM, Kalan I, Atikeler GF, Altinova AE, Yetkin I, Arslan M, Toruner F. A Case With Immunoassay Interferences in the Measurement of Multiple Hormones. J Clin Endocrinol Metab 2015; 100:2147-2153
  301. Klee GG. Interferences in hormone immunoassays. Clin Lab Med 2004; 24:1-18
  302. Sturgeon CM, Viljoen A. Analytical error and interference in immunoassay: minimizing risk. Ann Clin Biochem 2011; 48:418-432
  303. King RI, Florkowski CM. How paraproteins can affect laboratory assays: spurious results and biological effects. Pathology 2010; 42:397-401
  304. Imperiali M, Jelmini P, Ferraro B, Keller F, della Bruna R, Balerna M, Giovanella L. Interference in thyroid-stimulating hormone determination. Eur J Clin Invest 2010; 40:756-758
  305. LeGatt DF, Higgins TN. Paraprotein interference in immunoassays. Ther Drug Monit 2015; 37:417
  306. Covinsky M, Laterza O, Pfeifer JD, Farkas-Szallasi T, Scott MG. Lamda antibody to Esherichia coli produces false-positive results in multiple immunometric assays. Clin Chem 2000; 46:1157-1161
  307. Luzzi VI, Scott MG, Gronowski AM. Negative thyrotropin assay interference associated with an IgGkappa paraprotein. Clin Chem 2003; 49:709-710
  308. Glinoer D, de Nayer P, Bourdoux P, Lemone M, Robyn C, van Steirteghem A, Kinthaert J, Lejeune B. Regulation of maternal thyroid during pregnancy. J Clin Endocrinol Metab 1990; 71:276-287
  309. Glinoer D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocrinol Rev 1997; 18:404-433
  310. Ramsden DB, Sheppard MC, Sawers RS, Smith SC, Hoffenberg R. Serum free thyroxine concentrations in normal euthyroid subjects and ones with high serum thyroxine binding globulin concentration. Clin Chim Acta 1983; 130:211-217
  311. Guven S, Alver A, Mentese A, Ilhan FC, Calapoglu M, Unsal MA. The novel ischemia marker 'ischemia-modified albumin' is increased in normal pregnancies. Acta Obstet Gynecol Scand 2009; 88:479-482
  312. Cameron SJ, Hagedorn JC, Sokoll LJ, Caturegli P, Ladenson PW. Dysprealbuminemic hyperthyroxinemia in a patient with hyperthyroid graves disease. Clin Chem 2005; 51:1065-1069
  313. DeCosimo DR, Fang SL, Braverman LE. Prevalence of familial dysalbuminemic hyperthyroxinemia in Hispanics. Ann Intern Med 1987; 107:780-781
  314. Kricka LJ. Human anti-animal antibody interference in immunological assays. Clin Chem 1999; 45:942-956
  315. Levinson SS, Miller JJ. Towards a better understanding of heterophile (and the like) antibody interference with modern immunoassays. Clin Chim Acta 2002; 325:1-15
  316. Despres N, Grant AM. Antibody interference in thyroid assays: a potential for clinical misinformation. Clin Chem 1998; 44:440-454
  317. Mongolu S, Armston AE, Mozley E, Nasruddin A. Heterophilic antibody interference affecting multiple hormone assays: Is it due to rheumatoid factor? Scand J Clin Lab Invest 2016; 76:240-242
  318. Astarita G, Gutierrez S, Kogovsek N, Mormandi E, Otero P, Calabrese C, Alcaraz G, Vazquez A, Abalovich M. False positive in the measurement of thyroglobulin induced by rheumatoid factor. Clin Chim Acta 2015; 447:43-46
  319. Weber TH, Käpyaho KI, Tanner P. Endogenous interference in immunoassays in clinical chemistry. A review. Scand J Clin Lab Invest 1990; 201:77-82
  320. Bjerner J, Nustad K, Norum LF, Olsen KH, Børmer OP. Immunometric assay interference: incidence and prevention. Clin Chem 2002; 48:613-621
  321. Ghosh S, Howlett M, Boag D, Malik I, Collier A. Interference in free thyroxine immunoassay. Eur J Intern Med 2008; 19:221-222
  322. Preissner CM, Dodge LA, O'Kane DJ, Singh RJ, Grebe SK. Prevalence of heterophilic antibody interference in eight automated tumor marker immunoassays. Clin Chem 2005; 51:208-210
  323. Preissner CM, O'Kane DJ, Singh RJ, Morris JC, Grebe SK. Phantoms in the assay tube: heterophile antibody interferences in serum thyroglobulin assays. J Clin Endocrinol Metab 2003; 88:3069-3074
  324. Verburg FA, Wäschle K, Reiners C, Giovanella L, Lentjes EG. Heterophile Antibodies Rarely Influence the Measurement of Thyroglobulin and Thyroglobulin Antibodies in Differentiated Thyroid Cancer Patients. Horm Metab Res 2010; 42:736-739
  325. Bjerner J, Olsen KH, Bormer OP, Nustad K. Human heterophilic antibodies display specificity for murine IgG subclasses. Clin Biochem 2005; 38:465-472
  326. Choi WW, Srivatsa S, Ritchie JC. Aberrant thyroid testing results in a clinically euthyroid patient who had received a tumor vaccine. Clin Chem 2005; 51:673-675
  327. Monchamp T, Chopra IJ, Wah DT, Butch AW. Falsely elevated thyroid hormone levels due to anti-sheep antibody interference in an automated electrochemiluminescent immunoassay. Thyroid 2007; 17:271-275
  328. Chin KP, Pin YC. Heterophile antibody interference with thyroid assay. Intern Med 2008; 47:2033-2037
  329. Tan MJ, Tan F, Hawkins R, Cheah WK, Mukherjee JJ. A hyperthyroid patient with measurable thyroid-stimulating hormone concentration - a trap for the unwary. Ann Acad Med Singapore 2006; 35:500-503
  330. Ross HA, Menheere PP, Thomas CM, Mudde AH, Kouwenberg M, Wolffenbuttel BH. Interference from heterophilic antibodies in seven current TSH assays. Ann Clin Biochem 2008; 45:616
  331. Giovanella L, Ghelfo A. Undetectable serum thyroglobulin due to negative interference of heterophile antibodies in relapsing thyroid carcinoma. Clin Chem 2007; 53:1871-1872
  332. Giovanella L, Keller F, Ceriani L, Tozzoli R. Heterophile antibodies may falsely increase or decrease thyroglobulin measurement in patients with differentiated thyroid carcinoma. Clin Chem Lab Med 2009; 47:952-954
  333. Petrovic. I, Mandel. S, Fatemi. S, J LoPresti, C Spencer. Heterophile Antibodies (HAb/HAMA) Interfere with Automated TgAb IMA Tests. Thyroid 2016; 26:A120
  334. Papapetrou PD, Polymeris A, Karga H, Vaiopoulos G. Heterophilic antibodies causing falsely high serum calcitonin values. J Endocrinol Invest 2006; 29:919-923
  335. Kim JM, Chung KW, Kim SW, Choi SH, Min HS, Kim JN, Won WJ, Kim SK, Lee JI, Chung JH, Kim SW. Spurious hypercalcitoninemia in patients with nodular thyroid disease induced by heterophilic antibodies. Head Neck 2010; 32:68-75
  336. Giovanella L, Suriano S. Spurious hypercalcitoninemia and heterophilic antibodies in patients with thyroid nodules. Head Neck 2011; 33:95-97
  337. Bories PN, Broutin A, Delette A, Labelle G, Popovici T. Comparison of the Elecsys calcitonin assay with the Immulite 1000 assay. Describing one case with heterophilic antibody interference. Clin Chem Lab Med 2016; 54:e45-47
  338. Nakano K, Yasuda K, Shibuya H, Moriyama T, Kahata K, Shimizu C. Transient human anti-mouse antibody generated with immune enhancement in a carbohydrate antigen 19-9 immunoassay after surgical resection of recurrent cancer. Ann Clin Biochem 2016; 53:511-515
  339. Sapin R, Agin A, Gasser F. Efficacy of a new blocker against anti-ruthenium antibody interference in the Elecsys free triiodothyronine assay. Clin Chem Lab Med 2007; 45:416-418
  340. Ando T, Yasui J, Inokuchi N, Usa T, Ashizawa K, Kamihara S, Eguchi K. Non-specific activities against ruthenium crosslinker as a new cause of assay interference in an electrochemilluminescent immunoassay. Intern Med 2007; 46:1225-1229
  341. Buijs MM, Gorgels JP, Endert E. Interference by antiruthenium antibodies in the Roche thyroid-stimulating hormone assay. Ann Clin Biochem 2011; 48:276-281
  342. Ohba K, Noh JY, Unno T, Satoh T, Iwahara K, Matsushita A, Sasaki S, Oki Y, Nakamura H. Falsely elevated thyroid hormone levels caused by anti-ruthenium interference in the Elecsys assay resembling the syndrome of inappropriate secretion of thyrotropin. Endocr J 2012; 59:663-667
  343. Gessl A, Blueml S, Bieglmayer C, Marculescu R. Anti-ruthenium antibodies mimic macro-TSH in electrochemiluminescent immunoassay. Clin Chem Lab Med 2014; 52:1589-1594
  344. Rulander NJ, Cardamone D, Senior M, Snyder PJ, Master SR. Interference from anti-streptavidin antibody. Arch Pathol Lab Med 2013; 137:1141-1146
  345. Vos MJ, Rondeel JM, Mijnhout GS, Endert E. Immunoassay interference caused by heterophilic antibodies interacting with biotin. Clin Chem Lab Med 2016;
  346. Kwok JS, Chan IH, Chan MH. Biotin interference on TSH and free thyroid hormone measurement. Pathology 2012; 44:278-280
  347. Wijeratne NG, Doery JC, Lu ZX. Positive and negative interference in immunoassays following biotin ingestion: a pharmacokinetic study. Pathology 2012; 44:674-675
  348. Elston MS, Sehgal S, Du Toit S, Yarndley T, Conaglen JV. Factitious Graves' Disease Due to Biotin Immunoassay Interference-A Case and Review of the Literature. J Clin Endocrinol Metab 2016; 101:3251-3255
  349. Pedersen IB, P L. Biochemical Hyperthyroidism in a Newborn Baby Caused by Assay Interaction from Biotin Intake. Eur Thyroid J 2016; 5:212-215
  350. Barbesino G. Misdiagnosis of Graves' Disease with Apparent Severe Hyperthyroidism in a Patient Taking Biotin Megadoses. Thyroid 2016; 26:860-863
  351. Lazarus JH, John R, Ginsberg J, Hughes IA, Shewring G, Smith BR, Woodhead JS, Hall R. Transient neonatal hyperthyrotrophinaemia: a serum abnormality due to transplacentally acquired antibody to thyroid stimulating hormone. British medical journal (Clinical research ed) 1983; 286:592-594
  352. Newman JD, Bergman PB, Doery JC, Balazs ND. Factitious increase in thyrotropin in a neonate caused by a maternally transmitted interfering substance. Clin Chem 2006; 52:541-542
  353. Benvenga S, Ordookhani A, Pearce EN, Tonacchera M, Azizi F, Braverman LE. Detection of circulating autoantibodies against thyroid hormones in an infant with permanent congenital hypothyroidism and her twin with transient congenital hypothyroidism: possible contribution of thyroid hormone autoantibodies to neonatal and infant hypothyroidism. J Pediatr Endocrinol Metab 2008; 21:1011-1020
  354. Rix M, Laurberg P, Porzig C, Kristensen SR. Elevated thyroid-stimulating hormone level in a euthyroid neonate caused by macro thyrotropin-IgG complex. Acta paediatrica (Oslo, Norway : 1992) 2011; 100:e135-137
  355. Sakata S, Matsuda M, Ogawa T, Takuno H, Matsui I, Sarui H, Yasuda K. Prevalence of thyroid hormone autoantibodies in healthy subjects. Clin Endocrinol (Oxf) 1994; 41:365-370
  356. Benvenga S, Trimarchi F. Thyroid hormone autoantibodies in Hashimoto's thyroiditis: often transient but also increasingly frequent. Thyroid 2003; 13:995-996; author reply 996
  357. Gangemi S, Saitta S, Lombardo G, Patafi M, Benvenga S. Serum thyroid autoantibodies in patients with idiopathic either acute or chronic urticaria. J Endocrinol Invest 2009; 32:107-110
  358. Colucci R, Lotti F, Dragoni F, Arunachalam M, Lotti T, Benvenga S, Moretti S. High prevalence of circulating autoantibodies against thyroid hormones in vitiligo and correlation with clinical and historical parameters of patients. The British journal of dermatology 2014; 171:786-798
  359. Garber JR, Cobin RH, Gharib H, Hennessey JV, Klein I, Mechanick JI, Pessah-Pollack R, Singer PA, Woeber KA. Clinical Practice Guidelines for Hypothyroidism in Adults: Co-sponsored by American Association of Clinical Endocrinologists and the American Thyroid Association. Endocr Pract 2012; 18:1-207
  360. Cobb WE, Lamberton RP, Jackson IMD. Use of a rapid, sensitive immunoradiometric assay for Thyrotropin to distinguish normal from hyperthyroid subjects. Clin Chem 1984; 30:1558-1560
  361. Piketty ML, Talbot JN, Askienazy S, Milhaud G. Clinical significance of a low concentration of Thyrotropin" five immunometric "kit" assays compared. Clin Chem 1987; 33:1237-1241
  362. Spencer CA, Nicoloff JT. Improved radioimmunoassay for human TSH. Clin Chim Acta 1980; 108:415-424
  363. Mori T, Imura H, Bito S, Ikekubo K, Inoue S, Hashida S, Ishikawa E, Ogawa H. Clinical usefulness of a highly sensitive enzyme-immunoassay of TSH. Clin Endocrinol 1987; 27:1-10
  364. Evans MC. Ten commercial kits compared for assay of Thyrotropin in the normal and thyrotoxic range. Clin Chem 1988; 34:123-127
  365. Yalow RS, Berson SA. Immunoassay of endogenous plasma insulin in man. J Clin Invest 1960; 39:1157-1175
  366. Utiger RD. Radioimmunoassay of human plasma thyrotropin. J Clin Invest 1965; 44:1277-1286
  367. Odell WD, Wilber JF, Paul WE. Radioimmunoassay of thyrotropin in human serum. J Clin Endocrinol 1965; 25:1179-1188
  368. Patel YC, Burger HG, Hudson B. Radioimmunoassay of serum thyrotropin: Sensitivity and Specificity. J Clin Endocrinol 1971; 33:768-774
  369. Hall R, Amos J, Ormston BJ. Radioimmunoassay of human serum thyrotropin. Br Med J 1971; 1:582-?
  370. Hershman JM. Utility of the radioimmunoassay of serum thyrotropin in man. Ann Intern Med 1971; 74:481-490
  371. Haigler ED, Pittman JA, Hershman JM, Baugh CM. Direct evaluation of pituitary Thyrotropin reserve utilizing synthetic Thyrotropin Releasing Hormone. J Clin Endocrinol Metab 1971; 33:573-581
  372. Hall R, Ormston BJ, Besser GM, Cryer RJ, McKendrick M. The thyrotropin-releasing hormone test in diseases of the pituitary and hypothalamus. Lancet 1972; i:759-763
  373. Evans M, Croxson MS, Wilson TM, Ibbertson HK. The screening of patients with suspected thyrotoxicosis using a sensitive TSH radioimmunoassay. Clin Endocrinol 1985; 22:445-451
  374. Spencer C, Schwarzbein D, Guttler R, LoPresti J, Nicoloff J. TRH stimulation test responses employing 3rd. and 4th. generation TSH assay technology. J Clin Endocrinol Metab 1993; 76:494-499
  375. Atmaca H, Tanriverdi F, Gokce C, Unluhizarci K, Kelestimur F. Do we still need the TRH stimulation test? Thyroid 2007; 17:529-533
  376. Duntas LH, Emerson CH. On the Fortieth Anniversary of Thyrotropin-Releasing Hormone: The Hormone that Launched a New Era. Thyroid 2009; 19:1299-1301
  377. Miles LEM, Hales CN. Labeled antibodies and immunological assay systems. Nature 1968; 219:186-189
  378. Kohler G, Milstein C. Continuous culture of fused cells secreting specific antibody of predefined specificity. Nature 1975; 256:495-497
  379. Winter G, Milstein C. Man-made antibodies. Nature 1991; 349:293-299
  380. Seth J, Kellett HA, Caldwell G, Sweeting VM, Beckett GJ, Gow SM, Toft AD. A sensitive immunoradiometric assay for serum thyroid stimulating hormone: a replacement for the thyrotropin releasing test. Br Med J 1984; 289:1334-1336
  381. Bernutz C, Horn K, Konig A, Pickardt CR. Advantages of sensitive assays for Thyrotropin in the diagnosis of thyroid disorders. J Clin Chem Clin Biochem 1985; 23:851-856
  382. Wiersinga WM, Endert E, Trip MD, Verhaest-de Jong N. Immunoradiometric assay of Thyrotropin in plasma: its value in predicting response to thyroliberin stimulation and assessing thyroid function in amiodarone-treated patients. Clin Chem 1986; 32:433-436
  383. Bassett F, Eastman CJ, Ma G, Maberly GF, Smith HC. Diagnostic value of Thyrotropin concentrations in serum as measured by a sensitive immunoradiometric assay. Clin Chem 1986; 32:461-464
  384. Martino E, Bambini G, Bartalena L, Mammoli C, Aghini-Lombardi F, Baschieri L, Pinchera A. Human serum thyrotropin measurement by ultrasensitive immunoradiometric assay as a first-line test in the evaluation of thyroid function. Clin Endocrinol 1986; 24:141-148
  385. Thornes HM, McLeod DT, Carr D. Economy and efficiency in routine thyroid-function testing: use of a sensitive immunoradiometric assay for thyrotropin in a General Hospital Laboratory. Clin Chem 1987; 33:1636-1638
  386. Klee GG, Hay ID. Assessment of sensitive Thyrotropin assays for an expanded role in thyroid function testing: proposed criteria for analytic performance and clinical utility. J Clin Endocrinol Metab 1987; 64:461-471
  387. Kasagi K, Kousaka T, Misaki T, Iwata M, Alam MS, Konishi J. Comparison of serum thyrotrophin concentrations determined by a third generation assay in patients with various types of overt and subclinical thyrotoxicosis. Clin Endocrinol 1999; 50:185-189
  388. Taimela E, Tahtela R, Koskinen P, Nuutila P, Forsstrom J, Taimela S, Karonen SL, Valimaki M, Irjala K. Ability of two new thyrotropin (TSH) assays to separate hyperthyroid patients from euthyroid patients with low TSH. Clin Chem 1994; 40:101-105
  389. Ross DS, Daniels GH, Gouveia D. The use and limitations of a chemiluminescent thyrotropin assay as a single thyroid function test in an out-patient endocrine clinic. J Clin Endocrinol Metab 1990; 71:764-769
  390. Hay ID, Bayer MF, Kaplan MM, Klee GG, Larsen PR, Spencer CA. American Thyroid Association Assessment of Current Free Thyroid Hormone and Thyrotropin Measurements and Guidelines for Future Clinical Assays. Clin Chem 1991; 37:2002 - 2008
  391. Spencer CA, Takeuchi M, Kazarosyn M, MacKenzie F, Beckett GJ, Wilkinson E. Interlaboratory/intermethod differences in functional sensitivity of immunometric assays for thyrotropin (TSH): impact on reliability of measurement of subnormal concentration. Clin Chem 1995; 41:367-374
  392. Vogeser M, Weigand M, Fraunberger P, Fischer H, Cremer P. Evaluation of the ADVIA Centaur TSH-3 assay. Clin Chem Lab Med 2000; 38:331-334
  393. Hendriks HA, Kortlandt W, Verweij WM. Standardized comparison of processing capacity and efficiency of five new-generation immunoassay analyzers. Clin Chem 2000; 46:105-111
  394. Thienpont LM, Van Houcke SK. Traceability to a common standard for protein measurements by immunoassay for in-vitro diagnostic purposes. Clin Chim Acta 2010; 411:2058-2061
  395. Owen WE, Gantzer ML, Lyons JM, Rockwood AL, Roberts WL. Functional sensitivity of seven automated thyroid stimulating hormone immunoassays. Clin Chim Acta 2011; 412:2336-2339
  396. Sarkar R. TSH Comparison Between Chemiluminescence (Architect) and Electrochemiluminescence (Cobas) Immunoassays: An Indian Population Perspective. Indian journal of clinical biochemistry : IJCB 2014; 29:189-195
  397. Nicoloff J SC. Use and misuse of the sensitive thyrotropin assays. J Clin Endocrinol Metab 1990; 71:553-558
  398. Fraser CG, Browning MCK. A plea for abandonment of the term "Highly Sensitive" for Thyrotropin assays. Clin Chem 1986; 32:569-570
  399. Caldwell G, Gow SM, Sweeting VM, Beckett GJ, Seth J, Toft AD. Value and limitations of a highly sensitive immunoradioimetric assay for Thyrotropin in the study of thyrotroph function. Clin Chem 1987; 33:303-305
  400. Hildebrandt L, White GH. Is a "Super-Sensitive" thyrotropin assay ("Magic Lite") of more diagnostic value? Clin Chem 1988; 34:2584-2585
  401. Surmont DWA, Alexandre JA. Adaptations to keep a Thyrotropin immunoradiometric assay "supersensitive" with automated pipetting. Clin Chem 1988; 34:370-371
  402. Gaines-Das RE, Brettschneider H, Bristow AF. International Federation of Clinical Chemistry. The effects of common matrices for assay standards on the performance of "ultra sensitive" immunometric assays for TSH. Clin Chim Acta 1991; 203:S5-16
  403. Sadler WA, Murray LM, Turner JG. What does "functional sensitivity" mean? Clin Chem 1996; 42:2051
  404. Giovanella L, Feldt-Rasmussen U, Verburg FA, Grebe SK, Plebani M, Clark PM. Thyroglobulin measurement by highly sensitive assays: focus on laboratory challenges. Clin Chem Lab Med 2015; 53:1301-1314
  405. Reix N, Massart C, Gasser F, Heurtault B, Agin A. Should functional sensitivity of a new thyroid stimulating hormone immunoassay be monitored routinely? The ADVIA Centaur TSH3-UL assay experience. Clin Biochem 2012; 45:1260-1262
  406. Rigo RB, Panyella MG, Bartolome LR, Ramos PA, Soria PR, Navarro MA. Variations observed for insulin concentrations in an interlaboratory quality control program may be due to interferences between reagents and the matrix of the control materials. Clin Biochem 2007; 40:1088-1091
  407. Ross HA, Netea-Maier RT, Schakenraad E, Bravenboer B, Hermus AR, Sweep FC. Assay bias may invalidate decision limits and affect comparability of serum thyroglobulin assay methods: an approach to reduce interpretation differences. Clin Chim Acta 2008; 394:104-109
  408. Algeciras-Schimnich A, Bruns DE, Boyd JC, Bryant SC, La Fortune KA, Grebe SK. Failure of current laboratory protocols to detect lot-to-lot reagent differences: findings and possible solutions. Clin Chem 2013; 59:1187-1194
  409. Hayden JA, Schmeling M, Hoofnagle AN. Lot-to-lot variations in a qualitative lateral-flow immunoassay for chronic pain drug monitoring. Clin Chem 2014; 60:896-897
  410. Thaler MA, Iakoubov R, Bietenbeck A, Luppa PB. Clinically relevant lot-to-lot reagent difference in a commercial immunoturbidimetric assay for glycated hemoglobin A1c. Clin Biochem 2015; 48:1167-1170
  411. Armbruster DA, Pry T. Limit of blank, limit of detection and limit of quantitation. Clin Biochem Rev 2008; 29 Suppl 1:S49-52
  412. Spencer C, Fatemi S, Singer P, Nicoloff J, Lopresti J. Serum Basal thyroglobulin measured by a second-generation assay correlates with the recombinant human thyrotropin-stimulated thyroglobulin response in patients treated for differentiated thyroid cancer. Thyroid 2010; 20:587-595
  413. Giovanella L, Clark PM, Chiovato L, Duntas L, Elisei R, Feldt-Rasmussen U, Leenhardt L, Luster M, Schalin-Jantti C, Schott M, Seregni E, Rimmele H, Smit J, Verburg FA. Thyroglobulin measurement using highly sensitive assays in patients with differentiated thyroid cancer: a clinical position paper. Eur J Endocrinol 2014; 171:R33-46
  414. Rawlins ML, Roberts WL. Performance characteristics of six third-generation assays for thyroid-stimulating hormone. Clin Chem 2004; 50:2338-2344
  415. Iervasi A, Iervasi G, Bottoni A, Boni G, Annicchiarico C, Di Cecco P, Zucchelli GC. Diagnostic performance of a new highly sensitive thyroglobulin immunoassay. J Endocrinol 2004; 182:287-294
  416. Smallridge RC, Meek SE, Morgan MA, Gates GS, Fox TP, Grebe S, Fatourechi V. Monitoring thyroglobulin in a sensitive immunoassay has comparable sensitivity to recombinant human TSH-stimulated thyroglobulin in follow-up of thyroid cancer patients. J Clin Endocrinol Metab 2007; 92:82-87
  417. Andersen S, Pedersen KM, Bruun NH, Laurberg P. Narrow individual variations in serum T(4) and T(3) in normal subjects: a clue to the understanding of subclinical thyroid disease. J Clin Endocrinol Metab 2002; 87:1068-1072
  418. Boas M, Forman JL, Juul A, Feldt-Rasmussen U, Skakkebaek NE, Hilsted L, Chellakooty M, Larsen T, Larsen JF, Petersen JH, Main KM. Narrow intra-individual variation of maternal thyroid function in pregnancy based on a longitudinal study on 132 women. Eur J Endocrinol 2009; 161:903-910
  419. Meikle AW, Stringham JD, Woodward MG, Nelson JC. Hereditary and environmental influences on the variation of thyroid hormones in normal male twins. J Clin Endocrinol Metab 1988; 66:588-592
  420. Panicker V, Wilson SG, Spector TD, Brown SJ, Falchi M, Richards JB, Surdulescu GL, Lim EM, Fletcher SJ, Walsh JP. Heritability of serum TSH, free T4 and free T3 concentrations: a study of a large UK twin cohort. Clin Endocrinol 2008; 68:652-659
  421. Nilsson SE, Read S, Berg S, Johansson B. Heritabilities for fifteen routine biochemical values: findings in 215 Swedish twin pairs 82 years of age or older. Scand J Clin Lab Invest 2009; 69:562-569
  422. Hansen PS, Brix TH, Sørensen TI, Kyvik KO, Hegedüs L. Major genetic influence on the regulation of the pituitary-thyroid axis: a study of healthy Danish twins. J Clin Endocrinol Metab 2004; 89:1181-1187
  423. Peeters RP, van der Deure WM, Visser TJ. Genetic variation in thyroid hormone pathway genes; polymorphisms in the TSH receptor and the iodothyronine deiodinases. Eur J Endocrinol 2006; 155:655-662
  424. Arnaud-Lopez L, Usala G, Ceresini G, Mitchell BD, Pilia MG, Piras MG, Sestu N, Maschio A, Busonero F, Albai G, Dei M, Lai S, Mulas A, Crisponi L, Tanaka T, Bandinelli S, Guralnik JM, Loi A, Balaci L, Sole G, Prinzis A, Mariotti S, Shuldiner AR, Cao A, Schlessinger D, Uda M, Abecasis GR, Nagaraja R, Sanna S, Naitza S. Phosphodiesterase 8B gene variants are associated with serum TSH levels and thyroid function. American journal of human genetics 2008; 82:1270-1280
  425. Shields BM, Freathy RM, Knight BA, Hill A, Weedon MN, Frayling TM, Hattersley AT, Vaidya B. Phosphodiesterase 8B gene polymorphism is associated with subclinical hypothyroidism in pregnancy. J Clin Endocrinol Metab 2009; 94:4608-4612
  426. Bertalan R, Sallai A, Solyom J, Lotz G, Szabo I, Kovacs B, Szabo E, Patocs A, Racz K. Hyperthyroidism caused by a germline activating mutation of the thyrotropin receptor gene: difficulties in diagnosis and therapy. Thyroid 2010; 20:327-332
  427. Biebermann H, Winkler F, Handke D, Gruters A, Krude H, Kleinau G. Molecular description of non-autoimmune hyperthyroidism at a neonate caused by a new thyrotropin receptor germline mutation. Thyroid research 2011; 4 Suppl 1:S8
  428. Scaglia PA, Chiesa A, Bastida G, Pacin M, Domene HM, Gruneiro-Papendieck L. Severe congenital non-autoimmune hyperthyroidism associated to a mutation in the extracellular domain of thyrotropin receptor gene. Arq Bras Endocrinol Metabol 2012; 56:513-518
  429. Agretti P, Segni M, De Marco G, Ferrarini E, Di Cosmo C, Corrias A, Weber G, Larizza D, Calcaterra V, Pelizzo MR, Cesaretti G, Vitti P, Tonacchera M. Prevalence of activating thyrotropin receptor and Gsalpha gene mutations in paediatric thyroid toxic adenomas: a multicentric Italian study. Clin Endocrinol (Oxf) 2013; 79:747-749
  430. Grob F, Deladoey J, Legault L, Spigelblatt L, Fournier A, Vassart G, Van Vliet G. Autonomous adenomas caused by somatic mutations of the thyroid-stimulating hormone receptor in children. Horm Res Paediatr 2014; 81:73-79
  431. Nakamura A, Morikawa S, Aoyagi H, Ishizu K, Tajima T. A Japanese family with nonautoimmune hyperthyroidism caused by a novel heterozygous thyrotropin receptor gene mutation. Pediatr Res 2014; 75:749-753
  432. Kleinau G, Biebermann H. Constitutive activities in the thyrotropin receptor: regulation and significance. Advances in pharmacology (San Diego, Calif) 2014; 70:81-119
  433. Larsen CC, Karaviti LP, Seghers V, Weiss RE, Refetoff S, Dumitrescu AM. A new family with an activating mutation (G431S) in the TSH receptor gene: a phenotype discussion and review of the literature. International journal of pediatric endocrinology 2014; 2014:23
  434. Alberti L, Proverbio MC, Costagliola S, Romoli R, Boldrighini B, Vigone MC, Weber G, Chiumello G, Beck-Peccoz P, Persani L. Germline mutations of TSH receptor gene as cause of nonautoimmune subclinical hypothyroidism. J Clin Endocrinol Metab 2002; 87:2549-2555
  435. Persani L, Calebiro D, Cordella D, Weber G, Gelmini G, Libri D, de Filippis T, Bonomi M. Genetics and phenomics of hypothyroidism due to TSH resistance. Mol Cell Endocrinol 2010; 322:72-82
  436. Calebiro D, Gelmini G, Cordella D, Bonomi M, Winkler F, Biebermann H, de Marco A, Marelli F, Libri DV, Antonica F, Vigone MC, Cappa M, Mian C, Sartorio A, Beck-Peccoz P, Radetti G, Weber G, Persani L. Frequent TSH receptor genetic alterations with variable signaling impairment in a large series of children with nonautoimmune isolated hyperthyrotropinemia. J Clin Endocrinol Metab 2012; 97:E156-160
  437. Rapa A, Monzani A, Moia S, Vivenza D, Bellone S, Petri A, Teofoli F, Cassio A, Cesaretti G, Corrias A, de Sanctis V, Di Maio S, Volta C, Wasniewska M, Tatò L, Bona G. Subclinical hypothyroidism in children and adolescents: a wide range of clinical, biochemical, and genetic factors involved. J Clin Endocrinol Metab 2009; 94:2414-2420
  438. De Marco G, Agretti P, Camilot M, Teofoli F, Tatò L, Vitti P, Pinchera A, Tonacchera M. Functional studies of new TSH receptor (TSHr) mutations identified in patients affected by hypothyroidism or isolated hyperthyrotrophinaemia. Clin Endocrinol 2009; 70:335-338
  439. Brouwer JP, Appelhof BC, Peeters RP, Hoogendijk WJ, Huyser J, Schene AH, Tijssen JG, Van Dyck R, Visser TJ, Wiersinga WM, Fliers E. Thyrotropin, but not a polymorphism in type II deiodinase, predicts response to paroxetine in major depression. Eur J Endocrinol 2006; 154:819-825
  440. Takeda K, Mishiba M, Sugiura H, Nakajima A, Kohama M, Hiramatsu S. Evaluated reference intervals for serum free thyroxine and thyrotropin using the conventional outliner rejection test without regard to presence of thyroid antibodies and prevalence of thyroid dysfunction in Japanese subjects. Endoc J 2009; 56:1059-1066
  441. Andersen S, Bruun NH, Pedersen KM, Laurberg P. Biologic variation is important for interpretation of thyroid function tests. Thyroid 2003; 13:1069-1078
  442. Jensen E, Petersen PH, Blaabjerg O, Hegedüs L. Biological variation of thyroid autoantibodies and thyroglobulin. Clin Chem Lab Med 2007; 45:1058-1064
  443. Ankrah-Tetteh T, Wijeratne S, Swaminathan R. Intraindividual variation in serum thyroid hormones, parathyroid hormone and insulin-like growth factor-1. Ann Clin Biochem 2008; 45:167-169
  444. van de Ven AC, Netea-Maier RT, Medici M, Sweep FC, Ross HA, Hofman A, de Graaf J, Kiemeney LA, Hermus AR, Peeters RP, Visser TJ, den Heijer M. Underestimation of effect of thyroid function parameters on morbidity and mortality due to intra-individual variation. J Clin Endocrinol Metab 2011; 96:E2014-2017
  445. Biondi B, Cooper DS. The clinical significance of subclinical thyroid dysfunction. Endocr Rev 2008; 29:76-131
  446. Cooper DS, Biondi B. Subclinical thyroid disease. Lancet 2012; 379:1142-1154
  447. Silvio R, Swapp KJ, La'ulu SL, Hansen-Suchy K, Roberts WL. Method specific second-trimester reference intervals for thyroid-stimulating hormone and free thyroxine. Clin Biochem 2009; 42:750-753
  448. Coene KL, Demir AY, Broeren MA, Verschuure P, Lentjes EG, Boer AK. Subclinical hypothyroidism: a 'laboratory-induced' condition? Eur J Endocrinol 2015; 173:499-505
  449. Strich D, Karavani G, Levin S, Edri S, Gillis D. Normal limits for serum thyrotropin vary greatly depending on method. Clin Endocrinol (Oxf) 2016; 85:110-115
  450. Solberg HE. The IFCC recommendation on estimation of reference intervals. Clin Chem Lab Med 2004; 42:710-714
  451. Kahapola-Arachchige KM, Hadlow N, Wardrop R, Lim EM, Walsh JP. Age-specific TSH reference ranges have minimal impact on the diagnosis of thyroid dysfunction. Clin Endocrinol (Oxf) 2012; 77:773-779
  452. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, Braverman LE. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab 2002; 87:489-499
  453. Hollowell JG, Staehling NW, Hannon WH, Flanders DW, Gunter EW, Maberly GF, Braverman LE, Pino S, Miller DT, Garbe PL, DeLozier DM, Jackson RJ. Iodine nutrition in the United States. Trends and public health implications: iodine excretion data from National Health and Nutrition Examination Surveys I and III (1971-1974 and 1988-1994). J Clin Endocrinol Metab 1998; 83:3401-3408
  454. McNeil AR, Stanford PE. Reporting Thyroid Function Tests in Pregnancy. Clin Biochem Rev 2015; 36:109-126
  455. Kristensen GB, Rustad P, Berg JP, Aakre KM. Analytical Bias Exceeding Desirable Quality Goal in 4 out of 5 Common Immunoassays: Results of a Native Single Serum Sample External Quality Assessment Program for Cobalamin, Folate, Ferritin, Thyroid-Stimulating Hormone, and Free T4 Analyses. Clin Chem 2016; 62:1255-1263
  456. Spencer CA, Hollowell JG, Kazarosyan M, Braverman LE. National Health and Nutrition Examination Survey III thyroid-stimulating hormone (TSH)-thyroperoxidase antibody relationships demonstrate that TSH upper reference limits may be skewed by occult thyroid dysfunction. J Clin Endocrinol Metab 2007; 92:4236-4240
  457. Bjoro T, Holmen J, Kruger O, Midthjell K, Hunstad K, Schreiner T, Sandnes L, Brochmann H. Prevalence of thyroid disease, thyroid dysfunction and thyroid peroxidase antibodies in a large, unselected population. The Health Study of Nord-Trondelag (HUNT). Eur J Endocrinol 2000; 143:639-647
  458. Pedersen OM, Aardal NP, Larssen TB, Varhaug JE, Myking O, Vik-Mo H. The value of ultrasonography in predicting autoimmune thyroid disease. Thyroid 2000; 10:251-259
  459. Jensen E, Hyltoft Petersen P, Blaabjerg O, Hansen PS, Brix TH, Kyvik KO, Hegedus L. Establishment of a serum thyroid stimulating hormone (TSH) reference interval in healthy adults. The importance of environmental factors, including thyroid antibodies. Clin Chem Lab Med 2004; 42:824-832
  460. Surks MI, Hollowell JG. Age-specific distribution of serum thyrotropin and antithyroid antibodies in the US population: implications for the prevalence of subclinical hypothyroidism. J Clin Endocrinol Metab 2007; 92:4575-4582
  461. Zelaya AS, Stotts A, Nader S, Moreno CA. Antithyroid peroxidase antibodies in patients with high normal range thyroid stimulating hormone. Fam Med 2010; 42:111-115
  462. Sichieri R, Baima J, Marante T, de Vasconcellos MT, Moura AS, Vaisman M. Low prevalence of hypothyroidism among black and Mulatto people in a population-based study of Brazilian women. Clin Endocrinol 2007; 66:803-807
  463. Boucai L SM. Reference limits of serum TSH and free T4 are significantly influenced by race and age in an urban outpatient medical practice. Clin Endocrinol 2009; 70:788-793
  464. Vejbjerg P, Knudsen N, Perrild H, Carlé A, Laurberg P, Pedersen IB, Rasmussen LB, Ovesen L, Jørgensen T. The impact of smoking on thyroid volume and function in relation to a shift towards iodine sufficiency. Eur J Epidemiol 2008; 23:423-429
  465. Buchinger W, Lorenz-Wawschinek O, Semlitsch G, Langsteger W, Binter G, Bonelli RM, Eber O. Thyrotropin and thyroglobulin as an index of optimal iodine intake: correlation with iodine excretion of 39,913 euthyroid patients. Thyroid 1997; 7:593-597
  466. Nyrnes A, Jorde R, Sundsfjord J. Serum TSH is positively associated with BM. Int J Obes 2006; 30:100-105
  467. Knudsen N, Laurberg P, Rasmussen LB, Bulow I, Perrild H, Ovesen L, Jorgensen T. Small differences in thyroid function may be important for body mass index and the occurrence of obesity in the population. J Clin Endocrinol Metab 2005; 90:4019-4024
  468. Kok P, Roelfsema F, Langendonk JG, Frolich M, Burggraaf J, Meinders AE, Pijl H. High circulating thyrotropin levels in obese women are reduced after body weight loss induced by caloric restriction. J Clin Endocrinol Metab 2005; 90:4659-4663
  469. Chikunguwo S, Brethauer S, Nirujogi V, Pitt T, Udomsawaengsup S, Chand B, Schauer P. Influence of obesity and surgical weight loss on thyroid hormone levels. Surg Obes Relat Dis 2007; 3:631-635
  470. Fox CS, Pencina MJ, D'Agostino RB, Murabito JM, Seely EW, Pearce EN, Vasan RS. Relations of thyroid function to body weight: cross-sectional and longitudinal observations in a community-based sample. Arch Intern Med 2008; 168:587-592
  471. Friedrich N, Rosskopf D, Brabant G, Völzke H, Nauck M, Wallaschofski H. Associations of anthropometric parameters with serum TSH, prolactin, IGF-I, and testosterone levels: results of the study of health in Pomerania (SHIP). Exp Clin Endocrinol Diabetes 2010; 118:266-273
  472. Biondi B. Thyroid and obesity: an intriguing relationship. J Clin Endocrinol Metab 2010; 95:3614-3617
  473. Solanki A, Bansal S, Jindal S, Saxena V, Shukla US. Relationship of serum thyroid stimulating hormone with body mass index in healthy adults. Indian journal of endocrinology and metabolism 2013; 17:S167-169
  474. Taylor PN, Razvi S, Pearce SH, Dayan CM. Clinical review: A review of the clinical consequences of variation in thyroid function within the reference range. J Clin Endocrinol Metab 2013; 98:3562-3571
  475. Duntas LH, Biondi B. The interconnections between obesity, thyroid function, and autoimmunity: the multifold role of leptin. Thyroid 2013; 23:646-653
  476. Santini F, Marzullo P, Rotondi M, Ceccarini G, Pagano L, Ippolito S, Chiovato L, Biondi B. Mechanisms in endocrinology: the crosstalk between thyroid gland and adipose tissue: signal integration in health and disease. Eur J Endocrinol 2014; 171:R137-152
  477. Betry C, Challan-Belval MA, Bernard A, Charrie A, Drai J, Laville M, Thivolet C, Disse E. Increased TSH in obesity: Evidence for a BMI-independent association with leptin. Diabetes & metabolism 2015; 41:248-251
  478. Asvold BO, Vatten LJ, Nilsen TI, Bjoro T. The association between TSH within the reference range and serum lipid concentrations in a population-based study. The HUNT Study. Eur J Endocrinol 2007; 156:181-186
  479. Soldin OP, Goughenour BE, Gilbert SZ, Landy HJ, Soldin SJ. Thyroid Hormone Levels Associated with Active and Passive Cigarette Smoking. Thyroid 2010; 20:0-0
  480. Surks MI, Boucai L. Age- and race-based serum Thyrotropin Reference Limits. J Clin Endocrinol Metab 2010; 95:496 -502
  481. Atzmon G, Barzilai N, Hollowell JG, Surks MI, Gabriely I. Extreme longevity is associated with increased serum thyrotropin. J Clin Endocrinol Metab 2009; 94:1251-1254
  482. Bremner AP, Feddema P, Leedman PJ, Brown SJ, Beilby JP, Lim EM, Wilson SG, O'Leary PC, Walsh JP. Age-related changes in thyroid function: a longitudinal study of a community-based cohort. J Cln Endocrinol Metab 2012; 97:1554-1562
  483. Hoogendoorn EH, Hermus AR, de Vegt F, Ross HA, Verbeek AL, Kiemeney LA, Swinkels DW, Sweep FC, den Heijer M. Thyroid function and prevalence of anti-thyroperoxidase antibodies in a population with borderline sufficient iodine intake: influences of age and sex. Clin Chem 2006; 52:104-111
  484. Volzke H, Alte D, Kohlmann T, Ludemann J, Nauck M, John U, Meng W. Reference intervals of serum thyroid function tests in a previously iodine-deficient area. Thyroid 2005; 15:279-285
  485. Kratzsch J, Fiedler GM, Leichtle A, Brugel M, Buchbinder S, Otto L, Sabri O, Matthes G, Thiery J. New reference intervals for thyrotropin and thyroid hormones based on national academy of clinical biochemistry criteria and regular ultrasonography of the thyroid. Clin Chem 2005; 51:1480-1486
  486. Berghout A, Wiersinga WM, Smits NJ, touber JL. Interrelationships between age, thyroid volume, thyroid nodularity, and thyroid function in patients with sporadic nontoxic goiter. Amer J Med 1990; 89:602-608
  487. Meisinger C, Ittermann T, Wallaschofski H, Heier M, Below H, Kramer A, Doring A, Nauck M, Volzke H. Geographic variations in the frequency of thyroid disorders and thyroid peroxidase antibodies in persons without former thyroid disease within Germany. Eur J Endocrinol 2012; 167:363-371
  488. Ittermann T, Khattak RM, Nauck M, Cordova CM, Volzke H. Shift of the TSH reference range with improved iodine supply in Northeast Germany. Eur J Endocrinol 2015; 172:261-267
  489. Gussekloo J, van Exel E, de Craen AJ, Meinders AE, Frölich M, Westendorp RG. Thyroid status, disability and cognitive function, and survival in old age. JAMA 2004; 292:2591-2599
  490. Simonsick EM, Newman AB, Ferrucci L, Satterfield S, Harris TB, Rodondi N, Bauer DC. Subclinical hypothyroidism and functional mobility in older adults. Arch Intern Med 2009; 169:2011-2017
  491. Atzmon G, Barzilai N, Surks MI, Gabriely I. Genetic predisposition to elevated serum thyrotropin is associated with exceptional longevity. J Clin Endocrinol Metab 2009; 94:4768-4775
  492. Rozing MP, Houwing-Duistermaat JJ, Slagboom PE, Beekman M, Frölich M, de Craen AJ, Westendorp RG, van Heemst D. Familial longevity is associated with decreased thyroid function. J Clin Endocrinol Metab 2010; 95:4979-4984
  493. van den Beld AW, Visser TJ, Feelders RA, Grobbee DE, Lamberts SW. Thyroid hormone concentrations, disease, physical function, and mortality in elderly men. J Clin Endocrinol Metab 2005; 90:6403-6409
  494. Akirov A, Gimbel H, Grossman A, Shochat T, Shimon I. Elevated TSH in adults treated for hypothyroidism is associated with increased mortality. Eur J Endocrinol 2016;
  495. Taylor PN, Panicker V, Sayers A, Shields B, Iqbal A, Bremner AP, Beilby JP, Leedman PJ, Hattersley AT, Vaidya B, Frayling T, Evans J, Tobias JH, Timpson NJ, Walsh JP, Dayan CM. A meta-analysis of the associations between common variation in the PDE8B gene and thyroid hormone parameters, including assessment of longitudinal stability of associations over time and effect of thyroid hormone replacement. Eur J Endocrinol 2011; 164:773-780
  496. Somwaru LL, Rariy CM, Arnold AM, Cappola AR. The natural history of subclinical hypothyroidism in the elderly: the cardiovascular health study. J Clin Endocrinol Metab 2012; 97:1962-1969
  497. Magner JA. Thyroid-Stimulating Hormone: Biosynythesis, Cell Biology, and Bioactivity. Endocrinol Rev 1990; 11:354-385
  498. Estrada JM, Soldin D, Buckey TM, Burman KD, Soldin OP. Thyrotropin isoforms: implications for thyrotropin analysis and clinical practice. Thyroid 2014; 24:411-423
  499. Persani L. Hypothalamic thyrotropin-releasing hormone and thyrotropin biological activity. Thyroid 1998; 8:941-946
  500. Sergi I, Papandreou MJ, Medri G, Canonne C, Verrier B, Ronin C. Immunoreactive and bioactive isoforms of human thyrotropin. Endocrinology 1991; 128:3259-3268
  501. Donadio S, Morelle W, Pascual A, Romi-Lebrun R, Michalski JC, Ronin C. Both core and terminal glycosylation alter epitope expression in thyrotropin and introduce discordances in hormone measurements. Clin Chem Lab Med 2005; 43:519-530
  502. Schaaf L, Trojan J, Helton TE, Usadel KH, Magner JA. Serum thyrotropin (TSH) heterogeneity in euthyroid subjects and patients with subclinical hypothyroidism: the core fucose content of TSH-releasing hormone-released TSH is altered, but not the net charge of TSH. J Endocrinol 1995; 144:561-571
  503. Szkudlinski MW, Fremont V, Ronin C, Weintraub BD. Thyroid-stimulating hormone and thyroid-stimulating hormone receptor structure-function relationships. Physiol Rev 2002; 82:473-502
  504. Oliveira JH, Barbosa ER, Kasamatsu T, Abucham J. Evidence for thyroid hormone as a positive regulator of serum thyrotropin bioactivity. J Clin Endocrinol Metab 2007; 92:3108-3113
  505. Barreca T, Franceschini R, Messina V, Bottaro L, Rolandi E. 24-hour thyroid-stimulating hormone secretory pattern in elderly men. Gerentology 1985; 31:119-123
  506. van Coevorden A, Laurent E, Decoster C, Kerkhofs M, Neve P, van Cauter E, Mockel J. Decreased basal and stimulated thyrotropin secretion in healthy elderly men. J Clin Endocrinol Metab 1989; 69:177-185
  507. Romijn JA, Wiersinga WM. Decreased nocturnal surge of thyrotropin in nonthyroidal illness. J Clin Endocrinol Metab 1990; 70:35-42
  508. Persani L, Asteria C, Tonacchera M, Vitti P, Krishna V, Chatterjee K, Beck-Peccoz P. Evidence for the secretion of thyrotropin with enhanced bioactivity in syndromes of thyroid hormone resistance. J Clin Endocrinol Metab 1994; 78:1034-1039
  509. Persani L. Clinical review: Central hypothyroidism: pathogenic, diagnostic, and therapeutic challenges. J Clin Endocrinol Metab 2012; 97:3068-3078
  510. Chan MK, Seiden-Long I, Aytekin M, Quinn F, Ravalico T, Ambruster D, Adeli K. Canadian Laboratory Initiative on Pediatric Reference Interval Database (CALIPER): pediatric reference intervals for an integrated clinical chemistry and immunoassay analyzer, Abbott ARCHITECT ci8200. Clin Biochem 2009; 42:885-891
  511. Zurakowski D, Di Canzio J, Majzoub JA. Pediatric reference intervals for serum thyroxine, triiodothyronine, thyrotropin and free thyroxine. Clin Chem 1999; 45:1087-1091
  512. Kapelari K, Kirchlechner C, Högler W, Schweitzer K, Virgolini I, Moncayo R. Pediatric reference intervals for thyroid hormone levels from birth to adulthood: a retrospective study. BMC endocrine disorders 2008; 8:15
  513. Lazar L, Frumkin RB, Battat E, Lebenthal Y, Phillip M, Meyerovitch J. Natural history of thyroid function tests over 5 years in a large pediatric cohort. J Clin Endocrinol Metab 2009; 94:1678-1682
  514. Strich D, Edri S, Gillis D. Current normal values for TSH and FT3 in children are too low: evidence from over 11,000 samples. J Pediatr Endocrinol Metab 2012; 25:245-248
  515. d'Herbomez M, Jarrige V, Darte C. Reference intervals for serum thyrotropin (TSH) and free thyroxine (FT4) in adults using the Access Immunoassay System. Clin Chem Lab Med 2005; 43:102-105
  516. Zarković M, Cirić J, Beleslin B, Cirić S, Bulat P, Topalov D, Trbojević B. Further studies on delineating thyroid-stimulating hormone (TSH) reference range. Horm Metab Res 2011; 43:970-976
  517. Hamilton TE, Davis S, Onstad L, Kopecky KJ. Thyrotropin levels in a population with no clinical, autoantibody, or ultrasonographic evidence of thyroid disease: implications for the diagnosis of subclinical hypothyroidism. J Clin Endocrinol Metab 2008; 93:1224-1230
  518. Goichot B, Sapin R, Schlienger JL. Subclinical hyperthyroidism: considerations in defining the lower limit of the thyrotropin reference interval. Clin Chem 2009; 55:420-424
  519. Völzke H, Schmidt CO, John U, Wallaschofski H, Dörr M, Nauck M. Reference levels for serum thyroid function tests of diagnostic and prognostic significance. Horm Metab Res 2010; 42:809-814
  520. O'Leary PC, Feddema PH, Michelangeli VP, Leedman PJ, Chew GT, Knuiman M, Kaye J, Walsh JP. Investigations of thyroid hormones and antibodies based on a community health survey: the Busselton thyroid study. Clin Endocrinol 2006; 64:97-104
  521. Canaris GJ, Manowitz NR, Mayor G, Ridgway EC. The Colorado Thyroid Disease Prevalence Study. Arch Intern Med 2000; 160:19-27
  522. Sawin CT. Subclinical hyperthyroidism and atrial fibrillation. Thyroid 2002; 12:501-503
  523. Choi AR, Manning P. Overshooting the mark: subclinical hyperthyroidism secondary to excess thyroid hormone treatment may be more prevalent than we realise. NZ Med J 2009; 122:93-94
  524. Ochs N, Auer R, Bauer DC, Nanchen D, Gussekloo J, Cornuz J, Rodondi N. Meta-analysis: subclinical thyroid dysfunction and the risk for coronary heart disease and mortality. Ann Intern Med 2008; 148:832-845
  525. Murphy E, Gluer CC, Reid DM, Felsenberg D, Roux C, Eastell R, Williams GR. Thyroid function within the upper normal range is associated with reduced bone mineral density and an increased risk of nonvertebral fractures in healthy euthyroid postmenopausal women. J Clin Endocrinol Metab 2010; 95:3173-3181
  526. Blum MR, Bauer DC, Collet TH, Fink HA, Cappola AR, da Costa BR, Wirth CD, Peeters RP, Asvold BO, den Elzen WP, Luben RN, Imaizumi M, Bremner AP, Gogakos A, Eastell R, Kearney PM, Strotmeyer ES, Wallace ER, Hoff M, Ceresini G, Rivadeneira F, Uitterlinden AG, Stott DJ, Westendorp RG, Khaw KT, Langhammer A, Ferrucci L, Gussekloo J, Williams GR, Walsh JP, Juni P, Aujesky D, Rodondi N. Subclinical thyroid dysfunction and fracture risk: a meta-analysis. Jama 2015; 313:2055-2065
  527. Collet TH, Gussekloo J, Bauer DC, den Elzen WP, Cappola AR, Balmer P, Iervasi G, Asvold BO, Sgarbi JA, Volzke H, Gencer B, Maciel RM, Molinaro S, Bremner A, Luben RN, Maisonneuve P, Cornuz J, Newman AB, Khaw KT, Westendorp RG, Franklyn JA, Vittinghoff E, Walsh JP, Rodondi N. Subclinical hyperthyroidism and the risk of coronary heart disease and mortality. Arch Intern Med 2012; 172:799-809
  528. Wartofsky L, Dickey RA. The evidence for a narrower thyrotropin reference range is compelling. J Clin Endocrinol Metab 2005; 90:5483-5488
  529. Surks MI. Should the upper limit of the normal reference range for TSH be lowered? Nat Clin Pract Endocrinol Metab 2008; 4:370-371
  530. Laurberg P, Andersen S, Carlé A, Karmisholt J, Knudsen N, Pedersen IB. The TSH upper reference limit: where are we at? Nat Rev Endocrinol 2011; 7:232-239
  531. McQuade C, Skugor M, Brennan DM, Hoar B, Stevenson C, Hoogwerf BJ. Hypothyroidism and moderate subclinical hypothyroidism are associated with increased all-cause mortality independent of coronary heart disease risk factors: a PreCIS database study. Thyroid 2011; 21:837-843
  532. Vanderpump MPJ, Tunbridge WMG, French JM, Appleton D, Bates D, Rodgers H, Evans JG, Clark F, Tunbridge F, Young ET. The incidence of thyroid disorders in the community; a twenty year follow up of the Whickham survey. Clin Endocrinol 1995; 43:55-68
  533. Helfand M. Screening for subclinical thyroid dysfunction in nonpregnant adults: a summary of the evidence for the U.S. Preventive Services Task Force. Ann Intern Med 2004; 140:128-141
  534. Rugge B, Balshem H, Sehgal R, Relevo R, Gorman P, Helfand M. AHRQ Comparative Effectiveness Reviews. Screening and Treatment of Subclinical Hypothyroidism or Hyperthyroidism. Rockville (MD): Agency for Healthcare Research and Quality (US); 2011.
  535. Asvold BO, Vatten LJ, Midthjell K, Bjoro T. Serum TSH within the reference range as a predictor of future hypothyroidism and hyperthyroidism: 11-year follow-up of the HUNT Study in Norway. J Clin Endocrinol Metab 2012; 97:93-99
  536. Asvold BO, Bjoro T, Vatten LJ. Association of thyroid function with estimated glomerular filtration rate in a population-based study: the HUNT study. Eur J Endocrinol 2011; 164:101-105
  537. Ittermann T, Thamm M, Wallaschofski H, Rettig R, Volzke H. Serum thyroid-stimulating hormone levels are associated with blood pressure in children and adolescents. J Clin Endocrinol Metab 2012; 97:828-834
  538. Cappola AR, Ladenson PW. Hypothyroidism and atherosclerosis. J Clin Endocrinol Metab 2003; 88:2438-2444
  539. Iqbal A, Figenschau Y, Jorde R. Blood pressure in relation to serum thyrotropin: The Tromso study. J Hum Hypertens 2006; 20:932-936
  540. Rodondi N, den Elzen WP, Bauer DC, Cappola AR, Razvi S, Walsh JP, Asvold BO, Iervasi G, Imaizumi M, Collet TH, Bremner A, Maisonneuve P, Sgarbi JA, Khaw KT, Vanderpump MP, Newman AB, Cornuz J, Franklyn JA, Westendorp RG, Vittinghoff E, Gussekloo J. Subclinical hypothyroidism and the risk of coronary heart disease and mortality. Jama 2010; 304:1365-1374
  541. Daswani R, Jayaprakash B, Shetty R, Rau NR. Association of Thyroid Function with Severity of Coronary Artery Disease in Euthyroid Patients. Journal of clinical and diagnostic research : JCDR 2015; 9:Oc10-13
  542. Andersen MN, Olsen AS, Madsen JC, Kristensen SL, Faber J, Torp-Pedersen C, Gislason GH, Selmer C. Long-Term Outcome in Levothyroxine Treated Patients With Subclinical Hypothyroidism and Concomitant Heart Disease. J Clin Endocrinol Metab 2016; 101:4170-4177
  543. Ittermann T, Lorbeer R, Dorr M, Schneider T, Quadrat A, Hesselbarth L, Wenzel M, Lehmphul I, Kohrle J, Mensel B, Volzke H. High levels of thyroid-stimulating hormone are associated with aortic wall thickness in the general population. European radiology 2016; 26:4490-4496
  544. Iqbal A, Jorde R, Figenschau Y. Serum lipid levels in relation to serum thyroid-stimulating hormone and the effect of thyroxine treatment on serum lipid levels in subjects with subclinical hypothyroidism: the Tromso Study. J Intern Med 2006; 260:53-61
  545. Asvold BO, Bjoro T, Vatten LJ. Associations of TSH levels within the reference range with future blood pressure and lipid concentrations: 11-year follow-up of the HUNT study. Eur J Endocrinol 2013; 169:73-82
  546. Witte T, Ittermann T, Thamm M, Riblet NB, Volzke H. Association between serum thyroid-stimulating hormone levels and serum lipids in children and adolescents: a population-based study of german youth. J Clin Endocrinol Metab 2015; 100:2090-2097
  547. Javed Z, Sathyapalan T. Levothyroxine treatment of mild subclinical hypothyroidism: a review of potential risks and benefits. Therapeutic advances in endocrinology and metabolism 2016; 7:12-23
  548. Ajmani SN, Aggarwal D, Bhatia P, Sharma M, Sarabhai V, Paul M. Prevalence of overt and subclinical thyroid dysfunction among pregnant women and its effect on maternal and fetal outcome. Journal of Obstetrics and Gynaecology of India 2014; 64:105-110
  549. Krassas G, Karras SN, Pontikides N. Thyroid diseases during pregnancy: a number of important issues. Hormones (Athens) 2015; 14:59-69
  550. Nazarpour S, Ramezani Tehrani F, Simbar M, Azizi F. Thyroid dysfunction and pregnancy outcomes. Iranian journal of reproductive medicine 2015; 13:387-396
  551. Casey BM, Dashe JS, Wells CE, McIntire DD, Leveno KJ, Cunningham FG. Subclinical hyperthyroidism and pregnancy outcomes. Obstet Gynecol 2006; 107:337-341
  552. Casey BM, Dashe JS, Wells CE, McIntire DD, Byrd W, Leveno KJ, Cunningham FG. Subclinical hypothyroidism and pregnancy outcomes. Obstet Gynecol 2005; 105:239-245
  553. Shan ZY, Chen YY, Teng WP, Yu XH, Li CY, Zhou WW, Gao B, Zhou JR, Ding B, Ma Y, Wu Y, Liu Q, Xu H, Liu W, Li J, Wang WW, Li YB, Fan CL, Wang H, Guo R, Zhang HM. A study for maternal thyroid hormone deficiency during the first half of pregnancy in China. Eur J Clin Invest 2009; 39:37-42
  554. Krassas GE, Poppe K, Glinoer D. Thyroid function and human reproductive health. Endocr Rev 2010; 31:702-755
  555. Lazarus JH. Thyroid function in pregnancy. British medical bulletin 2011; 97:137-148
  556. Brabant G, Peeters RP, Chan SY, Bernal J, Bouchard P, Salvatore D, Boelaert K, Laurberg P. Management of subclinical hypothyroidism in pregnancy: are we too simplistic? Eur J Endocrinol 2015; 173:P1-p11
  557. Negro R, Schwartz A, Gismondi R, Tinelli A, Mangieri T, Stagnaro-Green A. Increased pregnancy loss rate in thyroid antibody negative women with TSH levels between 2.5 and 5.0 in the first trimester of pregnancy. J Clin Endocrinol Metab 2010; 95:E44-48
  558. Liu H, Shan Z, Li C, Mao J, Xie X, Wang W, Fan C, Wang H, Zhang H, Han C, Wang X, Liu X, Fan Y, Bao S, Teng W. Maternal subclinical hypothyroidism, thyroid autoimmunity, and the risk of miscarriage: a prospective cohort study. Thyroid 2014; 24:1642-1649
  559. Maraka S, Ospina NM, O'Keeffe DT, Espinosa De Ycaza AE, Gionfriddo MR, Erwin PJ, Coddington CC, 3rd, Stan MN, Murad MH, Montori VM. Subclinical Hypothyroidism in Pregnancy: A Systematic Review and Meta-Analysis. Thyroid 2016; 26:580-590
  560. Kaprara A, Krassas GE. Thyroid autoimmunity and miscarriage. Hormones (Athens) 2008; 7:294-302
  561. Wang S, Teng WP, Li JX, Wang WW, Shan ZY. Effects of maternal subclinical hypothyroidism on obstetrical outcomes during early pregnancy. J Endocrinol Invest 2012; 35:322-325
  562. Negro R, Schwartz A, Stagnaro-Green A. Impact of Levothyroxine in Miscarriage and Preterm Delivery Rates in First Trimester Thyroid Antibody-Positive Women with TSH<2.5mIU/L. J Clin Endocrinol Metab 2016:jc20161803
  563. Wilson KL, Casey BM, McIntire DD, Halvorson LM, Cunningham FG. Subclinical thyroid disease and the incidence of hypertension in pregnancy. Obstet Gynecol 2012; 119:315-320
  564. Negro R, Formoso G, Mangieri T, Pezzarossa A, Dazzi D, Hassan H. Levothyroxine treatment in euthyroid pregnant women with autoimmune thyroid disease: effects on obstetrical complications. J Clin Endocrinol Metab 2006; 91:2587-2591
  565. Chen X, Jin B, Xia J, Tao X, Huang X, Sun L, Yuan Q. Effects of Thyroid Peroxidase Antibody on Maternal and Neonatal Outcomes in Pregnant Women in an Iodine-Sufficient Area in China. International journal of endocrinology 2016; 2016:6461380
  566. Mannisto T, Vaarasmaki M, Pouta A, Hartikainen AL, Ruokonen A, Surcel HM, Bloigu A, Jarvelin MR, Suvanto-Luukkonen E. Perinatal outcome of children born to mothers with thyroid dysfunction or antibodies: a prospective population-based cohort study. J Clin Endocrinol Metab 2009; 94:772-779
  567. Saki F, Dabbaghmanesh MH, Ghaemi SZ, Forouhari S, Ranjbar Omrani G, Bakhshayeshkaram M. Thyroid function in pregnancy and its influences on maternal and fetal outcomes. International journal of endocrinology and metabolism 2014; 12:e19378
  568. Chen LM, Zhang Q, Si GX, Chen QS, Ye EL, Yu LC, Peng MM, Yang H, Du WJ, Zhang C, Lu XM. Associations between thyroid autoantibody status and abnormal pregnancy outcomes in euthyroid women. Endocrine 2015; 48:924-928
  569. Haddow JE, Palomaki GE, Allan WC, Williams JR, Knight GJ, Gagnon J, O'Heir CE, Mitchell ML, Hermos RJ, Waisbren SE, Faix JD, Klein RZ. Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child. N Engl J Med 1999; 341:549-555
  570. Li Y, Shan Z, Teng W, Yu X, Li Y, Fan C, Teng X, Guo R, Wang H, Li J, Chen Y, Wang W, Chawinga M, Zhang L, Yang L, Zhao Y, Hua T. Abnormalities of maternal thyroid function during pregnancy affect neuropsychological development of their children at 25-30 months. Clin Endocrinol (Oxf) 2010; 72:825-829
  571. Nazarpour S, Ramezani Tehrani F, Simbar M, Tohidi M, Alavi Majd H, Azizi F. Effects of levothyroxine treatment on pregnancy outcomes in pregnant women with autoimmune thyroid disease. Eur J Endocrinol 2016;
  572. Smallridge RC, Ladenson PW. Hypothyroidism in pregnancy: consequences to neonatal health. J Clin Endocrinol Metab 2001; 86:2349-2353
  573. Korevaar TI, Steegers EA, de Rijke YB, Visser WE, Jaddoe VW, Visser TJ, Medici M, Peeters RP. Placental Angiogenic Factors Are Associated With Maternal Thyroid Function and Modify hCG-Mediated FT4 Stimulation. J Clin Endocrinol Metab 2015; 100:E1328-1334
  574. Korevaar TI, Steegers EA, Pop VJ, Broeren MA, Chaker L, de Rijke YB, Jaddoe VW, Medici M, Visser TJ, Tiemeier H, Peeters RP. Thyroid autoimmunity impairs the thyroidal response to hCG: two population-based prospective cohort studies. J Clin Endocrinol Metab 2016:jc20162942
  575. Pekonen F, Alfthan H, Stenman UH, Ylikorkala O. Human chorionic gonadotropin (hCG) and thyroid function in early human pregnancy: circadian variation and evidence for intrinsic thyrotropic activity of hCG. J Clin Endocrinol Metab 1988; 66:853-856
  576. Goodwin TM, Montoro M, Mestman JH, Pekary AE, Hershman JM. The role of chorionic gonadotropin in transient hyperthyroidism of hyperemesis gravidarum. J Clin Endocrinol Metab 1992; 75:1333-1337
  577. Grun JP, Meuris S, De Nayer P, Glinoer D. The thyrotrophic role of human chorionic gonadotrophin (hCG) in the early stages of twin (versus single) pregnancies. Clin Endocrinol (Oxf) 1997; 46:719-725
  578. Li C, Shan Z, Mao J, Wang W, Xie X, Zhou W, Li C, Xu B, Bi L, Meng T, Du J, Zhang S, Gao Z, Zhang X, Yang L, Fan C, Teng W. Assessment of thyroid function during first-trimester pregnancy: what is the rational upper limit of serum TSH during the first trimester in Chinese pregnant women? J Clin Endocrinol Metab 2014; 99:73-79
  579. Medici M, Korevaar TI, Schalekamp-Timmermans S, Gaillard R, de Rijke YB, Visser WE, Visser W, de Muinck Keizer-Schrama SM, Hofman A, Hooijkaas H, Bongers-Schokking JJ, Tiemeier H, Jaddoe VW, Visser TJ, Peeters RP, Steegers EA. Maternal early-pregnancy thyroid function is associated with subsequent hypertensive disorders of pregnancy: the generation R study. J Clin Endocrinol Metab 2014; 99:E2591-2598
  580. Rajput R, Singh B, Goel V, Verma A, Seth S, Nanda S. Trimester-specific reference interval for thyroid hormones during pregnancy at a Tertiary Care Hospital in Haryana, India. Indian journal of endocrinology and metabolism 2016; 20:810-815
  581. Lambert-Messerlian G, McClain M, Haddow JE, Palomaki GE, Canick JA, Cleary-Goldman J, Malone FD, Porter TF, Nyberg DA, Bernstein P, D'Alton ME. First- and second-trimester thyroid hormone reference data in pregnant women: a FaSTER (First- and Second-Trimester Evaluation of Risk for aneuploidy) Research Consortium study. Am J Obstet Gyneol 2008; 199:62: e61-66
  582. Tozzoli R, D'Aurizio F, Ferrari A, Castello R, Metus P, Caruso B, Perosa AR, Sirianni F, Stenner E, Steffan A, Villalta D. The upper reference limit for thyroid peroxidase autoantibodies is method-dependent: A collaborative study with biomedical industries. Clin Chim Acta 2016; 452:61-65
  583. Weeke J, Gundersen HJ. Circadian and 30 minute variations in serum TSH and thyroid hormones in normal subjects. Acta Endocrinol 1978; 89:659-672
  584. Evans PJ, Weeks I, Jones MK, Woodhead JS, Scanlon MF. The circadian variation of thyrotrophin in patients with primary thyroidal disease. CLIN Endocrinol (Oxf) 1986; 24:343-348
  585. Brabant G, Prank K, Hoang-Vu C, von zur Muhlen A. Hypothalamic regulation of pulsatile thyrotropin secretion. J Clin Endocrinol Metab 1991; 72:145-150
  586. Roelfsema F, Pereira AM, Veldhuis JD, Adriaanse R, Endert E, Fliers E, Romijn JA. Thyrotropin secretion profiles are not different in men and women. J Clin Endocrinol Metab 2009; 94:3964-3967
  587. Ladenson PW, Singer PA, Ain KB, Bagchi N, Bigos ST, Levy EG, Smith SA, Daniels GH, Cohen HD. American Thyroid Association guidelines for detection of thyroid dysfunction. Arch Intern Med 2000; 160:1573-1575
  588. Singer PA, Cooper DS, Levy EG, Ladenson PW, Braverman LE, Daniels G, Greenspan FS, McDougall IR, Nikolai TF. Treatment guidelines for patients with hyperthyroidism and hypothyroidism. Standards of Care Committee, American Thyroid Association. JAMA 1995; 273:808-812
  589. Carr D, McLeod DT, Parry G, Thornes HM. Fine adjustment of thyroxine replacement dosage: comparison of the thyrotrophin releasing hormone test using a sensitive thyrotrophin assay with measurement of free thyroid hormones and clinical assessment. Clin Endocrinol 1988; 28:325-333
  590. Walsh JP, Ward LC, Burke V, Bhagat CI, Shiels L, Henley D, Gillett MJ, Gilbert R, Tanner M, Stuckey BG. Small changes in thyroxine dosage do not produce measurable changes in hypothyroid symptoms, well-being, or quality of life: results of a double-blind, randomized clinical trial. J Clin Endocrinol Metab 2006; 91:2624-2630
  591. Jonklaas J, Sarlis NJ, Litofsky D, Ain KB, Bigos ST, Brierley JD, Cooper DS, Haugen BR, Ladenson PW, Magner J, Robbins J, Ross DS, Skarulis M, Maxon HR, Sherman SI. Outcomes of patients with differentiated thyroid carcinoma following initial therapy. Thyroid 2006;
  592. Pujol P, Daures JP, Nsakala N, Baldet L, Bringer J, Jaffiol C. Degree of thyrotropin suppression as a prognostic determinant in differentiated thyroid cancer. J Clin Endocrinol Metab 1996; 81:4318-4323
  593. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL, Mandel SJ, Mazzaferri EL, McIver B, Pacini F, Schlumberger M, Sherman SI, Steward DL, Tuttle RM. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 2009; 19:1167-1124
  594. Wartofsky L, Burman KD. Alterations in thyroid function in patients with systemic illness: the "euthyroid sick syndrome". Endocrinol Rev 1982; 3:164-217
  595. Mebis L, van den Berghe G. The hypothalamus-pituitary-thyroid axis in critical illness. Neth J Med 2009; 67:332-340
  596. Lamb EJ, Martin J. Thyroid function tests: often justified in the acutely ill. Ann Clin Biochem 2000; 37:158-164
  597. Mebis L, Paletta D, Debaveye Y, Ellger B, Langouche L, D'Hoore A, Darras VM, Visser TJ, Van den Berghe G. Expression of thyroid hormone transporters during critical illness. Eur J Endocrinol 2009; 161:243-250
  598. Bunevicius R, Steibliene V, Prange AJ, Jr. Thyroid axis function after in-patient treatment of acute psychosis with antipsychotics: a naturalistic study. BMC psychiatry 2014; 14:279
  599. Verhoye E, Van den Bruel A, Delanghe JR, Debruyne E, Langlois MR. Spuriously high thyrotropin values due to anti-thyrotropin antibodies in adult patients. Clin Chem Lab Med 2009; 47:604-606
  600. Uy HL, Reasner CA, Samuels MH. Pattern of recovery of the hypothalamic-pituitary-thyroid axis following radioactive iodine therapy in patients with Graves' disease. Am J Med 1995; 99:173-179
  601. Kempers MJ, van der Sluijs Veer L, Nijhuis-van der Sanden RW, Lanting CI, Kooistra L, Wiedijk BM, Last BF, de Vijlder JJ, Grootenhuis MA, Vulsma T. Neonatal screening for congenital hypothyroidism in the Netherlands: cognitive and motor outcome at 10 years of age. J Clin Endocrinol Metab 2007; 92:919-924
  602. Schaaf L, Theodoropoulou M, Gregori A, Leiprecht A, Trojan J, Klostermeier J, Stalla GK. Thyrotropin-releasing hormone time-dependently influences thyrotropin microheterogeneity--an in vivo study in euthyroidism. J Endocrinol 2000; 166:137-143
  603. Tjornstrand A, Gunnarsson K, Evert M, Holmberg E, Ragnarsson O, Rosen T, Filipsson Nystrom H. The incidence rate of pituitary adenomas in western Sweden for the period 2001-2011. Eur J Endocrinol 2014; 171:519-526
  604. Beck-Peccoz P, Lania A, Beckers A, Chatterjee K, Wemeau J-L. 2013 European Thyroid Association Guidelines for the Diagnosis and treatment of TSH-secreting pituitary tumors. in press 2013;
  605. Refetoff S, Bassett JH, Beck-Peccoz P, Bernal J, Brent G, Chatterjee K, De Groot LJ, Dumitrescu AM, Jameson JL, Kopp PA, Murata Y, Persani L, Samarut J, Weiss RE, Williams GR, Yen PM. Classification and proposed nomenclature for inherited defects of thyroid hormone action, cell transport, and metabolism. Thyroid 2014; 24:407-409
  606. Refetoff S, Weiss RE, Usala SJ. The syndromes of resistance to thyroid hormone. Endocr Rev 1993; 14:348-399
  607. Refetoff S, DeGroot LJ, Benard B, DeWind LT. Studies of a sibship with apparent hereditary resistance to the intracellular action of thyroid hormone. Metabolism 1972; 21:723-756
  608. Dumitrescu AM, Refetoff S. The syndromes of reduced sensitivity to thyroid hormone. Biochim Biophys Acta 2013; 1830:3987-4003
  609. Laurberg P. Persistent problems with the specificity of immunometric TSH assays. Thyroid 1993; 4:279-283
  610. Sapin R, d'Herbomez M, Schlienger JL, Wemeau JL. Anti-thyrotropin antibody interference in thyrotropin assays. Clin Chem 1998; 44:2557-2559
  611. Hattori N, Ishihara T, Shimatsu A. Variability in the detection of macro TSH in different immunoassay systems. Eur J Endocrinol 2016; 174:9-15
  612. Drees JC1, Stone JA, Reamer CR, Arboleda VE, Huang K, Hrynkow J, Greene DN, Petrie MS, Hoke C, Lorey TS, Dlott RS. Falsely Undetectable TSH in a Cohort of South Asian Euthyroid Patients. J Clin Endocrinol Metab 2014; 99:1171-1179
  613. Pappa T, Johannesen J, Scherberg N, Torrent M, Dumitrescu A, Refetoff S. A TSHbeta Variant with Impaired Immunoreactivity but Intact Biological Activity and Its Clinical Implications. Thyroid 2015; 25:869-876
  614. Medeiros-Neto G, Herodotou DT, Rajan S, Kommareddi S, de Lacerda L, Sandrini R, Boguszewski MC, Hollenberg AN, Radovick S, Wondisford FE. A circulating, biologically inactive thyrotropin caused by a mutation in the beta subunit gene. J Clin Invest 1996; 97:1250-1256
  615. Sertedaki A, Papadimitriou A, Voutetakis A, Dracopoulou M, Maniati-Christidi M, Dacou-Voutetakis C. Low TSH congenital hypothyroidism: identification of a novel mutation of the TSH beta-subunit gene in one sporadic case (C85R) and of mutation Q49stop in two siblings with congenital hypothyroidism. Pediatr Res 2002; 52:935-941
  616. Grunert SC, Schmidts M, Pohlenz J, Kopp MV, Uhl M, Schwab KO. Congenital Central Hypothyroidism due to a Homozygous Mutation in the TSHbeta Subunit Gene. Case reports in pediatrics 2011; 2011:369871
  617. Saravanan P, Dayan CM. Thyroid autoantibodies. Endocrinol Metab Clin N Am 2001; 30:315-337
  618. Schardt CW, McLaughlan SM, Matheson J, Smith BR. An enzyme-linked immunoassay for thyroid microsomal antibodies. J Immunol Methods 1982; 55:155-168
  619. Mariotti S, Caturegli P, Piccolo P, Barbesino G, Pinchera A. Antithyroid peroxidase autoantibodies in thyroid diseases. J Clin Endocrinol Metab 1990; 71:661-669
  620. Tozzoli R, Bagnasco M, Giavarina D, Bizzaro N. TSH receptor autoantibody immunoassay in patients with Graves' disease: improvement of diagnostic accuracy over different generations of methods. Systematic review and meta-analysis. Autoimmun Rev 2012; 12:107-113
  621. Kahaly GJ. Bioassays for TSH Receptor Antibodies: Quo Vadis? Eur Thyroid J 2015; 4:3-5
  622. Massart C, Sapin R, Gibassier J, Agin A, d'Herbomez M. Intermethod variability in TSH-receptor antibody measurement: implication for the diagnosis of Graves disease and for the follow-up of Graves ophthalmopathy. Clin Chem 2009; 55:183-186
  623. Tozzoli R, Villalta D, Bizzaro N. Challenges in the Standardization of Autoantibody Testing: a Comprehensive Review. Clinical reviews in allergy & immunology 2016;
  624. Rapoport B, Chazenbalk GD, Jaume JC, McLachlan SM. The thyrotropin (TSH) receptor: interaction with TSH and autoantibodies. Endoc Rev 1998; 19:673-616
  625. Davies T, Marians R, Latif R. The TSH receptor reveals itself. J Clin Invest 2002; 110:161-164
  626. Smith BR, McLachlan SM, Furmaniak J. Autoantibodies to the thyrotropin receptor. Endocrinol Rev 1988; 9:106-121
  627. Adams DD. Long-acting thyroid stimulator: how receptor autoimmunity was discovered. Autoimmunity 1988; 1:3-9
  628. McKenzie MJ, Zakarija M. Antibodies in autoimmune thyroid disease. 6th. Edition ed. Philadelphia: J B Lippincott.
  629. Ando T, Latif R, Davies TF. Thyrotropin receptor antibodies: new insights into their actions and clinical relevance. Best Pract Res Clin Endocrinol Metab 2005; 19:33-52
  630. Noh JY, Hamada N, Inoue Y, Abe Y, Ito K, Ito K. Thyroid-Stimulating Antibody is Related to Graves' Ophthalmopathy, But Thyrotropin-Binding Inhibitor Immunoglobulin is Related to Hyperthyroidism in Patients with Graves' Disease. Thyroid 2000; 10:809-813
  631. Mizutori Y, Chen CR, Latrofa F, McLachlan SM, Rapoport B. Evidence that shed thyrotropin receptor A subunits drive affinity maturation of autoantibodies causing Graves' disease. J Clin Endocrinol Metab 2009; 94:927-935
  632. Woo YJ, Jang SY, Lim TH, Yoon JS. Clinical Association of Thyroid Stimulating Hormone Receptor Antibody Levels with Disease Severity in the Chronic Inactive Stage of Graves' Orbitopathy. Korean journal of ophthalmology : KJO 2015; 29:213-219
  633. Gupta MK. Thyrotropin-receptor antibodies in thyroid diseases: advances in detection techniques and clinical applications. Clin Chim Acta 2000; 293:1-29
  634. Kahaly GJ, Diana T, Glang J, Kanitz M, Pitz S, Konig J. Thyroid Stimulating Antibodies Are Highly Prevalent in Hashimoto's Thyroiditis and Associated Orbitopathy. J Clin Endocrinol Metab 2016; 101:1998-2004
  635. Morgenthaler NG, Ho SC, Minich WB. Stimulating and blocking thyroid-stimulating hormone (TSH) receptor autoantibodies from patients with Graves' disease and autoimmune hypothyroidism have very similar concentration, TSH receptor affinity, and binding sites. J Clin Endocrinol Metab 2007; 92:1058-1065
  636. Yoshida K, Aizawa Y, Kaise N, Fukazawa H, Kiso Y, Sayama N, Mori K, Hori H, Abe K. Relationship between thyroid-stimulating antibodies and thyrotropin-binding inhibitory immunoglobulins years after administration of radioiodine for Graves' disease: retrospective clinical survey. J Endocrinol Invest 1996; 19:682-686
  637. Quadbeck B, Hoermann R, Hahn S, Roggenbuck U, Mann K, Janssen OE. Binding, stimulating and blocking TSH receptor antibodies to the thyrotropin receptor as predictors of relapse of Graves' disease after withdrawal of antithyroid treatment. Horm Metab Res 2005; 37:745-750
  638. McLachlan SM, Rapoport B. Thyrotropin-blocking autoantibodies and thyroid-stimulating autoantibodies: potential mechanisms involved in the pendulum swinging from hypothyroidism to hyperthyroidism or vice versa. Thyroid 2013; 23:14-24
  639. Seetharamaiah GS, Kurosky A, Desai RK, Dallas JS, Prabhakar VS. A recombinant extracellular domain of the thyrotropin (TSH) receptor bindsTSH in the absence of membranes. Endocrinol 1994; 134:549-554
  640. Sugawa H, Ueda Y, M. U, al e. Immunization with the "immunogenic peptide" of TSH receptor induces oligoclonal antibodies with various biological activities. Peptides 1998; 19:1301-1307
  641. Davies TF, Ando T, Lin RY, Tomer Y, Latif R. Thyrotropin receptor-associated diseases: from adenomata to Graves disease. J Clin Invest 2005; 115:1972-1983
  642. Takasu N, oshiro C, Akamine H, Komiya I, Nagata A, Sato Y, Yoshimura H, Ito K. Thyroid-stimulating antibody and TSH-binding inhibitor immunoglobulin in 277 Graves' patients and in 686 normal subjects. J Endocrinol Invest 1997; 20:452-461
  643. Kung AW, Jones BM. A change from stimulatory to blocking antibody activity in Graves' disease during pregnancy. J Clin Endocrinol Metab 1998; 83:514-518
  644. Michalek K, Morshed SA, Latif R, Davies TF. TSH receptor autoantibodies. Autoimmun Rev 2009; 9:113-116
  645. Kamijo K. TSH-receptor antibodies determined by the first, second and third generation assays and thyroid-stimulating antibody in pregnant patients with Graves' disease. Endocr J 2007; 54:619-624
  646. Tozzoli R, D'Aurizio F, Villalta D, Giovanella L. Evaluation of the first fully automated immunoassay method for the measurement of stimulating TSH receptor autoantibodies in Graves' disease. Clin Chem Lab Med 2017; 55:58-64
  647. Kasagi K, Konishi J, Iida Y, Ikekubo K, Mori T, Kuma K, Torizuka K. A new in vitro assay for human thyroid stimulator using cultured thyroid cells: effect of sodium chloride on adenosine 3',5'-monophosphate increase. 54 1982:108-114
  648. Shewring G, Rees-Smith B. An improved radioreceptor assay for TSH receptor antibodies. Clin Endocrinol 1982; 17:409-417
  649. Tokuda Y, Kasagi K, LidaY, Hatabu H, Hidaka A, Misaki T, Konishi J. Sensitive, practical bioassay of thyrotropin, with use of FRTL-5 thyroid cells and magnetizable solid-phase-bound antibodies. Clin Chem 1988; 34:2360-2364
  650. McKenzie JM, Zakarija M. LATS in Graves' disease. Recent Prog Horm Res 1976; 33:29-57
  651. Kasagi K, Konishi J, Iida Y, Arai K, Endo K, Torizuka K. A sensitive and practical assay for thyroid stimulating antibodies using FRTL-5 thyroid cells. Acta Endocrinol 1987; 115:30-36
  652. Morris JC, Hay ID, Nelson RE, Jiang N-S. Clinical utility of thyrotropin-receptor antibody assays: comparison of radioreceptor and bioassay methods. Mayo Clin Proc 1988; 63:707-717
  653. Michelangeli VP, Munro DS, Poon CW, Frauman AG, Colman PG. Measurement of thyroid stimulating immunoglobulins in a new cell line transfected with a functional human TSH receptor (JPO9 cells), compared with an assay using FRTL-5 cells. Clin Endocrinol 1994; 40:645-652
  654. Minich WB, Behr M, Loos U. Expression of a functional tagged human thyrotropin receptor in HeLa cells using recombinant vaccinia virus. Exp Clin Endocrinol Diab 1997; 105:282-290
  655. Morgenthaler NG, Pampel I, Aust G, SeisslerJ, Scherbaum WA. Application of a bioassay with CHO cells for the routine detection of stimulating and blocking autoantibodies to the TSH-receptor. Horm Metab Res 1998; 30:162-168
  656. Kamijo K, Murayama H, Uzu T, Togashi K, Olivo PD, Kahaly GJ. Similar clinical performance of a novel chimeric thyroid-stimulating hormone receptor bioassay and an automated thyroid-stimulating hormone receptor binding assay in Graves' disease. Thyroid 2011; 21:1295-1299
  657. Atger M, Misrahi M, Young J, Jolivet A, Orgiazzi J, Schaison G, Milgrom E. Autoantibodies interacting with purified native thyrotropin receptor. Eur J Biochem 1999; 265:1022-1031
  658. Iida Y, Konishi J, Kasagi K, Kuma K, Torizuka K. Detection of TSH-binding inhibitor immunoglobulins by using the triton-solubilized receptor from human thyroid membranes. Endocrinol Jpn 1982; 29:227-231
  659. Costagliola S, Morganthaler NG, Hoermann R, Badenhoop K, Struck J, Freitag D, Poertl s, Weglohner W, Hollidt JM, Quadbeck B, Dumont JE, Schumm-Draeger PM, Bergmann A, Mann K, Vassart G, Usadel KH. Second generation assay for thyrotropin receptor antibodies has superior diagnostic sensitivity for Graves' disease. J Clin Endocrinol Metab 1999; 84:90-97
  660. Schott M, Feldkamp J, Bathan C, Fritzen R, Scherbaum WA, Seissler J. Detecting TSH-Receptor antibodies with the recombinant TBII assay: technical and clinical evaluation. Horm Metab Research 2000; 32:429-435
  661. Giovanella L, Ceriani L, Garancini S. Clinical applications of the 2nd. generation assay for anti-TSH receptor antibodies in Graves' disease. Evaluation in patients with negative 1st. generation test. Clin Chem Lab med 2001; 39:25-28
  662. Smith BR, Bolton J, Young S, Collyer A, Weeden A, Bradbury J, Weightman D, Perros P, Sanders J, Furmaniak J. A new assay for thyrotropin receptor autoantibodies. Thyroid 2004; 14:830-835
  663. Massart C, Gibassier J, d'Herbomez M. Clinical value of M22-based assays for TSH-receptor antibody (TRAb) in the follow-up of antithyroid drug treated Graves' disease: comparison with the second generation human TRAb assay. Clin Chim Acta 2009; 407:62-66
  664. Zöphel K, Roggenbuck D, Wunderlich G, Schott M. Continuously increasing sensitivity over three generations of TSH receptor autoantibody assays. Horm Metab Res 2010; 42:900-902
  665. Costagliola S, Swillens S, Niccoli P, Dumont JE, Vassart G, Ludgate M. Binding assay for thyrotropin receptor autoantibodies using the recombinant receptor protein. J Clin Endocrinol Metab 1992; 75:1540-1544
  666. Morgenthaler NG, Hodak K, Seissler J, Steinbrenner H, Pampel I, Gupta M, McGregor AM, Scherbaum WA, Banga JP. Direct binding of thyrotropin receptor autoantibody to in vitro translated thyrotropin receptor: a comparison to radioreceptor assay and thyroid stimulating bioassay. Thyroid 1999; 9:466-475
  667. Feldt-Rasmussen. Meta-analysis evaluation of the impact of thyrotropin receptor antibodies on long-term remission after medical therapy of Graves' disease. J Clin Endocrinol Metab 1994; 78:98-102
  668. Cho BY, Shong MH, Yi KH, Lee HK, Koh CS, Min HK. Evaluation of serum basal thyrotropin levels and thyrotropin receptor antibody levels and thyrotropin receptor antibody activities as prognostic markers for discontinuation of antithyroid drug treatment in patients with Graves' disease. Clin Endocrinol 1992; 36:585-590
  669. Shibayama K, Ohyama Y, Yokota Y, Ohtsu S, Takubo N, Matsuura N. Assays for thyroid-stimulating antibodies and thyrotropin-binding inhibitory immunoglobulins in children with Graves' disease. Endoc J 2005; 52:505-510
  670. Pedersen IB, Knudsen N, Perrild H, Ovesen L, Laurberg P. TSH-receptor antibody measurement for differentiation of hyperthyroidism into Graves' disease and multinodular toxic goitre: a comparison of two competitive binding assays. Clin Endocrinol 2001; 55:381-390
  671. Tan K, Loh TP, Sethi S. Lack of standardized description of TRAb assays. Endocrine 2012;
  672. Barbesino G, Tomer Y. Clinical review: Clinical utility of TSH receptor antibodies. J Clin Endocrinol Metab 2013; 98:2247-2255
  673. Evans M, Sanders J, Tagami T, Sanders P, Young S, Roberts E, Wilmot J, Hu X, Kabelis K, Clark J, Holl S, Richards T, Collyer A, Furmaniak J, Smith BR. Monoclonal autoantibodies to the TSH receptor, one with stimulating activity and one with blocking activity, obtained from the same blood sample. CLIN Endocrinol (Oxf) 2010; 73:404-412
  674. Bartalena L, Marcocci C, Bogazzi F, al e. Relation between therapy for hyperthyroidism and the course of Graves' disease. N Engl J Med 1998; 338:73-78
  675. Eckstein AK, Plicht M, Lax H, Neuhäuser M, Mann K, Lederbogen S, Heckmann C, Esser J, Morgenthaler NG. Thyrotropin receptor autoantibodies are independent risk factors for Graves' ophthalmopathy and help to predict severity and outcome of the disease. J Clin Endocrinol Metab 2006; 91:3464-3470
  676. Bech K. Immunological aspects of Graves' disease and importance of thyroid stimulating immunoglobulins. Acta Endocrinol (Copenh) Suppl 1983; 103:5-38
  677. Feldt-Rasmussen U. Serum thyroglobulin and thyroglobulin autoantibodies in thyroid disease. Allergy 1983; 38:369-387
  678. Nygaard B, Metcalfe RA, Phipps J, Weetman AP, Hegedus L. Graves' disease and thyroid-associated opthalmopathy triggered by 131I treatment of non-toxic goitre. J Endocrinol Invest 1999; 22:481-485
  679. Gerding MN, van der Meer Jolanda WC, Broenink M, Bakker O, WM W, Prummel MF. Association of thyrotropin receptor antibodies with the clinical features of Graves' opthalmopathy. Clin Endocrinol 2000; 52:267-271
  680. Takamura Y, Nakano K, Uruno T, Ito Y, Miya A, Kobayashi K, Yokozawa T, Matsuzuka F, Kuma K, Miyauchi A. Changes in serum TSH receptor antibody (TRAb) values in patients with Graves' disease after total or subtotal thyroidectomy. Endoc J 2003; 50:595-601
  681. Zimmermann-Belsing T, Nygaard B, Rasmussen AK, Feldt-Rasmussen U. Use of the 2nd generation TRAK human assay did not improve prediction of relapse after antithyroid medical therapy of Graves' disease. Eur J Endocrinol 2002; 146:173-177
  682. Schott M, Morgenthaler NG, Fritzen R, Feldkamp J, Willenberg HS, Scherbaum WA, Seissler J. Levels of autoantibodies against human TSH receptor predict relapse of hyperthyroidism in Graves' disease. Horm Metab Res 2004; 36:92-96
  683. Carella C, Mazziotti G, Sorvillo F, Piscopo M, Cioffi M, Pilla P, Nersita R, Iorio S, Amato G, Braverman LE, Roti E. Serum thyrotropin receptor antibodies concentrations in patients with Graves' disease before, at the end of methimazole treatment, and after drug withdrawal: evidence that the activity of thyrotropin receptor antibody and/or thyroid response modify during the observation period. Thyroid 2006; 16:295-302
  684. Morris JC. Clinical use of immunological assays of TSH Receptor autoantibodies. Thyroid Today 1998; 21:1-7
  685. Michelangeli V, Poon C, taft J, Newnham H, Topliss D, Colman P. The prognostic value of thyrotropin receptor antibody measurement in the early stages of treatment of Graves' disease with antithyroid drugs. Thyroid 1998; 8:119-124
  686. McKenzie JM, Zakarija M. Fetal and neonatal hyperthyroidism and hypothyroidism due to maternal TSH receptor antibodies [Review]. Thyroid 1992; 2:155-159
  687. Chan GW, Mandel SJ. Therapy insight: management of Graves' disease during pregnancy. Nat Clin Pract Endocrinol Metab 2007; 3:470-478
  688. Nor Azlin MI, Bakin YD, Mustafa N, Wahab NA, Johari MJ, Kamarudin NA, Jamil MA. Thyroid autoantibodies and associated complications during pregnancy. j Obstet Gynaecol 2010; 30:675-678
  689. Hamada N, Momotani N, Ishikawa N, Yoshimura Noh J, Okamoto Y, Konishi T, Ito K, Ito K. Persistent high TRAb values during pregnancy predict increased risk of neonatal hyperthyroidism following radioiodine therapy for refractory hyperthyroidism. Endoc J 2011; 58:55-58
  690. Heithorn R, Hauffa BP, Reinwein D. Thyroid antibodies in children of mothers with autoimmune thyroid disorders. Eur J Pediatr 1999; 158:24-28
  691. Abeillon-du Payrat J, Chikh K, Bossard N, Bretones P, Gaucherand P, Claris O, Charrie A, Raverot V, Orgiazzi J, Borson-Chazot F, Bournaud C. Predictive value of maternal second-generation thyroid-binding inhibitory immunoglobulin assay for neonatal autoimmune hyperthyroidism. Eur J Endocrinol 2014; 171:451-460
  692. McLachlan SM, Rapoport B. Thyroid peroxidase autoantibody epitopes revisited. Clin Endocrinol 2008; 69:526-527
  693. Jaume JC, Costante G, Nishikawa T, Phillips DI, Rapoport B, McLachlan SM. Thyroid peroxidase autoantibody fingerprints in hypothyroid and euthyroid individuals. I. Cross-sectional study in elderly women. J Clin Endocrinol Metab 1995; 80:994-999
  694. Jaume JC, Burek CL, Hoffman WH, Rose NR, McLachlan SM, Rapoport B. Thyroid peroxidase autoantibody epitopic 'fingerprints' in juvenile Hashimoto's thyroiditis: evidence for conservation over time and in families. J Clin Endocrinol Metab 1996; 104:115-123
  695. Czarnocka B, Szabolcs I, Pastuszko D, Feldkamp J, Dohán O, Podoba J, Wenzel B. In old age the majority of thyroid peroxidase autoantibodies are directed to a single TPO domain irrespective of thyroid function and iodine intake. Clin Endocrinol 1998; 48:803-808
  696. Nielsen CH, Brix TH, Gardas A, Banga JP, Hegedüs L. Epitope recognition patterns of thyroid peroxidase autoantibodies in healthy individuals and patients with Hashimoto's thyroiditis. Clin Endocrinol 2008; 69:664-668
  697. Ehlers M, Thiel A, Bernecker C, Porwol D, Papewalis C, Willenberg HS, Schinner S, Hautzel H, Scherbaum WA, Schott M. Evidence of a combined cytotoxic thyroglobulin and thyroperoxidase epitope-specific cellular immunity in Hashimoto's thyroiditis. J Clin Endocrinol Metab 2012; 97:1347-1354
  698. Trotter WR, Belyavin G, Waddams A. Precipitating and complement fixing antibodies in Hashimoto's disease. Proc R Soc Med 1957; 50:961-?
  699. Cayzer I, Chalmers SR, Doniach d, Swana G. An evaluation of two new haemagglutination tests for the rapid diagnosis of autoimmune thyroid disease. J Clin Pathol 1978; 31:1147-1151
  700. Mariotti S, Russova A, Pisani S, Pinchera A. A new solid phase immunoradiometric assay for antithyroid microsomal antibody. J Clin Endocrinol Metab 1983; 56:467-473
  701. Czarnocka B, Ruf J, Ferrand M, Carayon P, Lissitzky S. Purification of the human thyroid peroxidase and its identification as the microsomal antigen involved in autoimmune thyroid diseases. FEBS Lett 1985; 190:147-152
  702. Mariotti S, Anelli S, Ruf J, Bechi R, Czarnocka B, Lombardi A, Carayon P, Pinchera A. Comparison of serum thyroid microsomal and thyroid peroxidase autoantibodies in thyroid diseases. J Clin Endocrinol Metab 1987; 65:987-993
  703. Hoier-Madsen M, Feldt-Rasmussen U, Hegedus L, Perrild H, Hansen HS. Enzyme-linked immunosorbent assay for determination of thyroglobulin autoantibodies. Acta Pathol Microbiol Scand 1984; 92:377-382
  704. Ruf J, Czarnocka B, Ferrand M, Doullais F, Carayon P. Novel routine assay of thyroperoxidase autoantibodies. Clin Chem 1988; 34:2231-2234
  705. Yokoyama N, Taurog A, Klee GG. Thyroid peroxidase and thyroid microsomal autoantibodies. J Clin Endocrinol Metab 1989; 68:766-773
  706. Beever K, Bradbury J, Phillips D, McLachlan SM, Pegg C, Goral A, Overbeck W, Feifel G, BR< S. Highly sensitive assays of autoantibodies to thyroglobulin and to thyroid peroxidase. Clin Chem 1989; 35:1949-1954
  707. Groves CJ, Howells RD, Williams S, Darke C, Parkes AB. Primary standardization for the ELISA of serum thyroperoxidase and thyroglobulin antibodies and their prevalence in a normal Welsh population. J Clin Lab Immunol 1990; 32:147-151
  708. Laurberg P, Pedersen KM, Vittinghus E, Ekelund S. Sensitive enzyme-linked immunosorbent assay for measurement of autoantibodies to human thyroid peroxidase. Scand J Clin Lab Invest 1992; 52:663-669
  709. Finke R, Bogner u, Kotulla P, Schleusener H. Anti-TPO antibody determinations using different methods. Exp Clin Endocrinol 1994; 102:145-150
  710. La'ulu SL, Slev PR, Roberts WL. Performance characteristics of 5 automated thyroglobulin autoantibody and thyroid peroxidase autoantibody assays. Clin Chim Acta 2007; 376:88-95
  711. Feldt-Rasmussen U, Hoin-Madsen M, Beck K, al e. Anti-thyroid peroxidase antibodies in thyroid disorders and non thyroid autoimmune diseases. Autoimmunity 1991; 9:245-251
  712. Kasagi K, Takahashi N, Inoue G, Honda T, Kawachi Y, Izumi Y. Thyroid function in Japanese adults as assessed by a general health checkup system in relation with thyroid-related antibodies and other clinical parameters. Thyroid 2009; 19:937-944
  713. Spencer CA, Takeuchi M, Kazarosyan M, Wang CC, Guttler RB, Singer PA, Fatemi S, LoPresti JS, Nicoloff JT. Serum Thyroglobulin Autoantibodies: Prevalence, influence on serum thyroglobulin measurement and prognostic significance in patients with differentiated thyroid carcinoma. J Clin Endocrinol Metab 1998; 83:1121-1127
  714. Guo J, Jaume JC, Rapoport B, McLachlan SM. Recombinant thyroid peroxidase-specific Fab converted to immunoglobulin G (IgG)molecules: evidence for thyroid cell damage by IgG1, but not IgG4, autoantibodies. J Clin Endocrinol Metab 1997; 82:925-931
  715. Rebuffat SA, Nguyen B, Robert B, Castex F, Peraldi-Roux S. Antithyroperoxidase antibody-dependent cytotoxicity in autoimmune thyroid disease. J Clin Endocrinol Metab 2008; 93:929-934
  716. Chiovato L, Bassi P, Santini F, Mammoli C, Lapi P, Carayon P, Pinchera A. Antibodies producing complement-mediated thyroid cytotoxicity in patients with atrophic or goitrous autoimmune thyroiditis. J Clin Endocrinol Metab 1993; 77:1700-1705
  717. Karanikas G, Schuetz M, Wahl K, Paul M, Kontur S, Pietschmann P, Kletter K, Dudczak R, Willheim M. Relation of anti-TPO autoantibody titre and T-lymphocyte cytokine production patterns in Hashimoto's thyroiditis. Clin Endocrinol 2005; 63:191-196
  718. Carmel R, Spencer CA. Clinical and subclinical thyroid disorders associated with pernicious anemia. Observations on abnormal thyroid-stimulating hormone levels and on a possible association of blood group O with hyperthyroidism. Arch Intern Med 1982; 142:1465-1469
  719. Vestgaard M, Nielsen LR, Rasmussen AK, Damm P, Mathiesen ER. Thyroid peroxidase antibodies in pregnant women with type 1 diabetes: impact on thyroid function, metabolic control and pregnancy outcome. Acta Obstet Gynecol Scand 2008; 87:1336-1342
  720. Nakamura H, Usa T, Motomura M, Ichikawa T, Nakao K, Kawasaki E, Tanaka M, Ishikawa K, Eguchi K. Prevalence of interrelated autoantibodies in thyroid diseases and autoimmune disorders. J Endocrinol Invest 2008; 31:861-865
  721. Huber G, Staub JJ, Meier C, Mitrache C, Guglielmetti M, Huber P, Braverman LE. Prospective Study of the Spontaneous Course of Subclinical Hypothyroidism: Prognostic Value of Thyrotropin, Thyroid Reserve, and Thyroid Antibodies. J Clin Endocrinol Metab 2002; 87:3221-3226
  722. Mariotti S, Barbesino G, Caturegli P, Atzeni F, Manetti L, Marinò M, Grasso L, Velluzzi F, Loviselli A, Pinchera A. False negative results observed in anti-thyroid peroxidase autoantibody determination by competitive radioimmunoassays using monoclonal antibodies. Eur J Endocrinol 1994; 130:552-558
  723. Schmidt M, Voell M, Rahlff I, Dietlein M, Kobe C, Faust M, Schicha H. Long-term follow-up of antithyroid peroxidase antibodies in patients with chronic autoimmune thyroiditis (Hashimoto's thyroiditis) treated with levothyroxine. Thyroid 2008; 18:755-760
  724. Johnston AM, Eagles JM. Lithium-associated clinical hypothyroidism. Prevalence and risk factors. Br J Psychiatry 1999; 175:336-339
  725. Bell TM, Bansal AS, Shorthouse C, et al. Low titre autoantibodies predict autoimmune disease during interferon alpha treatment of chronic hepatitis C. J Gastroenterol Hepatol 1999; 14:419-422
  726. Daniels GH. Amiodarone-induced thyrotoxicosis. J Clin Endocrinol Metab 2001; 86:3-8
  727. Martino E, Bartalena L, Bogazzi F, Braverman LE. The effects of Amiodarone on the Thyroid. Endoc Rev 2001; 22:240-254
  728. Eskes SA, Wiersinga WM. Amiodarone and thyroid. Best Pract Res Clin Endocrinol Metab 2009; 23:735-751
  729. Bocchetta A, Mossa P, Velluzzi F, Mariotti S, Zompo MD, Loviselli A. Ten-year follow-up of thyroid function in lithium patients. J Clin Psychopharmacol 2001; 21:594-598
  730. Tsang W, Houlden RL. Amiodarone-induced thyrotoxicosis: a review. Can J Cardiol 2009; 25:421-424
  731. Mecacci F, Parretti E, Cioni R, Lucchetti R, Magrini A, La Torre P, Mignosa M, Acanfora L, Mello G. Thyroid autoimmunity and its association with non-organ-specific antibodies and subclinical alterations of thyroid function in women with a history of pregnancy loss or preeclampsia. J Reprod Immunol 2000; 46:39-50
  732. Stagnaro-Green A, Roman SH, Cobin RH, el-Harazy E, Alvarez-Marfany M, Davies TF. Detection of at-risk pregnancy by means of highly sensitive assays for thyroid autoantibodies. JAMA 1990; 264:1422-1425
  733. Glinoer D, Riahi M, Grun JP, Kinthaert J. Risk of subclinical hypothyroidism in pregnant women with asymptomatic autoimmune thyroid disorders. J Clin Endocrinol Metab 1994; 79:1970
  734. Premawardhana LD, Parkes AB, AMMARI F, John R, Darke C, Adams H, Lazarus JH. Postpartum thyroiditis and long-term thyroid status: prognostic influence of Thyroid Peroxidase Antibodies and ultrasound echogenicity. J Clin Endocrinol Metab 2000; 85:71-75
  735. Bussen S, Steck T, Dietl J. Increased prevalence of thyroid antibodies in euthyroid women with a history of recurrent in-vitro fertilization failure. Hum Reprod 2000; 15:545-548
  736. Muller AF, Drexhage HA, Berghout A. Postpartum thyroiditis and autoimmune thyroiditis in women of childbearing age: recent insights and consequences for antenatal and postnatal care. . Endocrinol Rev 2001; 22:605-630
  737. Vaquero E, Lazzarin N, De Carolis C, Valensise H, Moretti C, Romanini C. Mild thyroid abnormalities and recurrent spontaneous abortion: diagnostic and therapeutical approach. Am J Reprod Immunol 2000; 43:204-208
  738. Kim CH, Chae HD, Kang BM, Chang YS. Influence of antithyroid antibodies in euthyroid women on in vitro fertilization-embryo transfer outcome. Am J Reprod Immunol 1998; 40:2-8
  739. Poppe K, Glinoer D, tournaye H, Devroey P, Van Steirteghem a, Kaufman L, Velkeniers B. Assisted reproduction and thyroid autoimmunity: an unfortunate combination? J Clin Endocrinol Metab 2003; 88:4149-4152
  740. Negro R, Formoso G, Coppola L, Presicce G, Mangieri T, Pezzarossa A, Dazzi D. Euthyroid women with autoimmune disease undergoing assisted reproduction technologies: the role of autoimmunity and thyroid function. J Endocrinol Invest 2007; 30:3-8
  741. Karakosta P, Alegakis D, Georgiou V, Roumeliotaki T, Fthenou E, Vassilaki M, Boumpas D, Castanas E, Kogevinas M, Chatzi L. Thyroid Dysfunction and Autoantibodies in Early Pregnancy Are Associated with Increased Risk of Gestational Diabetes and Adverse Birth Outcomes. J Clin Endocrinol Metab 2012; 97:4464-4472
  742. He X, Wang P, Wang Z, He X, Xu D, Wang B. Thyroid antibodies and risk of preterm delivery: a meta-analysis of prospective cohort studies. Eur J Endocrinol 2012; 167:455-464
  743. McLachlan SM, Rapoport B. Why measure thyroglobulin autoantibodies rather than thyroid peroxidase autoantibodies. Thyroid 2004; 14:510-520
  744. Ericsson UB, Christensen SB, Thorell JI. A high prevalence of thyroglobulin autoantibodies in adults with and without thyroid disease as measured with a sensitive solid-phase immunosorbent radioassay. Clin Immunol Immunopathol 1985; 37:154-162
  745. Krahn J, Dembinski T. Thyroglobulin and anti-thyroglobulin assays in thyroid cancer monitoring. Clin Biochem 2009; 42:416-419
  746. Latrofa F, Ricci D, Montanelli L, Rocchi R, Piaggi P, Sisti E, Grasso L, Basolo F, Ugolini C, Pinchera A, Vitti P. Thyroglobulin autoantibodies in patients with papillary thyroid carcinoma: comparison of different assays and evaluation of causes of discrepancies. J Clin Endocrinol Metab 2012; 97:3974-3982
  747. Pickett AJ, Jones M, Evans C. Causes of discordance between thyroglobulin antibody assays. Ann Clin Biochem 2012; 49:463-467
  748. Pinchera A, Mariotti S, Vitti P, Tosi M, Grasso L, Facini F, Buti R, Baschieri L. Interference of serum thyroglobulin in the radioassay for serum antithyroglobulin antibodies. J Clin Endocrinol Metab 1977; 45:1077-1088
  749. Mariotti S, Pinchera A, Vitti P, Chiovato L, Marcocci C, Urbano C, Tosi M, Baschieri L. Comparison of radioassay and haemagglutination methods for anti-thyroid microsomal antibodies. Clin Exp Immunol 1978; 34:118-125
  750. Gao Y, Yuan Z, Yu Y, Lu H. Mutual interference between serum thyroglobulin and antithyroglobulin antibody in an automated chemiluminescent immunoassay. Clin Biochem 2007; 40:735-738
  751. Ruf J, Henry M, DeMicco C, Carayon P. Characterization of monoclonal and autoimmune antibodies to thyroglobulin : application to clinical investigation. In: Hufner M, Reiners C, eds Thyroglobulin and thyrolobulin antibodies in the follow-up of thyroid cancer and endemic goiter Stuttgart: G Thieme Verlag 1987:21-30
  752. Okosieme OE, Evans C, Moss L, Parkes AB, Premawardhana LD, Lazarus JH. Thyroglobulin antibodies in serum of patients with differentiated thyroid cancer: relationship between epitope specificities and thyroglobulin recovery. Clin Chem 2005; 51:729-734
  753. Taylor KP, Parkington D, Bradbury S, Simpson HL, Jefferies SJ, Halsall DJ. Concordance between thyroglobulin antibody assays. Ann Clin Biochem 2011; 48:367-369
  754. Jensen EA, Petersen PH, Blaabjerg O, Hansen PS, Brix TH, Hegedüs L. Establishment of reference distributions and decision values for thyroid antibodies against thyroid peroxidase (TPOAb), thyroglobulin (TgAb) and the thyrotropin receptor (TRAb). Clin Chem Lab Med 2006; 44:991-998
  755. Ruf J, Carayon P, Lissitzky S. Various expressions of a unique anti-human thyroglobulin antibody repertoire in normal state and autoimmune disease. Eur J Immunol 1985; 15:268-272
  756. Doullay F, Ruf J, Codaccioni JL, et al. Prevalence of autoantibodies to thyroperoxidase in patients with various thyroid and autoimmune diseases. Autoimmunity 1991; 9:237-244
  757. Aras G, Gültekin SS, Küçük NO. The additive clinical value of combined thyroglobulin and antithyroglobulin antibody measurements to define persistent and recurrent disease in patients with differentiated thyroid cancer. Nucl Med Commun 2008; 29:880-884
  758. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL, Mandel SJ, Mazzaferri EL, McIver B, Sherman SI, Tuttle RM. Management guidelines for patients with thyroid nodules and differentiated thyroid cancer. The American Thyroid Association Guidelines Taskforce. Thyroid 2006; 16:109-142
  759. Kumar A, Shah DH, Shrihari U, Dandekar SR, Vijayan U, Sharma SM. Significance of antithyroglobulin autoantibodies in differentiated thyroid carcinoma. Thyroid 1994; 4:199-202
  760. Gorges R, Maniecki M, Jentzen W, Sheu SN, Mann K, Bockisch A, Janssen OE. Development and clinical impact of thyroglobulin antibodies in patients with differentiated thyroid carcinoma during the first 3 years after thyroidectomy. Eur J Endocrinol 2005; 153:49-55
  761. Spencer CA. Clinical review: Clinical utility of thyroglobulin antibody (TgAb) measurements for patients with differentiated thyroid cancers (DTC). J Clin Endocrinol Metab 2011; 96:3615-3627
  762. Kim WG, Yoon JH, Kim WB, Kim TY, Kim EY, Kim JM, Ryu JS, Gong G, Hong SJ, Shong YK. Change of serum antithyroglobulin antibody levels is useful for prediction of clinical recurrence in thyroglobulin-negative patients with differentiated thyroid carcinoma. J Clin Endocrinol Metab 2008; 93:4683-4689
  763. Pacini F, Mariotti S, Formica N, Elisei R. Thyroid autoantibodies in thyroid cancer: Incidence and relationship with tumor outcome. Acta Endocrinol (Copenh) 1988; 119:373-380
  764. Rubello D, Casara D, Girelli ME, Piccolo M, Busnardo B. Clinical meaning of circulating antithyroglobulin antibodies in differentiated thyroid cancer: a prospective study. J Nucl Med 1992; 33:1478-1480
  765. Spencer CA. New Insights for Using Serum Thyroglobulin (Tg) Measurement for Managing Patients with Differentiated Thyroid Carcinomas. Thyroid International 2003; 4:1-14
  766. Chiovato L, Latrofa F, Braverman LE, Pacini F, Capezzone M, Masserini L, Grasso L, Pinchera A. Disappearance of humoral thyroid autoimmunity after complete removal of thyroid antigens. Ann Intern Med 2003; 139:346-351
  767. Tomer Y, Greenberg D. The thyroglobulin gene as the first thyroid-specific susceptibility gene for autoimmune thyroid disease. Trends Mol Med 2004; 10:306-308
  768. Van den Briel T, West CE, Hautvast JG, Vulsma T, de Vijlder JJ, Ategbo EA. Serum thyroglobulin and urinary iodine concentration are the most appropriate indicators of iodine status and thyroid function under conditions of increasing iodine supply in schoolchildren in Benin. J Nutr 2001; 131:2701-2706
  769. Vejbjerg P, Knudsen N, Perrild H, Laurberg P, Carlé A, Pedersen IB, Rasmussen LB, Ovesen L, Jørgensen T. Thyroglobulin as a marker of iodine nutrition status in the general population. Eur J Endocrinol 2009; 161:475-481
  770. Wang Z, Zhang H, Zhang X, Sun J, Han C, Li C, Li Y, Teng X, Fan C, Liu A, Shan Z, Liu C, Weng J, Teng W. Serum thyroglobulin reference intervals in regions with adequate and more than adequate iodine intake. Medicine (Baltimore) 2016; 95:e5273
  771. Bath SC, Pop VJ, Furmidge-Owen VL, Broeren MA, Rayman MP. Thyroglobulin as a Functional Biomarker of Iodine Status in a Cohort Study of Pregnant Women in the United Kingdom. Thyroid 2016;
  772. Vali M, Rose NR, Caturegli P. Thyroglobulin as autoantigen: structure-function relationships. Rev Endocr Metab Disord 2000; 1:69-77
  773. Knobel M, Medeiros-Neto G. An outline of inherited disorders of the thyroid hormone generating system. Thyroid 2003; 13:771-801
  774. Niu DM, Hsu JH, Chong KW, Huang CH, Lu YH, Kao CH, Yu HC, Lo MY, Jap TS. Six new mutations of the thyroglobulin gene discovered in taiwanese children presenting with thyroid dyshormonogenesis. J Clin Endocrinol Metab 2009; 94:5045-5052
  775. Citterio CE, Machiavelli GA, Miras MB, Gruñeiro-Papendieck L, Lachlan K, Sobrero G, Chiesa A, Walker J, Muñoz L, Testa G, Belforte FS, González-Sarmiento R, Rivolta CM, Targovnik HM. New insights into thyroglobulin gene: Molecular analysis of seven novel mutations associated with goiter and hypothyroidism. Mol Cell Endocrinol 2013; 365:277-291
  776. Fugazzola L, Persani L, Mannavola D, Reschini E, Vannucchi G, Weber G, Beck-Peccoz P. Recombinant human TSH testing is a valuable tool for differential diagnosis of congenital hypothyroidism during L-thyroxine replacement. Clin Endocrinol (Oxf) 2003; 59:230-236
  777. Simsek E, Karabay M, Kocabay K. Neonatal screening for congenital hypothyroidism in West Black Sea area, Turkey. Int J Clin Pract 2005; 59:336-341
  778. Mariotti S, Martino E, Cupini C, Lari R, Giani C, Baschieri L, Pinchera A. Low serum thyroglobulin as a clue to the diagnosis of thyrotoxicosis factitia. N Engl J Med 1982; 307:410-412
  779. Pacini F, Pinchera A. Serum and tissue thyroglobulin measurement: clinical applications in thyroid disease. Biochimie 1999; 81:463-467
  780. Chow E, Siddique F, Gama R. Thyrotoxicosis factitia: role of thyroglobulin. Ann Clin Biochem 2008; 45:447-448
  781. Torrens JI, Burch HB. Serum thyroglobulin measurement. Utility in clinical practice. Endocrinol Metab Clin N Amer 2001; 30:429-467
  782. Pacini F, Schlumberger M, Dralle H, Elisei R, Smit JW, Wiersinga W. European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur J Endocrinol 2006; 154:787-803
  783. Iervasi A, Iervasi G, Carpi A, Zucchelli GC. Serum thyroglobulin measurement: clinical background and main methodological aspects with clinical impact. Biomed Pharmacother 2006; 60:414-424
  784. Spencer CA, Petrovic I. Thyroglobuin measurement. In: Thyroid Function Testing Editor Gregory Brent MD Springer Science, MA, USA 2010
  785. Tuttle RM, Leboeuf R. Follow up Approaches in Thyroid Cancer: A Risk Adapted Paradigm. Endocrinol Metab Clin North Am 2008; 37:419-435
  786. Lin JD. Thyroglobulin and human thyroid cancer. Clin Chim Acta 2008; 388:15-21
  787. Pitoia F, Ward L, Wohllk N, Friguglietti C, Tomimori E, Gauna A, Camargo R, Vaisman M, Harach R, Munizaga F, Corigliano S, Pretell E, Niepomniszcze H. Recommendations of the Latin American Thyroid Society on diagnosis and management of differentiated thyroid cancer. Arq Bras Endocrinol Metabol 2009; 53:884-887
  788. Brassard M, Borget I, Edet-Sanson A, Giraudet AL, Mundler O, Toubeau M, Bonichon F, Borson-Chazot F, Leenhardt L, Schvartz C, Dejax C, Brenot-Rossi I, Toubert ME, Torlontano M, Benhamou E, Schlumberger M. Long-term follow-up of patients with papillary and follicular thyroid cancer: a prospective study on 715 patients. J Clin Endocrinol Metab 2011; 96:1352-1359
  789. Rosario PW, Borges MA, Fagundes TA, Franco AC, Purisch S. Is stimulation of thyroglobulin (Tg) useful in low-risk patients with thyroid carcinoma and undetectable Tg on thyroxin and negative neck ultrasound? Clin Endocrinol (Oxf) 2005; 62
  790. Grebe SKG. Diagnosis and management of thyroid carcinoma: a focus on serum thyroglobulin. Expert Rev Endocrinol Metab 2009; 4:25-43
  791. Malandrino P, Latina A, Marescalco S, Spadaro A, Regalbuto C, Fulco RA, Scollo C, Vigneri R, Pellegriti G. Risk-adapted management of differentiated thyroid cancer assessed by a sensitive measurement of basal serum thyroglobulin. J Clin Endocrinol Metab 2011; 96:1703-1709
  792. Chindris AM, Diehl NN, Crook JE, Fatourechi V, Smallridge RC. Undetectable sensitive serum thyroglobulin (<0.1 ng/ml) in 163 patients with follicular cell-derived thyroid cancer: results of rhTSH stimulation and neck ultrasonography and long-term biochemical and clinical follow-up. J Clin Endocrinol Metab 2012; 97:2714-2723
  793. Trimboli P, La Torre D, Ceriani L, Condorelli E, Laurenti O, Romanelli F, Ventura C, Signore A, Valabrega S, Giovanella L. High sensitive thyroglobulin assay on thyroxine therapy: can it avoid stimulation test in low and high risk differentiated thyroid carcinoma patients? Horm Metab Res 2013; 45:664-668
  794. Mariotti S, Barbesino G, Caturegli P, Marino M, Manetti L, Pacini F, Centoni R, Pinchera A. Assay of thyroglobulin in serum with thyroglobulin autoantibodies: an unobtainable goal? J Clin Endocrinol Metab 1995; 80:468-472
  795. Spencer CA, Takeuchi M, Kazarosyan M. Current status and performance goals for serum thyroglobulin assays. Clin Chem 1996; 42:164-173
  796. Spencer CA, Takeuchi M, Kazarosyan M, Wang CC, Guttler RB, Singer PA, Fatemi S, LoPresti JS, Nicoloff JT. Serum thyroglobulin autoantibodies: prevalence, influence on serum thyroglobulin measurement, and prognostic significance in patients with differentiated thyroid carcinoma. J Clin Endocrinol Metab 1998; 83:1121-1127
  797. Clark P, Franklyn J. Can we interpret serum thyroglobulin results? Ann Clin Biochem 2012; 49:313-322
  798. Latrofa F, Ricci D, Sisti E, Piaggi P, Nencetti C, Marino M, Vitti P. Significance of Low Levels of Thyroglobulin Autoantibodies Associated with Undetectable Thyroglobulin After Thyroidectomy for Differentiated Thyroid Carcinoma. Thyroid 2016; 26:798-806
  799. Netzel BC, Grebe SK, Carranza Leon BG, Castro MR, Clark PM, Hoofnagle AN, Spencer CA, Turcu AF, Algeciras-Schimnich A. Thyroglobulin (Tg) Testing Revisited: Tg Assays, TgAb Assays, and Correlation of Results With Clinical Outcomes. J Clin Endocrinol Metab 2015; 100:E1074-1083
  800. Jindal A, Khan U. Is Thyroglobulin Level by Liquid Chromatography Tandem-Mass Spectrometry Always Reliable for Follow-Up of DTC After Thyroidectomy: A Report on Two Patients. Thyroid 2016; 26:1334-1335
  801. Azmat U, Porter K, Senter L, Ringel MD, Nabhan F. Thyroglobulin Liquid Chromatography-Tandem Mass Spectrometry Has a Low Sensitivity for Detecting Structural Disease in Patients with Antithyroglobulin Antibodies. Thyroid 2016;
  802. Rosario PW, Mourao GF, Calsolari MR. Low postoperative nonstimulated thyroglobulin as a criterion for the indication of low radioiodine activity in patients with papillary thyroid cancer of intermediate risk 'with higher risk features'. Clin Endocrinol (Oxf) 2016; 85:453-458
  803. Spencer CA, Wang CC. Thyroglobulin measurement. Techniques, clinical benefits, and pitfalls. Endocrinol Metab Clin North Am 1995; 24:841-863
  804. Spencer CA, Wang CC. Thyroglobulin measurement:- Techniques, clinical benefits and pitfalls. Endocrinol Metab Clin N Amer 1995; 24:841-863
  805. Zucchelli G, Iervasi A, Ferdeghini M, Iervasi G. Serum thyroglobulin measurement in the follow-up of patients treated for differentiated thyroid cancer. Q J Nucl Med Mol Imaging 2009; 53:482-489
  806. Zophel K, Wunderlich G, Smith BR. Serum thyroglobulin measurements with a high sensitivity enzyme-linked immunosorbent assay: is there a clinical benefit in patients with differentiated thyroid carcinoma? Thyroid 2003; 13:861-865
  807. Iervasi A, Iervasi G, Ferdeghini M, Solimeo C, Bottoni A, Rossi L, Colato C, Zucchelli GC. Clinical relevance of highly sensitive Tg assay in monitoring patients treated for differentiated thyroid cancer. Clin Endocrinol (Oxf) 2007; 67:434-441
  808. Giovanella L. Highly sensitive thyroglobulin measurements in differentiated thyroid carcinoma management. Clin Chem Lab Med 2008; 46:1067-1073
  809. Tomoda C, Miyauchi A. Undetectable serum thyroglobulin levels in patients with medullary thyroid carcinoma after total thyroidectomy without radioiodine ablation. Thyroid 2012; 22:680-682
  810. Angell TE, Spencer CA, Rubino BD, Nicoloff JT, LoPresti JS. In Search of an Unstimulated Thyroglobulin Baseline Value in Low-Risk Papillary Thyroid Carcinoma Patients Not Receiving Radioactive Iodine Ablation. Thyroid 2014; 24:1127-1133
  811. Haugen BR, Ladenson PW, Cooper DS, Pacini F, Reiners C, Luster M, Schlumberger M, Sherman SI, Samuels M, Graham K, Braverman LE, Skarulis MC, Davies TF, DeGroot L, Mazzaferri EL, Daniels GH, Ross DC, Becker DV, Mazon HR, Cavalieri RR, Spencer CA, McEllin K, Weintraub BD, EC. R. A comparison of Recombinant Human Thyrotropin and Thyroid Hormone Withdrawal for the Detection of Thyroid Remnant or Cancer. J Clin Endocrinol Metab 1999; 84:3877-3885
  812. Pacini F, Castagna MG. Diagnostic and therapeutic use of recombinant human TSH (rhTSH) in differentiated thyroid cancer. Best Pract Res Clin Endocrinol Metab 2008; 22:1009-1021
  813. Kloos RT, Mazzaferri EL. A single recombinant human thyrotropin-stimulated serum thyroglobulin measurement predicts differentiated thyroid carcinoma metastases three to five years later. J Clin Endocrinol Metab 2005; 90:5047-5057
  814. Mazzaferri EL. Will highly sensitive thyroglobulin assays change the management of thyroid cancer? Clin Endocrinol (Oxf) 2007; 67:321-323
  815. Persoon AC, Jager PL, Sluiter WJ, Plukker JT, Wolffenbuttel BH, Links TP. A sensitive Tg assay or rhTSH stimulated Tg: what's the best in the long-term follow-up of patients with differentiated thyroid carcinoma. PLoS ONE 2007; 2:e816
  816. Spencer CA, Lopresti JS. Measuring thyroglobulin and thyroglobulin autoantibody in patients with differentiated thyroid cancer. Nat Clin Pract Endocrinol Metab 2008; 4:223-233
  817. Spencer CA, Lopresti JS. Measuring thyroglobulin and thyroglobulin autoantibody in patients with differentiated thyroid cancer. Nat Clin Pract Endocrinol Metab 2008; 4:223-233
  818. Bachelot A, Cailleux AF, Klain M, Baudin E, Ricard M, Bellon N, Caillou B, Travagli JP, Schlumberger M. Relationship between tumor burden and serum thyroglobulin level in patients with papillary and follicular thyroid carcinoma. Thyroid 2002; 12:707-711
  819. Pacini F, Molinaro E, Castagna MG, Agate L, Elisei R, Ceccarelli C, Lippi F, Taddei D, Grasso L, Pinchera A. Recombinant human thyrotropin-stimulated serum thyroglobulin combined with neck ultrasonography has the highest sensitivity in monitoring differentiated thyroid carcinoma. J Clin Endocrinol Metab 2003; 88:3668-3673
  820. Heilo A, Sigstad E, Fagerlid KH, Håskjold OI, Grøholt KK, Berner A, Bjøro T, Jørgensen LH. Efficacy of ultrasound-guided percutaneous ethanol injection treatment in patients with a limited number of metastatic cervical lymph nodes from papillary thyroid carcinoma. J Clin Endocrinol Metab 2011; 96:2750-2755
  821. Hay ID, Lee RA, Davidge-Pitts C, Reading CC, Charboneau JW. Long-term outcome of ultrasound-guided percutaneous ethanol ablation of selected "recurrent" neck nodal metastases in 25 patients with TNM stages III or IVA papillary thyroid carcinoma previously treated by surgery and 131I therapy. Surgery 2013; 154:1448-1454; discussion 1454-1445
  822. Feldt-Rasmussen U, Profilis C, Colinet E, Black E, Bornet H, Bourdoux P, Carayon P, Ericsson UB, Koutras DA, Lamas de Leon L, DeNayer P, Pacini F, Palumbo G, Santos A, Schlumberger M, Seidel C, Van Herle AJ, DeVijlder JJM. Human thyroglobulin reference material (CRM 457) 1st part: Assessment of homogeneity, stability and immunoreactivity. Ann Biol Clin 1996; 54:337-342
  823. Feldt-Rasmussen U, Profilis C, Colinet E, Black E, Bornet H, Bourdoux P, Carayon P, Ericsson UB, Koutras DA, Lamas de Leon L, DeNayer P, Pacini F, Palumbo G, Santos A, Schlumberger M, Seidel C, Van Herle AJ, DeVijlder JJM. Human thyroglobulin reference material (CRM 457) 2nd part: Physicochemical characterization and certification. Ann Biol Clin 1996; 54:343-348
  824. Kim M, Jeon MJ, Kim WG, Lee JJ, Ryu JS, Cho EJ, Ko DH, Lee W, Chun S, Min WK, Kim TY, Shong YK, Kim WB. Comparison of Thyroglobulin Measurements Using Three Different Immunoassay Kits: A BRAMHS Tg-Plus RIA Kit, a BRAMHS hTg Sensitive Kryptor Kit, and a Beckman Coulter ACCESS Immunoassay Kit. Endocrinology and metabolism (Seoul, Korea) 2016;
  825. Feldt-Rasmussen U, Petersen PH, Blaabjerg O, Horder M. Long-term variability in serum thyroglobulin and thyroid related hormones in healthy subjects. Acta Endocrinol (Copenh) 1980; 95:328-334
  826. Heilig B, Hufner M, Dorken B, Schmidt-Gayk H. Increased heterogeneity of serum thyroglobulin in thyroid cancer patients as determined by monoclonal antibodies. Klin Wochenschr 1986; 64:776-780
  827. Schulz R, Bethauser H, Stempka L, Heilig B, Moll A, Hufner M. Evidence for immunological differences between circulating and tissue-derived thyroglobulin in men. Eur J Clin Invest 1989; 19:459-463
  828. de Micco C, Ruf J, Carayon P, Chrestian MA, Henry JF, Toga M. Immunohistochemical study of thyroglobulin in thyroid carcinomas with monoclonal antibodies. Cancer 1987; 59:471-476
  829. Kim PS, Dunn AD, Dunn JT. Altered immunoreactivity of thyroglobulin in thyroid disease. J Clin Endocrinol Metab 1988; 67:161-168
  830. Cubero JM, Rodríguez-Espinosa J, Gelpi C, Estorch M, Corcoy R. Thyroglobulin autoantibody levels below the cut-off for positivity can interfere with thyroglobulin measurement. Thyroid 2003; 13:659-661
  831. Cole TG, Johnson D, Eveland BJ, Nahm MH. Cost-effective method for detection of "hook effect" in tumor marker immunometric assays. Clin Chem 1993; 39:695-696
  832. Basuyau JP, Leroy M, Brunelle P. Determination of Tumor Markers in Serum. Pitfalls and Good Practice. Clin Chem Lab Med 2001; 39:1227-1233
  833. Leboeuf R, Langlois MF, Martin M, Ahnadi CE, Fink GD. "Hook effect" in calcitonin immunoradiometric assay in patients with metastatic medullary thyroid carcinoma: case report and review of the literature. J Clin Endocrinol Metab 2006; 91:361-364
  834. Jassam N, Jones CM, Briscoe T, Horner JH. The hook effect: a need for constant vigilance. Ann Clin Biochem 2006; 43:314-317
  835. Selby C. Interference in immunoassay. Ann Clin Biochem 1999; 36:704-721
  836. Morgenthaler NG, Froehlich J, Rendl J, Willnich M, Alonso C, Bergmann A, Reiners C. Technical evaluation of a new immunoradiometric and a new immunoluminometric assay for thyroglobulin. Clin Chem 2002; 48:1077-1083
  837. Netzel BC, Grebe SK, Algeciras-Schimnich A. Usefulness of a thyroglobulin liquid chromatography-tandem mass spectrometry assay for evaluation of suspected heterophile interference. Clin Chem 2014; 60:1016-1018
  838. Schaadt B, Feldt-Rasmussen U, Rasmussen B, Torring H, Foder B, Jorgensen K, Sand Hansen H. Assessment of the influence of thyroglobulin (Tg) autoantibodies and other interfering factors on the use of serum Tg as tumor marker in differentiated thyroid carcinoma. Thyroid 1995; 5:165-170
  839. Spencer CA. Recoveries cannot be used to authenticate thyroglobulin (Tg) measurements when sera contain Tg autoantibodies. Clin Chem 1996; 42:661-663
  840. Weigle WO, High GJ. The behaviour of autologous thyroglobulin in the circulation of rabbits immunized with either heterologous or altered homologous thyroglobulin. J Immunol 1967; 98:1105-1114
  841. Sellitti DF, Suzuki K. Intrinsic regulation of thyroid function by thyroglobulin. Thyroid 2014; 24:625-638
  842. Igawa T, Haraya K, Hattori K. Sweeping antibody as a novel therapeutic antibody modality capable of eliminating soluble antigens from circulation. Immunological reviews 2016; 270:132-151
  843. Tozzoli R, Bizzaro N, Tonutti E, Pradella M, Manoni F, Vilalta D, Bassetti D, Piazza A, Rizzotti P. Immunoassay of anti-thyroid autoantibodies: high analytical variability in second generation methods. Clin Chem Lab Med 2002; 40:568-573
  844. Benvenga S, Burek CL, Talor M, Rose NR, Trimarchi F. Heterogeneity of the thyroglobulin epitopes associated with circulating thyroid hormone autoantibodies in hashimoto's thyroiditis and non-autoimmune thyroid diseases. J Endocrinol Invest 2002; 25:977-982
  845. Rosário PW, Maia FF, Fagundes TA, Vasconcelos FP, Cardoso LD, Purisch S. Antithyroglobulin antibodies in patients with differentiated thyroid carcinoma: methods of detection, interference with serum thyroglobulin measurement and clinical significance. Arq Bras Endocrinol Metabol 2004; 48:487-492
  846. McLachlan SM, B. R. Why measure thyroglobulin autoantibodies rather than thyroid peroxidase autoantibodies. Thyroid 2004; 14:510-520
  847. Schneider AB, Pervos R. Radioimmunoassay of human thyroglobulin: effect of antithyroglobulin autoantibodies. J Clin Endocrinol Metab 1978; 47:126-137
  848. Feldt-Rasmussen U, Rasmussen AK. Serum thyroglobulin (Tg) in presence of thyroglobulin autoantibodies (TgAb). Clinical and methodological relevance of the interaction between Tg and TgAb in vivo and in vitro. J Endocrinol Invest 1985; 8:571-576
  849. Crane MS, Strachan MW, Toft AD, Beckett GJ. Discordance in thyroglobulin measurements by radioimmunoassay and immunometric assay: a useful means of identifying thyroglobulin assay interference. Ann Clin Biochem 2013; 50:421-432
  850. Spencer C, Fatemi S. Thyroglobulin antibody (TgAb) methods - Strengths, pitfalls and clinical utility for monitoring TgAb-positive patients with differentiated thyroid cancer. Best Pract Res Clin Endocrinol Metab 2013; 27:701-712
  851. Feldt-Rasmussen U, Petersen PH, Date J, Madsen CM. Sequential changes in serum thyroglobulin (Tg) and its autoantibodies (TgAb) following subtotal thyroidectomy of patients with preoperatively detectable TgAb. Clin Endocrinol (Oxf) 1980; 12:29-38
  852. Fleck RA, Rapaport SI, Rao LV. Anti-prothrombin antibodies and the lupus anticoagulant. Blood 1988; 72:512-519
  853. van der Laken CJ, Voskuyl AE, Roos JC, Stigter van Walsum M, de Groot ER, Wolbink G, Dijkmans BA, Aarden LA. Imaging and serum analysis of immune complex formation of radiolabelled infliximab and anti-infliximab in responders and non-responders to therapy for rheumatoid arthritis. Ann Rheum Dis 2007; 66:253-256
  854. Richards DB, Cookson LM, Berges AC, Barton SV, Lane T, Ritter JM, Fontana M, Moon JC, Pinzani M, Gillmore JD, Hawkins PN, Pepys MB. Therapeutic Clearance of Amyloid by Antibodies to Serum Amyloid P Component. N Engl J Med 2015; 373:1106-1114
  855. Van Herle AJ, Uller RP. Elevated serum thyroglobulin: a marker of metastases in differentiated thyroid carcinomas. J Clin Invest 1975; 56:272-277
  856. Spencer CA, Platler BW, Nicoloff JT. The effect of 125-I thyroglobulin tracer heterogeneity on serum Tg RIA measurement. Clin Chim Acta 1985; 153:105-115
  857. Spencer CA, Platler B, Guttler RB, Nicoloff JT. Heterogeneity of 125-I labelled thyroglobulin preparations. Clin Chim Acta 1985; 151:121-132
  858. Black EG, Hoffenberg R. Should one measure serum thyroglobulin in the presence of anti-thyroglobulin antibodies? Clin Endocrinol (Oxf) 1983; 19:597-601
  859. Weightman DR, Mallick UK, Fenwick JD, Perros P. Discordant serum thyroglobulin results generated by two classes of assay in patients with thyroid carcinoma: correlation with clinical outcome after 3 years of follow-up. Cancer 2003; 98:41-47
  860. Jahagirdar VR, Strouhal P, Holder G, Gama R, Singh BM. Thyrotoxicosis factitia masquerading as recurrent Graves' disease: endogenous antibody immunoassay interference, a pitfall for the unwary. Ann Clin Biochem 2008; 45:325-327
  861. Bayer MF, Kriss JP. Immunoradiometric assay for serum thyroglobulin: semiquantitative measurement of thyroglobulin in antithyroglobulin-positive sera. J Clin Endocrinol Metab 1979; 49:557-564
  862. Latrofa F, Ricci D, Grasso L, Vitti P, Masserini L, Basolo F, Ugolini C, Mascia G, Lucacchini A, Pinchera A. Characterization of thyroglobulin epitopes in patients with autoimmune and non-autoimmune thyroid diseases using recombinant human monoclonal thyroglobulin autoantibodies. J Clin Endocrinol Metab 2008; 93:591-596
  863. Rahmoun MN, Bendahmane I. Anti-thyroglobulin antibodies in differentiated thyroid carcinoma patients: Study of the clinical and biological parameters. Ann Endocrinol (Paris) 2014; 75:15-18
  864. Marquet PY, Daver A, Sapin R, Bridgi B, Muratet JP, Hartmann DJ, Paolucci F, Pau B. Highly sensitive immunoradiometric assay for serum thyroglobulin with minimal interference from autoantibodies. Clin Chem 1996; 42:258-262
  865. Haapala AM, Soppi E, Morsky P, al. e. Thyroid antibodies in association with thyroid malignancy II: qualitative properties of thyroglobulin antibodies. Scand J Clin Lab Invest 1995; 55:317-322
  866. Netzel BC, Grant RP, Hoofnagle AN, Rockwood AL, Shuford CM, Grebe SK. First Steps toward Harmonization of LC-MS/MS Thyroglobulin Assays. Clin Chem 2016; 62:297-299
  867. Petrovic I, S. Fatemi, J. LoPresti, S.K. Grebe, A. Algeciras-Schimnich, B.C. Netzel, C. Spencer. SerumTg is frequently undetectable by mass spectrometry (Tg-MS) IN TgAb-positive differentiated thyroid cancer (DTC) patients with structural disease. . Thyroid 2015; 25:A251
  868. Chung JK, Park YJ, Kim TY, So Y, Kim SK, Park DJ, Lee DS, Lee MC, Cho BY. Clinical significance of elevated level of serum antithyroglobulin antibody in patients with differentiated thyroid cancer after thyroid ablation. Clin Endocrinol (Oxf) 2002; 57:215-221
  869. Küçük ON, Aras G, Kulak HA, Ibiş E. Clinical importance of anti-thyroglobulin auto-antibodies in patients with differentiated thyroid carcinoma: comparison with 99mTc-MIBI scans. Nucl Med Commun 2006; 27:873-876
  870. Thomas D, Liakos V, Vassiliou E, Hatzimarkou F, Tsatsoulis A, Kaldrimides P. Possible reasons for different pattern disappearance of thyroglobulin and thyroid peroxidase autoantibodies in patients with differentiated thyroid carcinoma following total thyroidectomy and iodine-131 ablation. J Endocrinol Invest 2007; 30:173-180
  871. Feldt-Rasmussen U, Rasmussen AK. Autoimmunity in differentiated thyroid cancer: significance and related clinical problems. Hormones (Athens) 2010; 9:109-117
  872. Hsieh CJ, Wang PW. Sequential changes of serum antithyroglobulin antibody levels are a good predictor of disease activity in thyroglobulin-negative patients with papillary thyroid carcinoma. Thyroid 2014; 24:488-493
  873. Feldt-Rasmussen U, Verburg FA, Luster M, Cupini C, Chiovato L, Duntas L, Elisei R, Rimmele H, Seregni E, Smit JW, Theimer C, Giovanella L. Thyroglobulin autoantibodies as surrogate biomarkers in the management of patients with differentiated thyroid carcinoma. Current medicinal chemistry 2014; 21:3687-3692
  874. Rosario PW, Carvalho M, Mourao GF, Calsolari MR. Comparison of Antithyroglobulin Antibody Concentrations Before and After Ablation with 131I as a Predictor of Structural Disease in Differentiated Thyroid Carcinoma Patients with Undetectable Basal Thyroglobulin and Negative Neck Ultrasonography. Thyroid 2016; 26:525-531
  875. Nascimento C, Borget I, Troalen F, Al Ghuzlan A, Deandreis D, Hartl D, Lumbroso J, Chougnet CN, Baudin E, Schlumberger M, Leboulleux S. Ultrasensitive serum thyroglobulin measurement is useful for the follow-up of patients treated with total thyroidectomy without radioactive iodine ablation. Eur J Endocrinol 2013; 169:689-693
  876. Spencer C, Fatemi S. Thyroglobulin antibody (TgAb) methods - Strengths, pitfalls and clinical utility for monitoring TgAb-positive patients with differentiated thyroid cancer. Best Pract Res Clin Endocrinol Metab 2013; 27:701-712
  877. Uller RP, Van Herle AJ. Effect of therapy on serum thyroglobulin levels in patients with Graves' disease. J Clin Endocrinol Metab 1978; 46:747-755
  878. Feldt-Rasmussen U, Blichert-Toft M, Christiansen C, Date J. Serum thyroglobulin and its autoantibody following subtotal thyroid resection of Graves' disease. Eur J Clin Invest 1982; 12:203-208
  879. Benvenga S, Bartolone L, Squadrito S, Trimarchi F. Thyroid hormone autoantibodies elicited by diagnostic fine needle biopsy. J Clin Endocrinol Metab 1997; 82:4217-4223
  880. Polyzos SA, Anastasilakis AD. Alterations in serum thyroid-related constituents after thyroid fine-needle biopsy: a systematic review. Thyroid 2010; 20:265-271
  881. Izumi M, Larsen PR. Correlation of sequential changes in serum thyroglobulin, triiodothyronine, and thyroxine in patients with Graves' disease and subacute thyroiditis. Metabolism 1978; 27:449-460
  882. Gardner DF, Rothman J, Utiger RD. Serum thyroglobulin in normal subjects and patients with hyperthyroidism due to Graves' disease: effects of T3, iodide, 131I and antithyroid drugs. Clin Endocrinol (Oxf) 1979; 11:585-594
  883. Feldt-Rasmussen U, Bech K, Date J, Hyltoft Pedersen P, Johansen K, Nistrup Madsen S. Thyroid stimulating antibodies, thyroglobulin antibodies and serum proteins during treatment of Graves' disease with radioiodine or propylthiouracil. Allergy 1982; 37:161-167
  884. Feldt-Rasmussen U, Bech K, Date J, Petersen PH, Johansen K. A prospective study of the differential changes in serum thyroglobulin and its autoantibodies during propylthiouracil or radioiodine therapy of patients with Graves' disease. Acta Endocrinol (Copenh) 1982; 99:379-385
  885. Yamada O, Miyauchi A, Ito Y, Nakayama A, Yabuta T, Masuoka H, Fukushima M, Higashiyama T, Kihara M, Kobayashi K, Miya A. Changes in serum thyroglobulin antibody levels as a dynamic prognostic factor for early-phase recurrence of thyroglobulin antibody-positive papillary thyroid carcinoma after total thyroidectomy. Endocr J 2014; 61:961-965
  886. Tsushima Y, Miyauchi A, Ito Y, Kudo T, Masuoka H, Yabuta T, Fukushima M, Kihara M, Higashiyama T, Takamura Y, Kobayashi K, Miya A, Kikumori T, Imai T, Kiuchi T. Prognostic significance of changes in serum thyroglobulin antibody levels of pre- and post-total thyroidectomy in thyroglobulin antibody-positive papillary thyroid carcinoma patients. Endocr J 2013; 60:871-876
  887. Tumino S, Belfiore A. Appearance of antithyroglobulin antibodies as the sole sign of metastatic lymph nodes in a patient operated on for papillary thyroid cancer: a case report. Thyroid 2000; 10:431-433
  888. Donegan D, McIver B, Algeciras-Schimnich A. Clinical Consequences of a Change in Anti-Thyroglobulin Antibody Assays During the Follow-Up of Patients with Differentiated Thyroid Cancer. Endocr Pract 2014:1-19
  889. Lupoli GA, Okosieme OE, Evans C, Clark PM, Pickett AJ, Premawardhana LD, Lupoli G, Lazarus JH. Prognostic significance of thyroglobulin antibody epitopes in differentiated thyroid cancer. J Clin Endocrinol Metab 2015; 100:100-108
  890. Sapin R, d'Herbomez M, Gasser F, Meyer L, Schlienger JL. Increased sensitivity of a new assay for anti-thyroglobulin antibody detection in patients with autoimmune thyroid disease. Clin Biochem 2003; 36:611-616
  891. Gianoukakis AG. Thyroglobulin antibody status and differentiated thyroid cancer: what does it mean for prognosis and surveillance? Current opinion in oncology 2015; 27:26-32
  892. Kim ES, Lim DJ, Baek KH, Lee JM, Kim MK, Kwon HS, Song KH, Kang MI, Cha BY, Lee KW, Son HY. Thyroglobulin Antibody Is Associated with Increased Cancer Risk in Thyroid Nodules. Thyroid 2010; 20:885-891
  893. Vasileiadis I, Boutzios G, Charitoudis G, Koukoulioti E, Karatzas T. Thyroglobulin antibodies could be a potential predictive marker for papillary thyroid carcinoma. Ann Surg Oncol 2014; 21:2725-2732
  894. Grani G, Calvanese A, Carbotta G, D'Alessandri M, Nesca A, Bianchini M, Del Sordo M, Vitale M, Fumarola A. Thyroid autoimmunity and risk of malignancy in thyroid nodules submitted to fine-needle aspiration cytology. Head Neck 2015; 37:260-264
  895. Karatzas T, Vasileiadis I, Zapanti E, Charitoudis G, Karakostas E, Boutzios G. Thyroglobulin antibodies as a potential predictive marker of papillary thyroid carcinoma in patients with indeterminate cytology. Am J Surg 2016;
  896. Petrovic I, S. Fatemi, J. LoPresti, C. Spencer. Follow-Up Time Needed for TgAb+ Differentiated Thyroid Cancer (DTC) Patients To Convert to TgAb-

Negativity Following Successful Surgery Relates to Initial TgAb Concentration and Not Radioiodine (RAI) Remnant

Ablation. Thyroid 2015; 25:A130

  1. Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immunity due to long-lived plasma cells. Immunity 1998; 8:363-372
  2. Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R. Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. Journal of cancer epidemiology 2013; 2013:965212
  3. Ahn HS, Kim HJ, Welch HG. Korea's thyroid-cancer "epidemic"--screening and overdiagnosis. N Engl J Med 2014; 371:1765-1767
  4. Oda H, Miyauchi A, Ito Y, Yoshioka K, Nakayama A, Sasai H, Masuoka H, Yabuta T, Fukushima M, Higashiyama T, Kihara M, Kobayashi K, Miya A. Incidences of Unfavorable Events in the Management of Low-Risk Papillary Microcarcinoma of the Thyroid by Active Surveillance Versus Immediate Surgery. Thyroid 2016; 26:150-155
  5. Davies L, Welch HG. Increasing incidence of thyroid cancer in the United States, 1973-2002. JAMA 2006; 295:2164-2167
  6. Cairong Zhu, Tongzhang Zheng, Briseis A. Kilfoy, Xuesong Han, Shuangge Ma, Yue Ba, Yana Bai, Rong Wang, Yong Zhu, Zhang Y. A Birth Cohort Analysis of the Incidence of Papillary Thyroid Cancer in the United States, 1973–2004. Thyroid 2009; 19:1061-1066
  7. Udelsman R, Zhang Y. The epidemic of thyroid cancer in the United States: the role of endocrinologists and ultrasounds. Thyroid 2014; 24:472-479
  8. Vaccarella S, Franceschi S, Bray F, Wild CP, Plummer M, Dal Maso L. Worldwide Thyroid-Cancer Epidemic? The Increasing Impact of Overdiagnosis. N Engl J Med 2016; 375:614-617
  9. Hundahl SA, Cady B, Cunningham MP, Mazzaferri E, McKee RF, Rosai J, Shah JP, Fremgen AM, Stewart AK, Holzer S. Initial results from a prospective cohort study of 5583 cases of thyroid carcinoma treated in the united states during 1996. U.S. and German Thyroid Cancer Study Group. An American College of Surgeons Commission on Cancer Patient Care Evaluation study. Cancer 2000; 89:202-217
  10. Mazzaferri EL, Kloos RT. Current Approaches to Primary Therapy for Papillary and Follicular Thyroid Cancer. J Clin Endocrinol Metab 2001; 86:1447-1463
  11. Hay ID, Thompson GB, Grant CS, Bergstralh EJ, Dvorak CE, Gorman CA, Maurer MS, McIver B, Mullan BP, Oberg AL, Powell CC, van Heerden JA, Goellner JR. Papillary thyroid carcinoma managed at the Mayo Clinic during six decades (1940-1999): temporal trends in initial therapy and long-term outcome in 2444 consecutively treated patients. World J Surg 2002; 26:879-885
  12. Pitoia F, Bueno F, Urciuoli C, Abelleira E, Cross G, Tuttle RM. Outcomes of patients with differentiated thyroid cancer risk-stratified according to the american thyroid association and latin american thyroid society risk of recurrence classification systems. Thyroid 2013; 23:1401-1407
  13. Ashcraft MW, Van Herle AJ. The comparative value of serum thyroglobulin measurements and iodine 131 total body scans in the follow-up of patients with treated differentiated thyroid cancer. Am J Med 1981; 71:806-814
  14. Pineda JD, Lee T, Ain K, Reynolds JC, Robbins J. Iodine-131 therapy for thyroid cancer patients with elevated thyroglobulin and negative diagnostic sca. J Clin Endocrinol Metab 1995; 80:1488-1492
  15. Baudin E, Do Cao C, Cailleux AF, Leboulleux S, Travagli JP, Schlumberger M. Positive predictive value of serum thyroglobulin levels, measured during the first year of follow-up after thyroid hormone withdrawal, in thyroid cancer patients. J Clin Endocrinol Metab 2003; 88:1107-1111
  16. Smallridge RC, Diehl N, Bernet V. Practice trends in patients with persistent detectable thyroglobulin and negative diagnostic radioiodine whole body scans: a survey of American Thyroid Association members. Thyroid 2014; 24:1501-1507
  17. Lamartina L, Durante C, Filetti S, Cooper DS. Low-risk differentiated thyroid cancer and radioiodine remnant ablation: a systematic review of the literature. J Clin Endocrinol Metab 2015; 100:1748-1761
  18. Giovanella L, Imperiali M, Ferrari A, Palumbo A, Furlani L, Graziani MS, Castello R. Serum thyroglobulin reference values according to NACB criteria in healthy subjects with normal thyroid ultrasound. Clin Chem Lab Med 2012; 50:891-893
  19. Djemli A, Van Vliet G, Belgoudi J, Lambert M, Delvin EE. Reference intervals for free thyroxine, total triiodothyronine, thyrotropin and thyroglobulin for Quebec newborns, children and teenagers. Clin Biochem 2004; 37:328-330
  20. Sobrero G, Munoz L, Bazzara L, Martin S, Silvano L, Iorkansky S, Bergoglio L, Spencer C, Miras M. Thyroglobulin reference values in a pediatric infant population. Thyroid 2007; 17:1049-1054
  21. Unger J, De Maertelaer V, Golstein J, Decoster C, Jonckheer MH. Relationship between serum thyroglobulin and intrathyroidal stable iodine in human simple goiter. Clin Endocrinol 1985; 23:1-6
  22. Grebe SK. Soluble thyroid tumor markers – old and new challenges and potential solutions. NZ J Med Lab Science 2013:76-87
  23. Shih ML, Lee JA, Hsieh CB, Yu JC, Liu HD, Kebebew E, Clark OH, Duh QY. Thyroidectomy for Hashimoto's thyroiditis: complications and associated cance. Thyroid 2008; 18:729-734
  24. Hrafnkelsson J, Tulinius H, Kjeld M, Sigvaldason H, Jonasson JG. Serum thyroglobulin as a risk factor for thyroid carcinoma. Acta Oncol 2000; 39:973-977
  25. Sands NB, Karls S, Rivera J, Tamilia M, Hier MP, Black MJ, Gologan O, Payne RJ. Preoperative serum thyroglobulin as an adjunct to fine-needle aspiration in predicting well-differentiated thyroid cancer. J Otolaryngol Head Neck Surg 2010; 39:669-673
  26. Petric R, Perhavec A, Gazic B, Besic N. Preoperative serum thyroglobulin concentration is an independent predictive factor of malignancy in follicular neoplasms of the thyroid gland. J Surg Oncol 2012; 105:351-356
  27. Lee EK, Chung KW, Min HS, Kim TS, Kim TH, Ryu JS, Jung YS, Kim SK, Lee YJ. Preoperative serum thyroglobulin as a useful predictive marker to differentiate follicular thyroid cancer from benign nodules in indeterminate nodules. J Korean Med Sci 2012; 27:1014-1018
  28. Rinaldi S, Plummer M, Biessy C, Tsilidis KK, Ostergaard JN, Overvad K, Tjonneland A, Halkjaer J, Boutron-Ruault MC, Clavel-Chapelon F, Dossus L, Kaaks R, Lukanova A, Boeing H, Trichopoulou A, Lagiou P, Trichopoulos D, Palli D, Agnoli C, Tumino R, Vineis P, Panico S, Bueno-de-Mesquita HB, Peeters PH, Weiderpass E, Lund E, Quiros JR, Agudo A, Molina E, Larranaga N, Navarro C, Ardanaz E, Manjer J, Almquist M, Sandstrom M, Hennings J, Khaw KT, Schmidt J, Travis RC, Byrnes G, Scalbert A, Romieu I, Gunter M, Riboli E, Franceschi S. Thyroid-stimulating hormone, thyroglobulin, and thyroid hormones and risk of differentiated thyroid carcinoma: the EPIC study. Journal of the National Cancer Institute 2014; 106:dju097
  29. Scheffler P, Forest VI, Leboeuf R, Florea AV, Tamilia M, Sands NB, Hier MP, Mlynarek AM, Payne RJ. Serum Thyroglobulin Improves the Sensitivity of the McGill Thyroid Nodule Score for Well-Differentiated Thyroid Cancer. Thyroid 2014; 24:852-857
  30. Trimboli P, Treglia G, Giovanella L. Preoperative measurement of serum thyroglobulin to predict malignancy in thyroid nodules: a systematic review. Horm Metab Res 2015; 47:247-252
  31. Ericsson UB, Tegler L, Lennquist S, Christensen SB, Ståhl E, Thorell JI. Serum thyroglobulin in differentiated thyroid carcinoma. Acta Chir Scand 1984; 150:367-375
  32. Durante C, Puxeddu E, Ferretti E, Morisi R, Moretti S, Bruno R, Barbi F, Avenia N, Scipioni A, Verrienti A, Tosi E, Cavaliere A, Gulino A, Filetti S, Russo D. BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism. J Clin Endocrinol Metab 2007; 92:2840-2843
  33. Gibelli B, Tredici P, De Cicco C, Bodei L, Sandri MT, Renne G, Bruschini R, Tradati N. Preoperative determination of serum thyroglobulin to identify patients with differentiated thyroid cancer who may present recurrence without increased thyroglobulin. Acta Otorhinolaryngol Ital 2005; 25:94-99
  34. Giovanella L, Ceriani L, Ghelfo A, Maffioli M, Keller F. Preoperative undetectable serum thyroglobulin in differentiated thyroid carcinoma: incidence, causes and management strategy. Clin Endocrinol 2007; 67:547-551
  35. Lazar V, Bidart JM, Caillou B, Mahe C, Lacroix L, Filetti S, Schlumberger M. Expression of the Na+/I- symporter gene in human thyroid tumors: a comparison study with other thyroid-specific genes. J Clin Endocrinol Metab 1999; 84:3228-3234
  36. Ronga G, Filesi M, Ventroni G, Vestri AR, Signore A. Value of the first serum thyroglobulin level after total thyroidectomy for the diagnosis of metastases from differentiated thyroid carcinoma. Eur J Nucl Med 1999; 26:1448-1452
  37. Lima N, Cavaliere E, Tomimori E, Knobel M, Medeieros-Neto G. Prognostic value of serial serum thyroglobulin determinations after total thyroidectomy for differentiated thyroid cancer. J Endocrinol Invest 2002; 25:110-115
  38. Lin JD, Huang MJ, Hsu BR, Chao TC, Hsueh C, Liu FH, Liou MJ, Weng HF. Significance of postoperative serum thyroglobulin levels in patients with papillary and follicular thyroid carcinomas. J Surg Oncol 2002; 80:45-51
  39. Hall FT, Beasley NJ, Eski SJ, Witterick IJ, Walfish PG, Freeman JL. Predictive value of serum thyroglobulin after surgery for thyroid carcinoma. Laryngoscope 2003; 113:77-81
  40. Toubeau M, Touzery C, Arveux P, Chaplain G, Vaillant G, Berriolo A, Riedinger JM, Boichot C, Cochet A, Brunotte F. Predictive value for disease progression of serum thyroglobulin levels measured in the postoperative period and after (131)I ablation therapy in patients with differentiated thyroid cancer. J Nucl Med 2004; 45:988-994
  41. Kim TY, Kim WB, Kim ES, Ryu JS, Yeo JS, Kim SC, Hong SJ, Shong YK. Serum thyroglobulin levels at the time of 131I remnant ablation just after thyroidectomy are useful for early prediction of clinical recurrence in low-risk patients with differentiated thyroid carcinoma. J Clin Endocrinol Metab 2005; 90:1440-1445
  42. Makarewicz J, Adamczewski Z, Knapska-Kucharska M, Lewiński A. Evaluation of the diagnostic value of the first thyroglobulin determination in detecting metastases after differentiated thyroid carcinoma surgery. Exp Clin Endocrinol Diabetes 2006; 114:485-489
  43. Heemstra KA, Liu YY, Stokkel M, Kievit J, Corssmit E, Pereira AM, Romijn JA, Smit JW. Serum thyroglobulin concentrations predict disease-free remission and death in differentiated thyroid carcinoma. Clin Endocrinol (Oxf) 2007; 66:58-64
  44. Giovanella L, Ceriani L, Suriano S, Ghelfo A, Maffioli M. Thyroglobulin measurement before rhTSH-aided (131)I ablation in detecting metastases from differentiated thyroid carcinoma. Clin Endocrinol (Oxf) 2008; 68:659-663
  45. Feldt-Rasmussen U, Petersen PH, Nielsen H, Date J. Thyroglobulin of varying molecular sizes with different disappearence rates in plasma following subtotal thyroidectomy. Clin Endocrinol (Oxf) 1978; 9:205-214
  46. Padovani RP, Robenshtok E, Brokhin M, Tuttle RM. Even without additional therapy, serum thyroglobulin concentrations often decline for years after total thyroidectomy and radioactive remnant ablation in patients with differentiated thyroid cancer. Thyroid 2012; 22:778-783
  47. Durante C, Montesano T, Attard M, Torlontano M, Monzani F, Costante G, Meringolo D, Ferdeghini M, Tumino S, Lamartina L, Paciaroni A, Massa M, Giacomelli L, Ronga G, Filetti S. Long-Term Surveillance of Papillary Thyroid Cancer Patients Who Do Not Undergo Postoperative Radioiodine Remnant Ablation: Is There a Role for Serum Thyroglobulin Measurement? J Clin Endocrinol Metab 2012; 97:2748-2753
  48. Pacini F, Agate L, Elisei R, Capezzone M, Ceccarelli C, Lippi F, Molinaro E, Pinchera A. Outcome of differentiated thyroid cancer with detectable serum Tg and negative diagnostic (131)I whole body scan: comparison of patients treated with high (131)I activities versus untreated patients. J Clin Endocrinol Metab 2001; 86:4092-4097
  49. Schaap J, Eustatia-Rutten CF, Stokkel M, Links TP, Diamant M, van der Velde EA, Romijn JA, Smit JW. Does radioiodine therapy have disadvantageous effects in non-iodine accumulating differentiated thyroid carcinoma. Clin Endocrinol (Oxf) 2002; 57:117-124
  50. Valadão MM, Rosário PW, Borges MA, Costa GB, Rezende LL, Padrão EL, Barroso AL, Purisch S. Positive predictive value of detectable stimulated tg during the first year after therapy of thyroid cancer and the value of comparison with Tg-ablation and Tg measured after 24 months. Thyroid 2006; 16:1145-1149
  51. Rosario P, Borges M, Reis J, Alves MF. Effect of suppressive therapy with levothyroxine on the reduction of serum thyroglobulin after total thyroidectomy. Thyroid 2006; 16:199-200
  52. Huang SH, Wang PW, Huang YE, Chou FF, Liu RT, Tung SC, Chen JF, Kuo MC, Hsieh JR, Hsieh HH. Sequential follow-up of serum thyroglobulin and whole body scan in thyroid cancer patients without initial metastasis. Thyroid 2006; 16:1273-1278
  53. Miyauchi A, Kudo T, Miya A, Kobayashi K, Ito Y, Takamura Y, Higashiyama T, Fukushima M, Kihara M, Inoue H, Tomoda C, Yabuta T, Masuoka H. Prognostic impact of serum thyroglobulin doubling-time under thyrotropin suppression in patients with papillary thyroid carcinoma who underwent total thyroidectomy. Thyroid 2011; 21:707-716
  54. Pacini F, Sabra MM, Tuttle RM. Clinical relevance of thyroglobulin doubling time in the management of patients with differentiated thyroid cancer. Thyroid 2011; 21:691-692
  55. Giovanella L, Trimboli P, Verburg FA, Treglia G, Piccardo A, Foppiani L, Ceriani L. Thyroglobulin levels and thyroglobulin doubling time independently predict a positive 18F-FDG PET/CT scan in patients with biochemical recurrence of differentiated thyroid carcinoma. Eur J Nucl Med Mol Imaging 2013; 40:874-880
  56. Miyauchi A, Kudo T, Kihara M, Higashiyama T, Ito Y, Kobayashi K, Miya A. Relationship of biochemically persistent disease and thyroglobulin-doubling time to age at surgery in patients with papillary thyroid carcinoma. Endocr J 2013; 60:415-421
  57. Yim JH, Kim EY, Bae Kim W, Kim WG, Kim TY, Ryu JS, Gong G, Hong SJ, Yoon JH, Shong YK. Long-term consequence of elevated thyroglobulin in differentiated thyroid cancer. Thyroid 2013; 23:58-63
  58. Elisei R, Agate L, Viola D, Matrone A, Biagini A, Molinaro E. How to manage patients with differentiated thyroid cancer and a rising serum thyroglobulin level. Endocrinol Metab Clin North Am 2014; 43:331-344
  59. Kelders A, Kennes LN, Krohn T, Behrendt FF, Mottaghy FM, Verburg FA. Relationship between positive thyroglobulin doubling time and 18F-FDG PET/CT-positive, 131I-negative lesions. Nucl Med Commun 2014; 34:176-181
  60. Rossing RM, Jentzen W, Nagarajah J, Bockisch A, Gorges R. Serum Thyroglobulin Doubling Time in Progressive Thyroid Cancer. Thyroid 2016; 26:1712-1718
  61. Schlumberger M CP, Fragu P, Lumbroso J, Parmentier C and Tubiana M,. Circulating thyrotropin and thyroid hormones in patients with metastases of differentiated thyroid carcinoma: relationship to serum thyrotropin levels. J Clin Endocrinol Metab 1980; 51:513-519
  62. Robbins RJ, Srivastava S, Shaha A, Ghossein R, Larson SM, Fleisher M, Tuttle RM. Factors influencing the basal and recombinant human thyrotropin-stimulated serum thyroglobulin in patients with metastatic thyroid carcinoma. J Clin Endocrinol Metab 2004; 89:6010-6016
  63. Spencer CA, LoPresti JS, Fatemi S, Nicoloff JT. Detection of residual and recurrent differentiated thyroid carcinoma by serum Thyroglobulin measurement. Thyroid 1999; 9:435-441
  64. Mazzaferri EL, Robbins RJ, Spencer CA, Braverman LE, Pacini F, Wartofsky L, Haugen BR, Sherman SI, Cooper DS, Braunstein GD, Lee S, Davies TF, Arafah BM, Ladenson PW, Pinchera A. A consensus report of the role of serum thyroglobulin as a monitoring method for low-risk patients with papillary thyroid carcinoma. J Clin Endocrinol Metab 2003; 88:1433-1441
  65. Mazzaferri EL, Kloos RT. Is diagnostic iodine-131 scanning with recombinant human TSH useful in the follow-up of differentiated thyroid cancer after thyroid ablation? J Clin Endocrinol Metab 2002; 87:1486-1489
  66. Vitale G, Lupoli GA, Ciccarelli A, Lucariello A, Fittipaldi MR, Fonderico F, Panico A, Lupoli G. Influence of body surface area on serum peak thyrotropin (TSH) levels after recombinant human TSH administration. J Clin Endocrinol Metab 2003; 88:1319-1322
  67. Montesano T, Durante C, Attard M, Crocetti U, Meringolo D, Bruno R, Tumino S, Rubello D, Al-Nahhas A, Colandrea M, Maranghi M, Travascio L, Ronga G, Torlontano M. Age influences TSH serum levels after withdrawal of l-thyroxine or rhTSH stimulation in patients affected by differentiated thyroid cancer. Biomed Pharmacother 2007; 61:468-471
  68. Braverman L, Kloos RT, Law B Jr, Kipnes M, Dionne M, Magner J. Evaluation of various doses of recombinant human thyrotropin in patients with multinodular goiters. Endoc Pract 2008; 14:832-839
  69. Over R, Nsouli-Maktabi H, Burman KD, Jonklaas J. Age modifies the response to recombinant human thyrotropin. Thyroid 2010; 20:1377-1384
  70. Schlumberger M, Charbord P, Fragu P, Lumbroso J, C P, Tubiana M. Circulating thyrotropin and thyroid hormones in patients with metastases of differentiated thyroid carcinoma: relationship to serum thyrotropin levels. J Clin Endocrinol Metab 1980; 51:513-519
  71. Nakabashi CC, Kasamatsu TS, Crispim F, Yamazaki CA, Camacho CP, Andreoni DM, Padovani RP, Ikejiri ES, Mamone MC, Aldighieri FC, Wagner J, Hidal JT, Vieira JG, Biscolla RP, Maciel RM. Basal serum thyroglobulin measured by a second-generation assay is equivalent to stimulated thyroglobulin in identifying metastases in patients with differentiated thyroid cancer with low or intermediate risk of recurrence. Eur Thyroid J 2014; 3:43-50
  72. Groen AH, Klein Hesselink MS, Plukker JT, Sluiter WJ, van der Horst-Schrivers AN, Brouwers AH, Lentjes EG, Muller Kobold AC, Links TP. Additional value of a high sensitive thyroglobulin assay in the follow-up of patients with differentiated thyroid carcinoma. Clin Endocrinol (Oxf) 2016;
  73. Rotman-Pikielny P, Reynolds JC, Barker WC, Yen PM, Skarulis MC, Sarlis NJ. Recombinant human thyrotropin for the diagnosis and treatment of a highly functional metastatic struma ovarii. J Clin Endocrinol Metab 2000; 85:237-244
  74. Russo M, Marturano I, Masucci R, Caruso M, Fornito MC, Tumino D, Tavarelli M, Squatrito S, Pellegriti G. Metastatic malignant struma ovarii with coexistence of Hashimoto's thyroiditis. Endocrinology, diabetes & metabolism case reports 2016:160030
  75. Trimboli P, D'Aurizio F, Tozzoli R, Giovanella L. Measurement of thyroglobulin, calcitonin, and PTH in FNA washout fluids. Clin Chem Lab Med 2016;
  76. Machens A, Holzhausen HJ, Dralle H. The prognostic value of primary tumor size in papillary and follicular thyroid carcinoma. Cancer 2005; 103:2269-2273
  77. Passler C, Scheuba C, Prager G, Kaczirek K, Kaserer K, Zettinig G, Niederle B. Prognostic factors of papillary and follicular thyroid cancer: differences in an iodine-replete endemic goiter region. Endocr Relat Cancer 2004; 11:131-139
  78. Randolph GW, Duh QY, Heller KS, LiVolsi VA, Mandel SJ, Steward DL, Tufano RP, Tuttle RM. The prognostic significance of nodal metastases from papillary thyroid carcinoma can be stratified based on the size and number of metastatic lymph nodes, as well as the presence of extranodal extension. Thyroid 2012; 22:1144-1152
  79. Pezzi TA, Sandulache VC, Pezzi CM, Turkeltaub AE, Feng L, Cabanillas ME, Williams MD, Lai SY. Treatment and survival of patients with insular thyroid carcinoma: 508 cases from the National Cancer Data Base. Head Neck 2016; 38:906-912
  80. Rosario PW, de Faria S, Bicalho L, Alves MF, Borges MA, Purisch S, Padrão EL, Rezende LL, Barroso AL. Ultrasonographic differentiation between metastatic and benign lymph nodes in patients with papillary thyroid carcinoma. J Ultrasound Med 2005; 24:1385-1389
  81. Pacini F, Fugazzola L, Lippi F, Ceccarelli C, Centoni R, Miccoli P, Elisei R, Pinchera A. Detection of thyroglobulin in fine needle aspirates of nonthyroidal neck masses: a clue to the diagnosis of metastatic differentiated thyroid cancer. J Clin Endocrinol Metab 1992; 74:1401-1404
  82. Uruno T, Miyauchi A, Shimizu K, Tomoda C, Takamura Y, Ito Y, Miya A, Kobayashi K, Matsuzuka F, Amino N, Kuma K. Usefulness of thyroglobulin measurement in fine-needle aspiration biopsy specimens for diagnosing cervical lymph node metastasis in patients with papillary thyroid cancer. World J Surg 2005; 29:483-485
  83. Boi F, Baghino G, Atzeni F, Lai ML, Faa G, Mariotti S. The diagnostic value for differentiated thyroid carcinoma metastases of thyroglobulin (Tg) measurement in washout fluid from fine-needle aspiration biopsy of neck lymph nodes is maintained in the presence of circulating anti-Tg antibodies. J Clin Endocrinol Metab 2006; 91:1364-1369
  84. Snozek CL, Chambers EP, Reading CC, Sebo TJ, Sistrunk JW, Singh RJ, Grebe SK. Serum thyroglobulin, high-resolution ultrasound, and lymph node thyroglobulin in diagnosis of differentiated thyroid carcinoma nodal metastases. J Clin Endocrinol Metab 2007; 92:4278-4281
  85. Bruno R, Giannasio P, Chiarella R, Capula C, Russo D, Filetti S, Costante G. Identification of a neck lump as a lymph node metastasis from an occult contralateral papillary microcarcinoma of the thyroid: key role of thyroglobulin assay in the fine-needle aspirate. Thyroid 2009; 19:531-533
  86. Jeon SJ, Kim E, Park JS, Son KR, Baek JH, Kim YS, Park do J, Cho BY, Na DG. Diagnostic benefit of thyroglobulin measurement in fine-needle aspiration for diagnosing metastatic cervical lymph nodes from papillary thyroid cancer: correlations with US features. Korean J Radiol 2009; 10:106-111
  87. Cunha N, Rodrigues F, Curado F, Ilhéu O, Cruz C, Naidenov P, Rascão MJ, Ganho J, Gomes I, Pereira H, Real O, Figueiredo P, Campos B, Valido F. Thyroglobulin detection in fine-needle aspirates of cervical lymph nodes: a technique for the diagnosis of metastatic differentiated thyroid cancer. Eur J Endocrinol 2007; 157:101-107
  88. Suh YJ, Son EJ, Moon HJ, Kim EK, Han KH, Kwak JY. Utility of Thyroglobulin Measurements in Fine-needle Aspirates of Space Occupying Lesions in the Thyroid Bed after Thyroid Cancer Operations. Thyroid 2012; 23:in press
  89. Cappelli C, Pirola I, De Martino E, Gandossi E, Cimino E, Samoni F, Agosti B, Rosei EA, Casella C, Castellano M. Thyroglobulin measurement in fine-needle aspiration biopsy of metastatic lymph nodes after rhTSH stimulation. Head Neck 2013; 35:E21-23
  90. Grani G, Fumarola A. Thyroglobulin in Lymph Node Fine-Needle Aspiration Washout: A Systematic Review and Meta-analysis of Diagnostic Accuracy. J Clin Endocrinol Metab 2014; 99:1970-1982
  91. Torres MR, Nóbrega Neto SH, Rosas RJ, Martins AL, Ramos AL, da Cruz TR. Thyroglobulin in the washout fluid of lymph-node biopsy: what is its role in the follow-up of differentiated thyroid carcinoma? Thyroid 2014; 24:7-18
  92. Chung J, Kim EK, Lim H, Son EJ, Yoon JH, Youk JH, Kim JA, Moon HJ, Kwak JY. Optimal indication of thyroglobulin measurement in fine-needle aspiration for detecting lateral metastatic lymph nodes in patients with papillary thyroid carcinoma. Head Neck 2014; 36:795-801
  93. Tang S, Buck A, Jones C, Sara Jiang X. The utility of thyroglobulin washout studies in predicting cervical lymph node metastases: One academic medical center's experience. Diagn Cytopathol 2016;
  94. Jeon MJ, Kim WG, Jang EK, Choi YM, Lee YM, Sung TY, Yoon JH, Chung KW, Hong SJ, Baek JH, Lee JH, Kim TY, Shong YK, Kim WB. Thyroglobulin level in fine-needle aspirates for preoperative diagnosis of cervical lymph node metastasis in patients with papillary thyroid carcinoma: two different cutoff values according to serum thyroglobulin level. Thyroid 2015; 25:410-416
  95. Zanella AB, Meyer EL, Balzan L, Silva AC, Camargo J, Migliavacca A, Guimaraes JR, Maia AL. Thyroglobulin measurements in washout of fine needle aspirates in cervical lymph nodes for detection of papillary thyroid cancer metastases. Arq Bras Endocrinol Metabol 2010; 54:550-554
  96. Baskin HJ. Detection of recurrent papillary thyroid carcinoma by thyroglobulin assessment in the needle washout after fine-needle aspiration of suspicious lymph nodes. Thyroid 2004; 14:959-963
  97. Shin HJ, Lee HS, Kim EK, Moon HJ, Lee JH, Kwak JY. A Study on Serum Antithyroglobulin Antibodies Interference in Thyroglobulin Measurement in Fine-Needle Aspiration for Diagnosing Lymph Node Metastasis in Postoperative Patients. PLoS One 2015; 10:e0131096
  98. Boi F, Maurelli I, Pinna G, Atzeni F, Piga M, Lai ML, Mariotti S. Calcitonin measurement in wash-out fluid from fine needle aspiration of neck masses in patients with primary and metastatic medullary thyroid carcinoma. J Clin Endocrinol Metab 2007; 92:2115-2118
  99. Abraham D, Gault PM, Hunt J, Bentz J. Calcitonin estimation in neck lymph node fine-needle aspirate fluid prevents misinterpretation of cytology in patients with metastatic medullary thyroid cancer. Thyroid 2009; 19:1015-1016
  100. Massaro F, Dolcino M, Degrandi R, Ferone D, Mussap M, Minuto F, Giusti M. Calcitonin assay in wash-out fluid after fine-needle aspiration biopsy in patients with a thyroid nodule and border-line value of the hormone. J Endocrinol Invest 2009; 32:308-312
  101. Barzon L, Boscaro M, Pacenti M, Taccaliti A, Palù G. Evaluation of circulating thyroid-specific transcripts as markers of thyroid cancer relapse. Int J Cancer 2004; 110:914-920
  102. Verburg FA, Lips CJ, Lentjes EG, Klerk Jd J. Detection of circulating Tg-mRNA in the follow-up of papillary and follicular thyroid cancer: how useful is it? Br J Cancer 2004; 91:1-5
  103. Gupta M, Chia SY. Circulating thyroid cancer markers. Curr Opin Endocrinol Diabetes Obes 2007; 14:383-388
  104. Ringel MD, Ladenson PW, Levine MA. Molecular diagnosis of residual and recurrent thyroid cancer by amplification of thyroglobulin messenger ribonucleic acid in peripheral blood. J Clin Endocrinol Metab 1998; 83:4435-4442
  105. Ausavarat S, Sriprapaporn J, Satayaban B, Thongnoppakhun W, Laipiriyakun A, Amornkitticharoen B, Chanachai R, Pattanachak C. Circulating thyrotropin receptor messenger ribonucleic acid is not an effective marker in the follow-up of differentiated thyroid carcinoma. Thyroid research 2015; 8:11
  106. Biscolla RP, Cerutti JM, Maciel RM. Detection of recurrent thyroid cancer by sensitive nested reverse transcription-polymerase chain reaction of thyroglobulin and sodium/iodide symporter messenger ribonucleic acid transcripts in peripheral blood. J Clin Endocrinol Metab 2000; 85:3623-3627
  107. Chinnappa P, Taguba L, Arciaga R, Faiman C, Siperstein A, Mehta AE, Reddy SK, Nasr C, Gupta MK. Detection of thyrotropin-receptor messenger ribonucleic acid (mRNA) and thyroglobulin mRNA transcripts in peripheral blood of patients with thyroid disease: sensitive and specific markers for thyroid cancer. J Clin Endocrinol Metab 2004; 89:3705-3709
  108. Barbosa GF, Milas M. Peripheral thyrotropin receptor mRNA as a novel marker for differentiated thyroid cancer diagnosis and surveillance. Expert Rev Anticancer Ther 2008; 8:1415-1424
  109. Milas M, Shin J, Gupta M, Novosel T, Nasr C, Brainard J, Mitchell J, Berber E, Siperstein A. Circulating thyrotropin receptor mRNA as a novel marker of thyroid cancer: clinical applications learned from 1758 samples. Ann Surg 2010; 252:643-651
  110. Ringel MD. Editorial: molecular detection of thyroid cancer: differentiating "signal" and "noise" in clinical assays. J Clin Endocrinol Metab 2004; 89:29-32
  111. Chia SY, Milas M, Reddy SK, Siperstein A, Skugor M, Brainard J, Gupta MK. Thyroid-stimulating hormone receptor messenger ribonucleic acid measurement in blood as a marker for circulating thyroid cancer cells and its role in the preoperative diagnosis of thyroid cancer. J Clin Endocrinol Metab 2007; 92:468-475
  112. Kaufmann S, Schmutzler C, Schomburg L, Körber C, Luster M, Rendl J, Reiners C, Köhrle J. Real time RT-PCR analysis of thyroglobulin mRNA in peripheral blood in patients with congenital athyreosis and with differentiated thyroid carcinoma after stimulation with recombinant human thyrotropin. Endocr Regul 2004; 38:41-49
  113. Chelly J, Concordet JP, Kaplan JC, Kahn A. Illegitimate transcription: transcription of any gene in any cell type. Proc Natl Acad Sci USA 1989; 86:2617-2621
  114. Ghossein RA, Bhattacharya S. Molecular detection and characterisation of circulating tumour cells and micrometastases in solid tumours. Eur J Cancer 2000; 36:1681-1694
  115. Savagner F, Rodien P, Reynier P, Rohmer V, Bigorgne JC, Malthiery Y. Analysis of Tg transcripts by real-time RT-PCR in the blood of thyroid cancer patients. J Clin Endocrinol Metab 2002; 87:635-639
  116. Elisei R, Vivaldi A, Agate L, Molinaro E, Nencetti C, Grasso L, Pinchera A, Pacini F. Low specificity of blood thyroglobulin messenger ribonucleic acid assay prevents its use in the follow-up of differentiated thyroid cancer patients. J Clin Endocrinol Metab 2004; 89:33-39
  117. Denizot A, Delfino C, Dutour-Meyer A, Fina F, Ouafik L. Evaluation of quantitative measurement of thyroglobulin mRNA in the follow-up of differentiated thyroid cancer. Thyroid 2003; 13:867-872
  118. Amakawa M, Kato R, Kameko F, Maruyama M, Tajiri J. Thyroglobulin mRNA expression in peripheral blood lymphocytes of healthy subjects and patients with thyroid disease. Clin Chim Acta 2008; 390:97-103
  119. Sellitti DF, Akamizu T, Doi SQ, Kim GH, Kariyil JT, Kopchik JJ, Koshiyama H. Renal expression of two 'thyroid-specific' genes: thyrotropin receptor and thyroglobulin. Exp Nephrol 2000; 8:235-243
  120. Bojunga J, Roddiger S, Stanisch M, Kusterer K, Kurek R, Renneberg H, Adams S, Lindhorst E, Usadel KH, Schumm-Draeger PM. Molecular detection of thyroglobulin mRNA transcripts in peripheral blood of patients with thyroid disease by RT-PCR. Br J Cancer 2000; 82:1650-1655
  121. Endo T, Kobayashi T. Thyroid-stimulating hormone receptor in brown adipose tissue is involved in the regulation of thermogenesis. Am J Physiol Endocrinol Metab 2008; 295:E514-518
  122. Zhang W, Tian LM, Han Y, Ma HY, Wang LC, Guo J, Gao L, Zhao JJ. Presence of thyrotropin receptor in hepatocytes: not a case of illegitimate transcription. J Cell Mol Med 2009; 13:4636-4642
  123. Cianfarani F, Baldini E, Cavalli A, Marchioni E, Lembo L, Teson M, Persechino S, Zambruno G, Ulisse S, Odorisio T, D'Armiento M. TSH receptor and thyroid-specific gene expression in human skin. J Invest Dermatol 2010; 130:93-101

 

Pathology And Pathogenesis Of Pituitary Adenomas And Other Sellar Lesions

ABSTRACT

The pituitary gland, or hypophysis, and adjacent structures of the sellar region can be affected by a wide range of pathologies leading to endocrine and neurological disorders. These include neoplasms arising from the adenohypophysis, such as pituitary adenomas associated with distinctive endocrine disorders such as acromegaly or Cushing’s disease; cysts or neoplasms derived from remnants of Rathke’s pouch (Rathke’s cleft cyst, craniopharyngioma); tumours of the neurohypophysis and pituitary stalk (pituicytoma, granular cell tumour) and neoplasms of the parasellar bone (chordoma). Further, conditions like lymphocytic or granulomatous hypophysitis may mimic pituitary neoplasms. Here, we provide an overview of the molecular pathogenesis and neuropathological features of these common lesions. For complete coverage of this and related areas of Endocrinology, please visit our free web –book, www.endotext.org.

PITUITARY ADENOMAS

Definition

Pituitary adenomas are benign clonal neoplasms of the neuroendocrine epithelial cells of the adenohypophysis.

General Features

Pituitary adenomas share characteristics with other adenomas of endocrine glands: granular cytoplasm, round nuclei with finely dispersed chromatin and multiple distinct nucleoli; they generally also express both markers of neurosecretory granules (synaptophysin, chromogranin) as well as epithelial differentiation (cytokeratins). However, they may present with a wide range of morphological features depending on hormonal or genetic subtype, or as a result of treatment effect. Although called benign, pituitary adenomas can be locally invasive and destructive, or clinically malignant due to the metabolic consequences of excess hormone secretion.

Pituitary adenomas are common intracranial neoplasms and may be clinically silent, detected incidentally on MRI scans of the brain (~ 22%), or found at autopsy (~ 14%)(1). It has been estimated that they represent approximately 25% of all clinically manifest intracranial neoplasms.

WHO Classification

Pituitary adenomas can be classified in various ways, according to size, clinically functional or silent manifestation, hormone or cytokeratin expression profile, defining somatic mutations, and histologic features. The 2004 edition of the WHO classification of endocrine tumours uses markers of cytodifferentiation as the principal classifier. In addition to the category of ‘typical pituitary adenoma’ and ‘pituitary carcinoma’, it also introduced the concept of ‘atypical pituitary adenoma’. However, the latter is controversial (2), as the criteria are to some degree subjective and the clinical significance of ‘atypia’ as currently defined remains to be determined in longitudinal studies (3). The current classification is summarised in Table 3b.1

Table 3b.1 Classification of pituitary adenomas (Adapted from reference (4))

Adenoma type Transcription Factors Hormones Cytokeratin
GH-producing adenomas      
Densely granulated somatotroph adenoma Pit-1 GH, a-SU diffuse
Sparsely granulated somatotroph adenoma Pit-1 GH dot-like
Mammosomatotroph adenoma Pit-1, ER GH, PRL, a-SU diffuse
Mixed somatotroph and lactotroph andenoma Pit-1, ER GH, PRL, a-SU diffuse
PRL-producing adenomas      
Sparsely granulated lactotroph adenoma Pit-1, ER PRL (Golgi) diffuse
Densely granulated lactotroph adenoma Pit-1, ER PRL (diffuse) diffuse
Acidophil stem-cell adenoma Pit-1, ER PRL (diffuse), GH rare dot-like
TSH-producing adenoma      
Thyrotroph adenoma Pit-1, GATA-2 b-TSH, a-SU diffuse
ACTH-producing adenomas      
Densely granulated corticotroph adenoma Tpit ACTH diffuse
Sparsely granulated corticotroph adenoma Tpit ACTH diffuse
Crooke’s cell adenoma Tpit ACTH ring-like
Gonadotropin-producing adenoma      
Gonadotroph adenoma SF-1, GATA-2, ER b-FSH, b-LH, a-SU diffuse
Plurihormonal adenomas      
Silent type III adenoma Pit-1 (?), ER multiple diffuse
Unusual plurihormonal adenoma (NOS) multiple multiple n/a
Hormone negative adenoma      
Null cell adenoma none none diffuse

Figure 3b-1: Principles of pituitary adenoma classification. Clinical and neuropathological classification schemes vary in their emphasis. This chapter will use a pathological / cell-lineage based approach. Compared with other intracranial neoplasms, molecular genetic, epigenetic or proteomic classification schemes that influence therapeutic decisions are only beginning to emerge.

Gh-Producing Adenoma

Definition

Benign lesions arising from Pit-1 lineage cells of the anterior pituitary that express, store and secrete growth hormone. The classical cause of acromegaly or gigantism.

Pathology

Somatotroph adenomas (SA) occur in the anterior pituitary, arising from growth hormone-producing cells, often in the lateral wings of the gland. They account for 10-15% of pituitary adenomas. T1-weighted MRI imaging shows a sellar structure that is hypointense relative to normal gland. Invasion of the sphenoid or cavernous sinus or suprasellar extension to give the characteristic snowman shape may be seen (5). Lesions are non- or slowly enhancing. Macroscopically, somatotroph adenomas are soft tan-to-grey lesions. Microscopically, somatotroph adenomas occur as two major variants: densely and sparsely granulated.

Densely Granulated Somatotroph Adenoma

Densely granulated somatotroph adenomas (DGSAs) are the most common finding and are composed of large, round, eosinophilic cells with spherical nuclei and prominent nucleoli that closely resemble somatotroph cells. They are diffusely and strongly immunopositive for growth hormone and may also variably express prolactin and less frequently, thyroid-stimulating hormone. Nuclei are strongly immunopositive for Pit1. Ultrastructurally, they contain a well-developed endoplasmic reticulum, a prominent Golgi complex and numerous, large (300-600nm) secretory granules containing growth hormone that are distributed throughout the cytosol. Growth hormone is expressed throughout the lesion. Immunostaining with CAM5.2 antibody against cytokeratin (predominantly cytokeratin 8) reveals a diffuse cytosolic pattern.

Figure 3b-2: Normal anterior gland (NG) and somatotroph adenoma (SOMA) interface. This figure illustrates histological principles of distinction of adenoma from normal gland, which may be difficult on routine HE stains (top left), but is greatly aided by a reticulin stain (top and bottom right). The normal adenohypophysis consists of very well demarcated cell nests separated by dense septa. Bottom left: Serial section to the top row images stained for GH. Dashed line: Border between normal gland and adenoma.

Figure 3b-3: Densely granulated somatotroph adenoma. Densely granulated adenomas consist of monomorphic cells that are eosinophilic on HE stain (top left) and intensely orangeophilic on PAS-OG histochemistry (top right). They show strong, diffuse GH expression (bottom left) and an evenly distributed, cell-membrane-anchored keratin cytoskeleton (bottom right).

Sparsely Granulated Somatotroph Adenoma

Sparsely granulated somatotroph adenomas (SGSAs) are less common and are composed of sheets of poorly cohesive, chromophobic cells often containing eccentric, pleiomorphic nuclei. SGSAs are weakly and focally immunopositive for growth hormone and nuclei are immunopositive for Pit1. They may also variably express prolactin and less frequently, thyroid-stimulating hormone. SGSAs contain dense juxtanuclear deposits of low-molecular weight cytokeratin, termed fibrous bodies that appear as pale spherical inclusions on H&E staining and are strongly immunopositive with CAM5.2 antibodies against cytokeratin (predominantly cytokeratin 8). Ultrastructurally, SGSAs contain few, small (100-250nm) growth-hormone containing granules that align along the plasma membrane. The distribution of cytokeratin and growth hormone-containing granules co-segregate with tumour variant type, so the presence of fibrous bodies is a diagnostic feature of SGSA.

There is a growing body of evidence that DGSAs and SGSAs behave differently with SGSAs being larger, more common in younger, female patients, more proliferative (higher MIB1 indices) and with a greater capacity to invade surrounding structures (6-11). Some studies have found that SGSAs are more poorly responsive to somatostatin treatment than DGSAs (7) although the extent of the impact of tumour subtype on behaviour is unclear.

Figure 3b-4: Sparsely granulated somatotroph adenoma. Sparsely granulated somatotroph adenoma cells are pleomorphic and chromophobe on HE stain (top left) and PAS-OG histochemistry (top right). Fibrous bodies can be seen as pale discs in the cytoplasm on routine stains (arrows). As the name implies, sparsely granulated cells show weak, patchy GH expression (bottom left) and their keratin cytoskeleton is disrupted and condensed into a paranuclear globular structure – the fibrous body (bottom right).

Mixed Pattern Somatotroph Adenoma

SAs that contain cells of both the densely granulated and sparsely granulated type are not uncommon and if more than 30% of cells differ from the predominant cell type, a diagnosis of mixed pattern is required. Very occasionally, SAs that are not immunopositive for low molecular weight cytokeratin are seen although their clinical significance is not known.

Figure 3b-5: Mixed densely and sparsely granulated somatotroph adenoma. A proportion of somatotroph adenomas demonstrate a clearly segregated mixed densely-sparsely phenotype. The respective cells remain true to their ‘pure’ counterparts: The sparsely granulated cells can be identified as chromophobe islands amongst orangeophilic densely granulated cells (left) and their contrasting cytokeratin pattern is absolutely clear following incubation with Cam5.2 antibody (right).

Somatotroph Adenoma With Neuronal Differentiation

A rare but pathologically intriguing subtype of SA, always associated with acromegaly and usually presenting as macroadenoma with or without hypothalamic involvement, shows sparsely granulated GH-producing cells admixed with large atypical ganglion cells. These resemble tumour cells seen in gangliocytomas and represent truly metaplastic tumour cells, as they express a mixture of lineage markers that otherwise are virtually never co-expressed (synaptophysin, neurofilament, cytokeratin and GH). This is of no known clinical relevance and the mechanisms of transdifferentiation remain unexplored.

Figure 3b-6: Sparsely granulated somatotroph adenoma with neuronal metaplasia. Atypical large, neoplastic ganglion cells can be seen in rare sparsely granulated somatotroph adenomas (long arrows). These cells show true metaplasia, expressing GH (top right), neurofilament (bottom left) and cytokeratins (bottom right). Note that both types of intermediate filaments aggregate in fibrous bodies (notched short arrows).

Somatostatin Analogue Effect On Somatotroph Adenomas

Densely granulated somatotroph adenomas tend to respond better to somatostatin analogue treatment than sparsely granulated tumours. This results in a distinct perivascular hyaline / fibrous reaction. The reaction of somatotroph adenomas to somatostatin analogues is morphologically distinct to that of prolactinomas to dopamine agonists (see figure 3b-9 later in this chapter).

Figure 3b-7: Somatostatin analogue effect in densely granulated somatotroph adenomas.
Densely granulated somatotroph adenomas that have responded to somatostatin analogues tend to show perivascular hyaline/fibrous degeneration. Left – untreated, right – treated.

Molecular Genetics

G protein α-subunit

One of the earliest mutations to be associated with sporadic somatotroph adenomas was at the GNAS complex locus. This locus contains four alternative promoters and 5’ exons and has a complex, imprinted expression pattern. Different isoforms of the G protein alpha subunit arise as a result of alternative splicing. The GNAS gene encodes the G protein alpha subunit Gsα, which couples seven-transmembrane receptors to adenylyl cyclase (12). Mutation at either Arg201 or Gln227, destroys GTPase activity (13). Gsα mutation leads to constitutive activation of adenylyl cyclase (termed the gsp oncogene) resulting in increased cAMP synthesis. Mutations in GNAS have been identified in 15-58% of somatotroph adenomas (6,8,11,13-19) (6,8,11,13-19)This mutation may promote tumorigenesis since cAMP can function as a mitogenic signal.

The functional implications of gsp mutation are not determined. Elevated cAMP may be countered by increases in the activity of phosphodiesterase enzymes (PDEs) especially PDE4, which is 7-fold more active in adenomas with a mutation in Gsα. Inhibition of this enzyme resulted in an increase in cellular cAMP (20). However, the effect of Gsα mutation on downstream target genes induced by CREB is uncertain and although increases in the expression of these genes were observed in some cases, they were not consistent (21). No association was observed between mutation in gsp and the granulation pattern of somatotroph adenomas (6,8,22)

Ghrelin And Receptor

There is growing evidence to suggest a role for ghrelin in somatotroph adenomas. Ghrelin (GHS) is a growth hormone secretagogue that acts on the pituitary and has been associated with increased cell migration and proliferation in certain cancers (23,24). Two forms of the ghrelin receptor (GHSR) GHSR1α and the non-functional splice-variant GHSR1β which contains all of exon 1 and some of the following intron are differentially expressed in normal somatotrophs compared to somatotroph adenomas, although there is disagreement concerning which isoform mRNA is more abundant in adenomas compared to normal pituitary (25,26). In different studies, GHSR1α mRNA has been shown to be both reduced (27) and increased (28) in somatotroph adenomas that have a GNAS mutation compared to wild type adenomas. It is unclear whether GHS/GHSR1α. expression is related to adenoma subtype.

Somatostatin receptor

Differing expression of the somatostatin receptor between adenoma subtypes has been observed and this pattern can be influenced by somatostatin analogue (SSA) treatment. A positive correlation has been observed between SSTR2 expression and reduction in GH after SSA treatment (19,29,30). Greater expression of SSTR2 has also been associated with densely granulated adenomas (31), while SSTR5 was associated with sparsely granulated tumours (15,22,30). One study suggests that the proportion of cells expressing SSTR2 is a more reliable indicator of response to SSA than overall expression level (7,32)

Aryl hydrocarbon interacting protein

Mutations in AIP associated with FIPA are covered elsewhere (Section 11a1, Stiles and Korbonits).

Mutations in AIP (aryl hydrocarbon interacting protein) are most frequently associated with somatotroph adenomas. They are generally truncations or nonsense mutations leading to loss of function, which has resulted in the classification of AIP as a tumour suppressor gene, although the mechanism by which it functions is not yet known. Consistent with its purported tumour suppressor role, multiple different mutations are seen in AIP, with some “hotspots” (33-41). Among patients with acromegaly, germline mutation in AIP is rare, but is relatively more common in the young; the reported incidence of AIP mutations in sporadic somatotroph adenomas varies from 4.2% (patients < 40 years) (42) to 5.5-13% (patients <30 years) (43,44). Mutation in AIP in somatotroph adenomas is associated with larger tumours and more invasive behaviour and more recurrences (45). Furthermore patients with AIP mutations are relatively resistant to treatment with somatostatin analogues although the mechanism of this resistance remains to be clarified (35,39). Treatment with SSAs leads to and is associated with upregulation of AIP expression, (34,46). The mechanism for this upregulation is not fully understood, but some authors have proposed that it is ZAC1- (zinc finger regulator of apoptosis and cell cycle arrest) mediated. ZAC1 induces G1 cell cycle arrest and apoptosis (47-49). Low levels of AIP expression have been linked to tumour invasiveness (46) suggesting that patients with AIP mutation require more stringent follow-up.

Gpr101 Mutations And X-Lag

A study of early childhood onset gigantism with growth hormone hypersecretion found heritable microduplications on chromosome Xq26.3. The condition was termed X-LAG or x-linked acrogigantism (50). Analysis of the expression of the genes encoded in this region in a small number of patients showed that GPR101 mRNA was upregulated by up to 1000-fold. In a screen of 248 patients with sporadic acromegaly, there were no microduplications at Xq26.3, but in 11 (4.4%) patients, a mutation in GPR101 (c.924G-C; pE308D) was found that was not present in control samples. In 3 cases, the mutation was also observed in blood and presumed to be germline; in one case, it was a somatic mutation. In a screens of 263 patients with gigantism or acromegaly and 579 patients with acromegaly, the incidence of GPR101 mutation was shown to be 1.1% and 0.69% respectively (51,52)

GPR101 encodes an orphan G-protein-coupled receptor that is predicted to bind the stimulatory G protein and regulate activation of adenylyl cyclase. Overexpression of this mutated form of GPR101 in rat GH3 somatotroph cells resulted in increased proliferation and growth hormone secretion, along with increased cAMP signalling. In rare cases of sporadic acromegaly, mutation in GPR101 may upregulate cAMP signalling and promote growth hormone secretion and tumorigenesis.

Micro Rna In Somatotroph Adenomas

Recently, miRNA profiling of pituitary adenomas has shown that miR-23a, miR-23b, and miR-24-2 expression were increased in these somatotroph adenomas along with prolactinomas (53). The function of these miRNAs is unknown. Microarray analysis of somatotroph adenomas and normal pituitary gland showed significant downregulation of miR-34b, miR-326, miR-432, miR-548c-3p, miR-570 and miR-603 in adenomas. Among the targets of these miRNAs are high-mobility group A1 (HMGA1), HMGA2 and E2F1, genes whose activation plays a role in pituitary tumorigenesis. Overexpression of these miRNAs resulted in reduced growth of pituitary adenoma cell lines (54).

Epigenetic Regulators Of Somatotroph Adenoma Progression

A number of studies propose an epigenetic mechanism of pituitary somatotroph tumorigenesis. The expression of the adherens junction component E-cadherin has been shown to be significantly lower in sparsely than densely granulated adenomas and lower levels of E-cadherin correlate with larger tumour size, invasiveness, GH and IGF-1 levels and poor acute response to SSAs (55). A regulator of alternative splicing that promotes the epithelial phenotype (ESRP1) was found to be expressed at much lower levels in tumours that did not express E-cadherin (56). The role of ESRP1 in somatotroph adenomas is yet to be clarified, but tumours expressing low levels of ESRP1 also expressed low levels of proteins involved in regulation of the SNARE complex, vesicle trafficking and calcium signalling (56).

Somatotroph Adenomas Are Not Associated With Recurrent Genetic Alterations

Whole genome and exome sequencing of somatotroph adenomas has not identified recurrent genetic alterations other than those in Gsα. Pathway analysis has suggested that mutation events were associated with the cAMP pathway and calcium signalling pathway (57,58).

 

PRL-PRODUCING ADENOMA

Definition

A Pit-1-lineage derived adenoma expressing mostly prolactin and containing characteristic ultrastructural secretory granules demonstrating ‘misplaced exocytosis’.

Pathology

Lactotroph adenomas are the most common hormone-secreting pituitary adenomas. Two types are distinguished according to their granularity – sparsely and densely granulated. A third, very rare subtype, is the so-called acidophil stem cell adenoma. Prolactinomas in women are often detected at younger age and smaller size than in men. This has been attributed to the clinical syndrome associated with these tumours in women, but some observations suggest that lactotroph macroadenomas in men may be biologically different and behave more aggressively. The typical functional lactotroph adenoma consists of sheets of either acidophilic or chromophobe cells, which are smaller than in other adenomas (even in patients not exposed to dopaminergic agonists). In drug-responders morphological effects may be striking, resulting in reduced granularity, shrunken cytoplasm and condensed, hyperchromatic nuclei. Most tumours are of the sparsely granulated subtype characterised by chromophobe cytoplasm and restriction of prolactin immunohistochemistry to the Golgi apparatus, resulting in a polarised or cap-like prolactin pattern. Densely granulated tumours show a diffuse pattern and are acidophil. The sparsely granulated tumour may be associated with spherical calcifications (psammoma bodies) or amyloid deposition.

The acidophil stem-cell adenoma is rare and its nosological status remains to be further defined. It shows eosinophilia on H&E due to accumulation of mitochondria (oncocytic change) and distinct clear cytoplasmic vacuoles may be seen on light microscopy. Occasional perinuclear dot-like fibrous bodies may be seen with cytokeratin stains. The acidophil stem-cell adenoma is considered to be more prone to recurrence than other adenomas.

Figure 3b-8: Prolactinoma with atypia and resistance to dopamine agonist treatment. Prolactinomas consist of sheets of relatively small, monomorphic cells (top left) with a cap-like (‘Golgi-pattern’) staining for prolactin (top right). Most pituitary adenomas of any lineage, including lactotroph adenomas, do not show any mitotic figure in routine stains. Some lactotroph adenomas in men can be resistant to dopamine agonists and histologically atypical, as in this example: Mitotic figures are common, the Ki67/MIB-1 proliferation index is high (10-20% [usually <3%], bottom left) and nuclear p53 is overexpressed (bottom right). This indicates a high likelihood of tumour recurrence if incompletely excised (as proved to be the case in this instance).

Figure 3b-9: Histology of dopamine agonist response in a prolactinoma. Bromocriptine treated prolactinomas respond with characteristic dense fibrosis and condensation of the cytoplasm and nucleus of the neoplastic cells. Left – untreated, right treated.

Molecular Genetics

To date, no mutational events have been unequivocally associated with prolactinomas. However, management of prolactinomas is predominantly medical, using dopamine agonists, with a high proportion of patients achieving disease control or remission, so surgery is not often indicated. Consequently, surgical specimens are few and represent patients who do not tolerate or respond to medical treatment. These tumours are likely to have acquired multiple alterations that allow escape from apoptosis or unrestrained replicative potential which could complicate analysis of mechanisms involved in dopamine agonist-responsive PRLomas.

Oestrogen Receptor Aib1 And Aromatase

A significant correlation between oestrogen receptor ERα mRNA and PRL level, tumour volume and TGFβ1 mRNA was observed in prolactinomas (59), suggesting a role for both ERα and TGFβ1 in prolactinoma tumorigenesis, but the mechanism by which this may occur is unclear.

AIB1 (Amplified in breast cancer) is a member of the p160/SRC family of nuclear co-activators and is a co-activator of the oestrogen receptor. It integrates extracellular signals from growth factors and – through MAPK activation - relays them to the oestrogen receptor, enhancing its transcriptional activity (60). Overexpression of AIB1 was observed in prolactinomas and was associated with expression of ERα and aromatase. In addition, subcellular distribution of AIB1 was linked to cell cycle phase and viability. Nuclear AIB1 expression correlated with nuclear PCNA (a marker of cell proliferation) and cytosolic expression correlated with caspase-3 activation (a marker of apoptosis) (61).

Increased expression of aromatase cytochrome P450 (an enzyme that converts androgens to oestrogens) was observed in prolactinoma relative to normal pituitary but its expression did not correlate with resistance to dopamine agonists or remission (62).

Dopamine Receptor And Downstream Signalling

Dopamine agonists are the first choice of treatment for PRLomas and act by increasing the inhibition of prolactin release mediated by dopamine signalling. The major dopamine receptors expressed in pituitary are D1R (adenylyl cyclase-stimulating) and D2R (adenylyl cyclase- inhibiting). Expression of D2R is more prevalent. The dopamine receptor D2 is expressed as long (D2L) and short (D2S) isoforms, with D2L being the predominant isoform. In patients who were poor responders and those with secondary DA resistance, levels of D2L were significantly reduced (63).

The molecular mechanism of resistance to dopamine agonists is not fully understood. There have been no reported mutations in dopamine receptors in prolactinoma, however, studies have been few (64). Downregulation of the dopamine receptor (D2R) and alterations in the downstream signalling pathway are thought to be involved (65) and binding of PREB (prolactin regulatory element-binding protein) has been shown to be essential for dopamine-mediated inhibition of PRL gene expression – mutation of the PREB consensus sequence in the PRL promoter of GH3 cells prevented cabergoline-induced suppression of PRL expression (66). Further support for the role of dopamine receptor in DA resistance came from studies in mice. In xenografts of prolactin-secreting GH3 cells, those overexpressing the short form of the dopamine receptor (D2S) showed increased sensitivity to bromocriptine in the form of reduced tumour growth (67).

Filamin –A (FLNA) is a cytoskeletal protein that is widely expressed and associates with D2R. FLNA is important for D2R signalling and targeting. In PRLomas with differing responses to DAs, the effect of FLNA on D2R expression and signalling was investigated. Silencing of FLNA in DA-sensitive PRLoma primary cultures resulted in reduced D2R expression and signalling, which could be restored by FLNA overexpression, however, in cells that do not express D2R, overexpression of FLNA did not induce D2R expression, suggesting a more complex mechanism of regulation of D2R expression and signalling (68).

Nerve Growth Factor

A series of studies of PRLomas that were totally resistant to DA therapy and lacked D2 receptors expressed NGFR (nerve growth factor receptor) and cells from these tumours could be induced to differentiate and express D2R upon NGF treatment, furthermore, this expression persisted after cessation of NGF treatment, a feature that was accompanied by reduced tumour growth (69). In female patients with microprolactinoma, hyperprolactinaemia correlated with increased serum NGF, suggesting that release of both molecules is regulated by a D2R-mediated mechanism (70). Further insight into the mechanism of NGF-mediated suppression of DA-resistant PRLoma growth came from the observation that in DA-resistant cells, p53 adopted a different conformation that prevented its nuclear translocation. Treatment with NGF restored p52 conformation and DNA-binding ability, an effect mediated by trkA through activation of PI-3-K (71).

Egfr Receptor Family

Signalling through ErbB and other EGFR family tyrosine kinase receptors occurs upstream of PRL synthesis. The subtype and distribution of expression of these receptors was correlated with therapeutic reduction of prolactin levels in DA resistant prolactinomas in patients receiving lapatinib treatment. Increased expression of ErbB3 was associated with optic chiasm compression, suprasellar extension and carotid artery encasement. Higher ErbB3 expression was also associated with increased response to DA therapy (72)

High mobility group A2 (HMGA2) is an abundant, non-histone DNA-binding protein that mediates the assembly of nucleoprotein complexes involved in the determination of chromatin architecture, transcriptional regulation and RNA processing. HMGA2 is involved in many aspects of cell function, including proliferation and tumour progression, but the exact role of HMGA2 is still not understood (reviewed in (50,73)). The HMGA2 gene was found to be amplified and overexpressed in PRLomas, which often have trisomy of chromosome 12 (containing the HMGA2 gene) (74,75). HMGA2 is thought to promote the activity of transcription factor E2F1, which is required for entry of cells into S-phase. In non-proliferating cells, this activity is repressed by interaction of E2F1 with retinoblastoma protein (pRB) (67). Expression of HMGA2 and HMGA1B have been shown to correlate with expression of PIT1, a transcription factor that regulates expression of PRL (along with GH, GHRHR and Pit1 itself), HMGA2 and HMGA1B bind the Pit1 promoter and enhance Pit1 expression, implicating HMGA2 (and HMGA1B) in pituitary tumorigenesis (76).

E-cadherin, α, β and γ catenins and p120

An immunohistochemical comparison of the expression of E-cadherin, α, β and γ catenins and p120 in normal pituitary, indolent and invasive prolactinomas showed that expression of these proteins was membranous and strong in normal pituitary, decreased in prolactinoma and markedly decreased or absent in invasive prolactinoma, with the exception of γ-catenin, which was expressed more highly in invasive prolactinoma (77). The expression of E-cadherin was inversely proportional to invasiveness, proliferation index (Ki67) and tumour size in prolactinoma. E-cadherin is a suppressor of invasion and participates in the formation of adherens junctions and a decrease in its expression is often seen associated with tumour invasiveness (reviewed in (78,79)).

Micro Rna In Prolactinomas

Little is known about the involvement of miRNAs in prolactinoma pathogenesis. A study examining the expression profiles of miRNAs in prolactinomas that had been treated with bromocriptine or were treatment naïve showed upregulation of miR-206, miR-516b and miR-550 and downregulation of  miR-671-5p was shown to be associated with bromocriptine treatment (80). A study examining miRNA expression profiles in bromocriptine-resistant and bromocriptine-sensitive prolactinomas showed that resistance was associated with increased expression of Hsa-mir-93, hsa-mir-17, hsa-mir-22*, hsa-mir-126*, hsa-mir-142-3p, hsa-mir-144*, hsa-mir-486-5p, hsa-mir-451, and hsa-mir-92a and decreased expression of hsa-mir-30a, hsa-mir-382, and hsa-mir-136 (81). The functional significance of this change in expression pattern is not understood, but silencing of mir-93 was shown to suppress p21 expression.

 

THS-PRODUCING ADENOMA

Definition

A Pit-1-lineage derived neoplasm that mostly expresses TSH and contains typical TSH-type granules on electron microscopy.

Pathology

Thyrotroph adenomas are rare (~1% of all pituitary adenomas). They arise usually in the 5th decade and present as functional macroadenomas resulting in diffuse goitre and hyperthyroidism. Longstanding primary hypothyroidism may lead to thyrotroph adenomas via thyrotroph hyperplasia. Histologically they comprise sheets of angulated or elongated, chromophobe cells, often accompanied by fibrosis. Staining for beta-TSH is usually patchy; tumour cells also express GATA-2 and Pit-1.

Figure 3b-10: Histology of thyrotroph adenoma. These rare tumours contain interlacing, relatively plump spindle cells (left) with strong, patchy TSH expression (right).

Molecular Genetics

The pathogenetic mechanisms of thyroid-hormone-producing adenomas (TSHomas) are not well understood. This may be in part due to the rarity of the lesion (thyrotroph adenomas are estimated to represent 1-3% of pituitary adenomas (82)). No mutations have so far been associated with TSHomas. Experiments that sequenced TSHomas show no mutations in G-protein subunits or the TRH receptor (83) . Pit 1 is overexpressed in these tumours, but not mutated. Expression of somatostatin receptors SSTR2A and SSTR5 was found in TSHomas (31). A high ratio of expression of SSTR5 to SSTR2 might indicate a better response to long-term treatment with somatostatin analogues in TSHomas (84,85) but this is not a consistent finding (86).

 

ACTH-PRODUCING ADENOMA

Definition

Corticotroph pituitary adenomas are Tpit-lineage derived tumours producing ACTH stored in ultrastructurally typical ACTH granules. They are the defining neoplasms of Cushing’s disease.

Pathology

Corticotroph adenomas associated with manifest Cushing’s disease are composed of deeply basophilic (PAS-positive) cells with granular cytoplasm and round nuclei; these comprise the densely granulated subtype of ACTH adenoma. Most tumours arise in women in the 4th or 5th decade (F:M = 8:1); prepubertal tumours are rare and equally distributed between the sexes, with a slight male predominance. Sparsely granulated tumours are weakly basophilic or chromophobe, and individuals may lack an overt Cushing’s phenotype (‘silent corticotroph adenomas’). The cytokeratin pattern in typical Cushing’s adenomas is diffuse. However, rare neoplasms may display ‘Crooke’s hyaline change’, classically interpreted as a morphological manifestation of intact feedback inhibition by excess systemic cortisol on non-neoplastic corticotrophs. This change is therefore seen in intact acini adjacent to a typical corticotroph adenoma and results in a ring-like accumulation of cytokeratins. If present in many adenoma cells, these tumours are called ‘Crooke’s cell adenomas’, possibly representing a subgroup with an adverse outcome and silent presentation. Immunohistochemistry of all corticotroph adenomas shows strong nuclear T-pit positivity. Densely granulated tumours show strong diffuse cytoplasmic ACTH expression, whilst chromophobe tumours show only patchy positivity. Following detection of somatic Usp8 mutations in a subgroup of ACTH-producing adenomas (see below), it has been suggested that nuclear translocation of Usp8 may represent an immunohistochemically detectable surrogate marker of these mutations (87). USP8-mutated corticotroph adenomas are more commonly microadenomas compared to USP8-wild-type Cushing’s adenomas. The ‘minimal pathological unit’ of Cushing’s disease is corticotroph hyperplasia. This is defined as a distention of normal adenohypophyseal acini by a homogeneous population of corticotrophs that does not lead to complete breakdown of the acinar reticulin border. The described morphological entities associated with Cushing’s disease are illustrated below in figures 3b-11 to 3b-14.

Figure 3b-11: Histology of corticotroph hyperplasia causing Cushing’s disease. Note distended but still intact reticulin network of hyperplastic pituitary acini (left) populated by a homogeneous population of deeply basophilic cells (center) expressing ACTH (right).

Figure 3b-12: Histology of a corticotroph microadenoma. These (often USP8-mutated) Cushing’s adenomas represent microscopic nodules well-demarcated from normal gland (NG). Reticulin stain is often essential for their detection (top left) and discrimination from corticotroph hyperplasia: a corticotroph adenoma results in completed destruction of the reticulin network as seen here (right side of the dashed line in top left image). Typical Cushing’s adenomas are deeply basophilic (top right) and show strong diffuse ACTH positivity (bottom left) and an intact keratin cytoskeleton (bottom right).

Figure 3b-13: Crooke’s hyaline change in non-neoplastic corticotroph cells in response to hypercortisolaemia. The physiological response of normal corticotrophs to exposure of excess cortisol (of any source, neoplastic or iatrogenic) is downregulation of ACTH synthesis and development of hyaline, cytokeratin-rich perinuclear rings: Crooke’s hyaline degeneration (named after the English endocrinologist Arthur Carleton Crooke).

Figure 3b-14: Histology of Crooke’s cell adenoma. In these corticotroph adenomas Crooke’s cell change is seen in the tumour cells, rather than non-neoplastic corticotrophs. This is clearly evident in PAS-OG histochemistry, where the hyaline ring displaces the deeply basophilic granules (left). It is also reflected in the dense ring-like cytokeratin pattern (right).

Molecular Genetics

ACTH-producing adenomas causing Cushing’s disease are associated with both an excess of corticotroph releasing hormone (CRH) and a loss of negative feedback inhibition by glucocorticoids. However, no mutations in either the CRH receptor or the glucocorticoid receptor have been reported.

USP8

Exome sequencing of corticotroph adenomas from patients with Cushing’s disease revealed recurrent heterozygous somatic mutations in the deubiquitinase USP8 in one third of cases (87,88), although one study has estimated the prevalence to be 62% (89). Mutations were clustered within the 14-3-3 binding motif of USP8: a highly evolutionarily conserved region that is rarely mutated in other human cancers. Mutations in USP8 were found to be more common in adult than paediatric cases and more common in females than males (ratio 5:2). Patients with a USP8 mutation were less likely to develop postoperative adrenal insufficiency (88). Tumours with a mutation in USP8 were also found to be smaller and to produce more ACTH than their wild-type counterparts (89)

USP8 is a ubiquitin-specific protease that regulates the fate of numerous cellular proteins. Conjugated ubiquitin molecules target a protein for degradation and these modifications are removed by deubiquitinases (DUBs). USP8 specifically targets the pathway whereby activated EGFR is targeted for lysosomal degradation and leads to increased cellular concentrations of EGFR and sustained levels of EGFR signalling. However, one study of 60 corticotroph adenomas did not find any association between USP8 mutation and EGFR expression; there was, however significantly higher expression of POMC, SSTR5 and MGMT (90). Binding of 14-3-3 proteins to Usp8 has a suppressive effect and so mutations in the 14-3-3 binding motif of USP8 that perturb this interaction lead to a gain of function of USP8 and increased EGFR signalling. Additionally, mutations in the 14-3-3 binding motif that abrogate 14-3-3 binding result in exposure of a cleavage site and an increase in proteolytic cleavage of USP8 between Lys714 and Arg715. This results in a shorter 40kDa C-terminal fragment of USP8 with increased deubiquitinase activity. Mutant USP8 also results in increased activation of the POMC promoter in the AtT-20 corticotroph adenoma mouse cell line.

Cyclins And Cyclin-Dependent Kinases

A study investigating the role of cell-cycle regulators and related transcription factors in ACTH-secreting and silent corticotroph adenomas found that CDKN2A expression was four times greater in ACTH-expressing than silent corticotroph adenomas, while cyclins D1, E1 and B1 were suppressed. It is suggested that the upregulation of a cell-cycle inhibitor combined with the downregulation of cyclins may restrict growth of ACTH-producing adenomas compared to their silent counterparts (91).

11β-Hydroxysteroid Dehydrogenase

Cortisol and inactive cortisone are interconverted by 11β-hydroxysteroid dehydrogenase. This enzyme exists as two isoforms: 11β-HSD1 and 11β-HSD2. Studies investigating the expression ratio of these two isoforms in ACTH-expressing adenomas found that Isoform 1 is downregulated in ACTHomas and 2 is upregulated compared to normal pituitary (92). The role of increased expression of 11β-HSD2 in ACTHoma tumorigenesis is unclear and findings are inconsistent (93).

Somatostatin Receptors

A comparison of the expression of somatostatin receptor subtypes SSTR2 and SSTR5 in silent corticotroph adenomas and adenomas responsible for Cushing’s disease showed that mRNA encoding SSTR1 and 2 was expressed in greater quantities in silent corticotrophs (SSTR2 5-fold increase), whereas in Cushing’s disease, SSTR5 was expressed more highly (14-fold increase) (94). Although the implications of this difference in expression are not fully understood, it may be that treatments that selectively target SSTR5 could be useful for ACTHoma treatment.

In a series of ACTH-secreting pituitary adenomas, levels of miR-26a were assessed by RT-qPCR. This micro-RNA was upregulated in all ACTHomas compared to normal pituitary. The putative target of this miRNA, PRKCD, was downregulated in tumours with elevated miR-26a (95). PRKCD encodes protein kinase C delta, a serine/threonine kinase involved in a diverse range of signalling pathways including regulation of growth, apoptosis and differentiation.

 

GONADOTROPHIN-PRODUCING ADENOMA

Definition

Pituitary adenomas derived from SF-1 expressing adenohypophyseal cells producing mainly FSH or LH and typical secretory granules.

Pathology

Classic gonadotroph adenomas are chromophobe adenomas with a growth pattern that may include papillae and perivascular pseudorosettes. Although all tumour cells express nuclear SF-1, FSH and LH are restricted to clusters of cells often demonstrating striking polarisation towards vascular lumina in well-differentiated examples. Gonadotroph adenomas are usually endocrinologically silent and therefore present as macroadenomas with compression of the optic chiasm or invasion of the cavernous sinus. They are often called non-functioning adenomas (NFAs) but it should be noted that adenomas of other lineages may also be clinically ‘non-functional’ (e.g. silent corticotroph or somatotroph adenomas). Rare functionally active tumours in females of reproductive age may be associated with ovarian hyperstimulation syndrome.

Figure 3b-15.1+15.2: MRI and macroscopic pathology of gonadotroph pituitary macroadenomas. Gonadotroph adenomas are usually clinically silent and thus tend to present as space occupying lesions compressing the optic chiasm, pituitary stalk or hypothalamus. 3b-15.1: Sagittal MRI (left) and post-mortem view of the same tumour. Note the suprasellar extension and compression of the hypothalamus. 3b-15.2: Two further gonadotroph macroadenomas seen in situ in the skull base (left) and the base of the brain (right) compressing the optic chiasm. Note in the left image the anatomical relationship to the sphenoid wings, right optic nerve and basal vessels of the brain (one of which is stuck to the rostral surface of the macroadenoma). The adenoma (asterisk) in the right image has grown through the diaphragma sellae and therefore is attached to dura mater – which is used to pull the adenoma away from the chiasmatic cistern to reveal the chiasm (arrow) and optic nerves.

Figure 3b-16: Histology of a gonadotroph (non-functioning) adenoma. In their typical form these adenomas have a distinct architecture comprising perivascular rosettes of neuroendocrine cells with a distinct polarity of their processes towards the vascular lumen (left) and always patchy and focal, rather than diffuse expression of FSH and LH (right). Note a rare mitosis in the HE image (left, arrow).

Molecular Genetics

Micorarray studies comparing functional gonadotroph tumours to normal post mortem pituitary found that downstream p53 target genes RPRM, p21/CDKN1A and PMAIP1 were consistently downregulated (96). These genes are mediators of cell cycle arrest and apoptosis. Members of the GADD45 family were differentially expressed, with GADD45β downregulated in gonadotroph adenomas compared to normal gland. Overexpression of GADD45β in gonadotroph cells inhibited proliferation and activated apoptosis in the absence of growth factor, however, the authors found no evidence of hypermethylation of GADD45β (96) and so the mechanism of downregulation remains unknown.

A whole-exome sequencing study of histologically typical, clinically non-functioning gonadotroph adenomas revealed 24 somatic variants in independent genes, none of which were recurrent. There were no mutations that had been previously associated with pituitary tumorigenesis and the authors conclude that mechanisms other than somatic mutation may be involved in sporadic NFPA tumorigenesis (74).

 

NULL CELL ADENOMA

Definition

Null cell adenomas are neoplasms derived from adenohypophyseal endocrine cells that cannot be assigned to any specific subtype based on transcription factor, hormone or ultrastructural features.

Pathology

These tumours are chromophobe and show usually a diffuse growth pattern. Increasing sensitivity and specificity of immunohistochemical techniques for detection of pituitary transcription factors and hormones make this a shrinking diagnostic subgroup. Distinction of this subtype from rare endocrine tumours not derived from adenohypophyseal cells (paraganglioma, metastatic endocrine carcinoma) is important but can be difficult.

Molecular Genetics

As tumours previously designated ‘null-cell’ or ‘non-functioning’ may actually represent SF-1 lineage tumours, interpretation of molecular studies is difficult. A study examining the expression of E-cadherin (CDH1), slug (SNAI2) and oestrogen receptor ERα and ERβ in invasive compared to non-invasive non-functioning pituitary adenomas (NFPAs) showed that E-cadherin is downregulated in more invasive tumours, while its repressor, slug, is upregulated. Expression of slug was positively correlated with ERα expression, while E-cadherin was positively correlated with ERβ expression. The relevance of these findings for patient prognosis and treatment has yet to be determined (75). MicroRNA profiling of NFPAs compared to normal pituitary showed that miRNAs predicted to target components of the TGFβ signalling pathway and result in their downregulation are overexpressed. This pathway is known to have a role in tumorigenesis, but the nature of its role in the pathogenesis of NFPAs is not well understood (97). Another pathway commonly disrupted in tumorigenesis, the Notch signalling pathway was investigated in NFPAs. Upregulation of Notch3 (a regulator of cell proliferation and apoptosis) and its ligand, Jagged1, was observed in NFPAs compared to normal gland. Owing to the complexity of Notch pathway regulation, the consequence of this upregulation is not yet clear (98,99). The pathogenic mechanisms of NFPAs are largely unclear and targeted treatments are not available.

 

PITUITARY CARCINOMA

Definition

Pituitary carcinoma is defined as a neoplasm of adenohypophyseal endocrine cells with cerebrospinal or systemic dissemination.

Pathology

Pituitary carcinoma is very rare, comprising approximately ~0.2% of operated pituitary neoplasms. Most pituitary carcinomas develop from recurrent endocrinologically functioning, invasive macroadenomas with a highly variable lag period. The majority represents corticotroph or lactotroph neoplasms. Bilateral adrenalectomy in the setting of Cushing’s syndrome with an undetected pituitary microadenoma may predispose to pituitary carcinoma (Nelson’s syndrome). It has also been suggested that silent corticotroph adenomas or Crooke’s cell adenomas may pose a risk, but data are scant. Histologically, pituitary carcinomas may show remarkably little pleomorphism; however, an increased MIB-1 index and p53 overexpression are usually present. Despite the introduction of the ‘atypical pituitary adenoma’ category, no reliable diagnostic markers are available that allow prediction of carcinomatous behaviour before dissemination has occurred. The prognosis is poor once systemic metastases are present. Treatment with temozolomide should be considered.

 

Molecular Genetics

Molecular studies of pituitary carcinomas are scant, presumably due to the rarity of the lesion. One study has observed a mutation in H-ras in a PRL-producing carcinoma. Unlike other pituitary tumours, pituitary carcinomas show aggressive tendencies and metastasise (100). A microarray study comparing expression levels in pituitary adenomas relative to an ACTH pituitary carcnioma identified the LGALS3 (galactin 3) gene as being upregulated in pituitary carcinomas (101)

 

PITUITARY BLASTOMA

Definition

Pituitary blastoma is a rare pediatric neoplasm of the anterior gland composed of primitive follicular structures of endocrine cells admixed with folliculo-stellate cells. It is pathognomonic of germline DICER1 syndrome or pleuropulmonary blastoma-familial tumor and dysplasia syndrome [online Mendelian inheritance in man (OMIM) #601200] (102)

Pathology

Tumours are variably cellular, likely reflecting different degrees of maturation, and consist of cells arranged in rosettes and glandular structures reminiscent of Rathke’s epithelium, undifferentiated cells (blastema) and larger granular (secretory cells) (103). Ultrastructurally FS-like cells may also be seen. There is usually ACTH-positivity in a few cells and GH may also be seen.

Mitoses are present but the MIB-1 index may be very variable. The designation as ‘blastoma’ reflects the original view that these tumours are highly malignant with a natural history similar to other embryonal neoplasms; however, more recent evidence suggests that the prognosis is not uniformly poor (102).

Figure 3b-17: Histology of a pituitary blastoma. These are primitive tumours resembling ‘small-blue round-cell’ neoplasms.

Reproduced from Acta Neuropathologica ”Pituitary blastoma: a pathognomonic feature of germ-line DICER1 mutations” volume 128, 2014 pages 111-122 de Kock L, et al. (102) with permission of Springer. Fig. 3a case 13, T1-weighted post-contrast midline sagittal MR image showing pituitary region mass (red arrow). b case 4, hematoxylin and eosin (H&E) staining ×250: three enlarged follicles lined by stem cells. c Immunohistochemical staining I case 10, growth hormone (GH) immunostaining ×400: enlarged GH/alpha subunit cells immunopositive for GH. II case 10, ACTH immunostaining ×400: small vessel surrounded by stem cells. Some cells display ACTH immunoreactivity

 

Molecular Genetics

The precise mechanisms driving tumorigenesis remain to be defined. The morphological evidence of stem-cell-like features of pituitary blastoma cells and known roles of micro RNAs in regulation of stem cell differentiation make it plausible that profound abnormalities in micro RNA profiles following mutations of Dicer, a key regulator of micro RNA maturation, are causative. Mutations occur in highly conserved regions of DICER1, particularly the RNase IIIb domain, resulting in predicted loss of function, following Knudson’s dual hit model.

 

CRANIOPHARYNGIOMA

Definition

Usually benign, but invasive epithelial lesions of the supra-sellar region or third ventricle that exist as two variants: adamantinomatous and papillary. The variants have distinct clinicopatholoigcal and genetic features which may represent different pathogenic mechanisms.

Pathology

Craniopharyngiomas occur with an incidence of 0.13 per 100 000 person years. There are two variants: adamantinomatous (aCP) and papillary (pCP) that occur in the ratio ~9:1 with no sex differences. aCP usually occurs in childhood (mean age 5-14 years) while the pCP is almost exclusively seen in adults (mean age 65-74 years). Craniopharyngiomas are complex, epithelial neoplasms that arise in the sellar region along the vestigial craniopharyngeal tract. Although some overlapping features have been observed, the two variants represent clinicopathologically distinct lesions.

Adamantinomatous Craniopharyngioma

aCPs are located predominantly in the suprasellar region although infrequently they have an intrasellar component. Rare locations include the sphenoid sinus and cerebello-pontine angle. They are multi-lobulated and often multi-cystic masses. On T1 weighted MRI imaging they are hypo- or iso-intense with areas of hyperintensity, corresponding to the cystic components. Enhancement is strong and heterogeneous.

Macroscopically, aCP are firm, lobular lesions with an irregular, but sharp interface strongly adherent to and invading surrounding structures. Cyst contents are variable and may contain necrotic or inflammatory debris or a dark, cholesterol-rich fluid resembling motor oil. Calcification is often present.

Microscopically, the architecture of aCP shows a well circumscribed, multicystic lesion with finger-like protrusions into surrounding brain parenchyma (Figure 3.b.18). The lesion is composed of a peripheral palisading epithelium surrounding a loose core of stellate reticulum. Nodules of anuclear “ghost cells” containing wet keratin are commonly found and are pathognomonic for this tumour type. Near the tumour invading edge, epithelial whorls of cells that often show translocation of beta-catenin from membrane to cytosol/nucleus are common. Degenerative changes include intra-cystic squamous debris, chronic inflammation and the appearance of cholesterol clefts and extensive calcification. These changes can elicit a granulomatous inflammatory response and brain invasion may cause Rosenthal fibre gliosis.

Figure 3b-18: Histology of adamantinomatous craniopharyngioma. This highly distinctive neoplasm consists of loosely arranged ‘stellate’ reticular stroma, palisading peripheral epithelium and nodules of ‘ghost’ cells (degenerated keratinocytes, arrow in the left image). Finger-like protrusions of neoplastic epithelium commonly invade the hypothalamic brain parenchyma resulting in a dense, hypereosinophilic Rosenthal fibre gliosis (arrow and bottom half of right image).

Figure 3b-19: Histology of adamantinomatous craniopharyngioma – beta-catenin. Nodules or whorls of tinctorially distinct epithelium is often seen in the invading edge of the adamantinomatous subtype (left, black arrow). The cells in these nests demonstrate nuclear translocation of beta-catenin (right, white arrow), indicating activated WNT signalling.

Papillary Craniopharyngioma

pCPs are located in the suprasellar region or within the third ventricle. They are usually more solid than aCPs but may have a cystic component. On T1-weighted MRI imaging, they appear hypointense with enhancement of the cyst wall.

Macroscopically, pCP are discrete, well circumscribed and often solid lesions with little adherence to surrounding structures. When cystic, the cyst contents are clear without cholesterol crystals. Clacification is not seen.

Microscopically, pCPs are composed of squamous, well differentiated, non-keratinizing epithelium. There is no stellate reticulum; these lesions have a fibrovascular stroma without a palisading layer. The lesions form pseudopapillae as a result of epithelial dehiscence and do not contain wet keratin. In these lesions, beta-catenin retains its membranous location. There may be scant foci of goblet or ciliated cells that resemble Rathke’s cleft cyst

Figure 3b-20: Histology of papillary craniopharyngioma. Papillary craniopharyngiomas lack ’stellate reticular’ stroma and keratin ‘ghost’ cell nests and nodules or whorls in the advancing edge. They have a more solid growth pattern prone to artifactual ‘cracking’ (arrow, left) giving it a (pseudo-) papillary appearance.

Figure 3b-21: Histology of papillary craniopharyngioma – beta-catenin and BRAFV600E. Papillary craniopharyngiomas lack any nuclear translocation of beta-catenin; it remains restricted to the adherens junction (left). Instead, the tumours are always positive for mutated BRAFV600E protein (right).

Molecular Genetics

Two alternate theories have been proposed to explain the pathogenesis of craniopharyngiomas. The embryogenetic theory states that CPs arise from neoplastic transformation of ectopic embryonic remnants of Rathke’s pouch. The metaplastic theory states that differentiated squamous epithelium that forms part of the anterior pituitary or pituitary stalk undergoes metaplastic transformation. In addition, a dual theory has also been proposed that the adamantinomatous type is formed via the embryogenetic mechanism while the papillary type follows the metaplastic route.

Mutations have been found in craniopharyngiomas that may co-segregate with subtype. Mutations in exon 3 of CTNNB1, the gene encoding β-catenin have been observed in around 70% of aCP cases, although estimates of frequency vary (104-106). Β-catenin is a mediator of the Wnt signaling pathway and exon 3 of CTNNB1 contains critical Ser and Thr residues S33, S37, T41 and S45 (107) that are phosphorylated during formation of the β-catenin degradation complex. This complex regulates Wnt signalling by targeting β-catenin for ubiquitination and degradation in the absence of receptor-bound Wnt ligand. Mutations in these critical Ser and Thr residues prevent formation of the β-catenin destruction complex, leading to a constitutively active Wnt signal and accumulation of β-catenin in the cytosol and nucleus. Nuclear and cytosolic β-catenin was observed in 90-100% of adamantinomatous craniopharyngiomas, but never in papillary craniopharyngiomas or other tumours of the sellar region (pituitary adenomas, arachnoid cysts, Rathke’s cleft cysts and xanthogranulomas), (104,108-110). A study that selectively expressed mutant CTNNB1 in developing mouse pituitary showed disrupted Pit1 lineage differentiation, hypopituitarism and large, cystic tumours resembling adamantinomatous craniopharyngiomas (111,112), suggesting that CTNNB1 mutation may be sufficient for aCP formation.

Despite the presence of a β-catenin mutation in all cells, nuclear and cytosolic accumulation of β-catenin is found only in small clusters of cells near the infiltrating edge of the tumour (104,108,109,113-115). It has been shown that these cells possess stem-cell-like properties and may perform a paracrine function by secretion of members of the SHH, BMP and FGF family that promote division of the surrounding tumour cells (113,116) These cells also show reduced expression of fascin and increased phosphorylation and activation of EGFR, suggesting increased capacity for migration (117-119).

Mutations in the protein kinase BRAF (V600E) have been shown to be associated with pCPs in 81-95% of cases (106,110). BRAF is a component of the MAP kinase signalling cascade and mutations in this pathway are associated with numerous neoplasms including melanoma, for which treatment with BRAF inhibitors is common. There have been two reports of targeted treatment of pCP. Dabrafenib (150mg, orally twice daily) and trametinib (2mg, orally twice daily), resulted in 85% reduction in tumour volume after 35 days (120). Vemurafenib (960mg twice daily) for three months resulted in significant reduction in tumour volume, but the tumour recurred within 6 weeks upon cessation of treatment (121).

The two subtypes of craniopharyngiomas are clinicopathologically distinct, but do have some overlapping features. This observation has led to the hypothesis that craniopharyngiomas fall on a histopathological continuum with other cystic epithelial sellar lesions (109,122-124). It has been suggested that papillary craniopharyngiomas represent an intermediate entity between Rathke’s cleft cysts and adamantinomatous craniopharyngiomas, as they have been found to contain ciliated epithelial cells and goblet cells characteristic of Rathke’s cleft cysts (122,125-127). Craniopharyngiomas, particularly the papillary form, can arise after treatment for Rathke’s cleft cysts although the possibility of coexisting lesions cannot be excluded (128,129).

 

RATHKE’S CLEFT CYST

Definition

A benign, non-neoplastic epithelial cyst arising from accumulation of mucinous material in remnants of Rathke’s pouch.

Pathology

Classical Rathke’s cleft cysts consist of a monolayer of cuboidal cells on with microvilli and scattered columnar and goblet cells. Cyst contents consist usually of amorphous eosinophilic material. Squamous metaplasia of the lining epithelium is common and may result in the differential diagnosis of craniopharyngioma. Xanthogranulomas with chronic inflammation and cholesterol crystals may also occur. Rathke’s cleft cysts have no neoplastic potential but may recur following incomplete excision.

Figure 3b-22: Histology of Rathke’s cleft cyst. Symptomatic Rathke’s cleft cysts are lined by a ciliated epithelium (top left) that is cytokeratin-positive (top right) and often contains PAS-positive vacuoles (bottom left). It often undergoes attenuation or squamous metaplasia (bottom right).

DIFFERENTIAL DIAGNOSIS OF CYSTIC LESIONS

A series of observations that note similarities between cystic sellar lesions has led to the hypothesis that there exists a histopathological continuum that includes epithelial, epidermoid and dermoid cysts, Rathke’s cleft cysts and both papillary and adamantinomatous craniopharyngiomas (109,122,125,130,131). Although experimental evidence is lacking there are reports of transitional lesions that lend support to this idea. Due to the rarity of these lesions and the paucity of material available for study, so far no genetic event has been unequivocally associated with the development of non-neoplastic cystic lesions arising in the sellar region. Immunohistochemistry and sequencing for BRAFV600E mutations in Rathke’s cleft cysts was negative (132).

 

TUMOURS OF THE NEUROHYPOPYSIS

GRANULAR CELL TUMOUR, PITUICYTOMA, SPINDLE CELL ONCOCYTOMA: TTF-1 FAMILY OF PITUITARY NEOPLASMS

Definition

Rare endocrinologically silent neoplasms of the posterior pituitary or infundibulum that share the expression of thyroid-transcription factor 1 (TTF-1).

Pathology

The neurohypophysis is derived from the floor of the diencephalon. The development of its specialised glial cells, termed pituicytes, is controlled in part by the expression of TTF-1, which is maintained throughout adulthood. Pituicytes are thought to provide structural and functional support for the axonal processes and neurosecretory terminals of oxytocin and vasopressin producing cells whose cell bodies are located in the hypothalamus. Electron microscopic studies have suggested that there are five different types of pituicytes, which (simplified) can be described as: light, dark, granular, ependymal and oncocytic (133). Neoplastic transformation of these cells is thought to give rise to three distinct neoplasms, termed granular cell tumour, pituicytoma and spindle cell oncocytoma. However, the precise relationship of these lesions remains to be defined, as some studies suggest that the spindle cell oncocytoma is arising from folliculo-stellate cells of the adenohypophysis.

Granular Cell Tumour

These lesions may be found incidentally at autopsy as microscopic nodules along the pituitary stalk. Clinically relevant lesions present as slow-growing, solid space-occupying tumours that mimic pituitary macroadenomas on preoperative imaging. Microscopically they are characterised by sheets of relatively large cells with eosinophilic, granular cytoplasm. Nuclei are round, sometimes eccentric and contain inconspicuous nucleoli. There are generally no mitoses. The cytoplasmic granules remain periodic-acid-Schiff (PAS) positive after diastase treatment. Ultrastructurally, the granules correspond to membrane-bound lysosomal organelles. This is reflected in patchy immunostaining with PGM-1 antibody against CD68, a membrane epitope belonging to the lysosomal/endosomal-associated membrane glycoprotein (LAMP) family. Tumour cells are also usually S100-positive but negative for cytokeratins, synaptophysin and pituitary hormones. In our experience granular cell tumours of the sellar region consistently show strong nuclear TTF-1 expression. The proliferation fraction is low (<5%) but tumours with mitoses and multiple recurrences have been described. Surgery is the preferred treatment modality.

Pituicytoma

This variant of posterior pituitary or infundibular TTF-1-positive neoplasm consists of fascicles of elongated yet plump bipolar cells that are pale eosinophilic and usually lack granularity (134). There may be moderate nuclear hyperchromasia but mitoses are generally absent. Electron microscopy demonstrates intermediate filaments and no secretory granules. Tumour cells express vimentin and S100 and show variable GFAP positivity. Proliferation is low. Again, surgery is the main treatment

Spindle Cell Oncocytoma

Spindle cell oncocytoma may share light-microscopic appearances with pituicytoma, particularly if accumulation of mitochondria, a defining feature, is not fully developed. Tumour cells are elongated, spindle-shaped, sometimes arranged in fascicles or epithelioid. Nuclei may be moderately pleomorphic and hyperchromatic but mitoses are again rare, although a few reports documented atypical variants with an increased recurrence rate. Scattered lymphocytic infiltration may be seen. Apart from abundant mitochondria, ultrastructural features that help to distinguish these tumours morphologically are well-formed desmosomes (135). Tumours show strong nuclear TTF-1 positivity and cytoplasmic annexin-1 expression. The latter is shared with folliculo-stellate cells of the adenohypophysis (but may also be seen in pituicytes). EMA may be expressed and is usually absent from pituicytomas and granular cell tumours. GFAP, cytokeratins and neuroendocrine markers are generally negative. The proliferation fraction is usually low but may reach 25% in recurrent tumours.

Figure 3b-23: Histology of TTF-1 positive neoplasms of the neurohypophysis. Granular cell tumour (left) and pituicytoma (right) are morphologically distinct but share nuclear TTF-1 expression, confirming their origin from specialised glial cells of the posterior gland or infundibulum. Both entities are S100 positive but only the pituicytoma expresses GFAP.

Molecular Genetics

No pathognomonic molecular genetic features have been identified for these neoplasms. Comparative genomic hybridization on one case demonstrated losses on chr 1p, 14q and 22q and gains on 5p (136). Presumed glial origin prompted examination of the IDH1 R132H and BRAF V600E mutations and BRAF-KIAA fusion gene in a recent study of 14 cases comprising all three pathologies (137). Systematic genomic and epigenomic analysis may clarify the aetiologic relationship of these tumours.

 

PRIMARY NEOPLASMS OF THE SELLAR REGION THAT MAY MIMIC PITUITARY ADENOMAS

Germinoma

Intracranial germ cell neoplasms have a predilection for midline structures and commonly involve the infundibular region. The most common form of this rare tumour is germinoma.

Definition

An extragonadal germ cell tumour arising in or above the pituitary fossa with histological features resembling gonadal seminoma.

Pathology

Tumours may present as large compressive lesions or subtle thickening of the (posterior) pituitary stalk. Diabetes insipidus is a classic presentation but delayed puberty due to hypopituitarism is also seen. Pituitary germinomas commonly manifest in children or young adults, mostly males. Historically, the incidence is far higher in East Asia than Western countries. CSF/blood tumour markers (alpha-fetoprotein and human chorionic gonadotrophin) that can be diagnostic in germ cell tumours with yolk-sac or choriocarcinoma components may not be helpful in pure germinoma, resulting in biopsy. Histological diagnosis may be difficult because some germinomas elicit a profound inflammatory or even granulomatous reaction that can obscure the neoplastic cells. Typical examples show a biphasic architecture of large tumour cells with vesicular nuclei, prominent nucleoli and mitoses accompanied by reactive lymphocytes.

Tumour cells express placental alkaline phosphatase (PLAP), CD117 (KIT) and the transcription factor POU5F1 (Oct3/4).

Figure 3b-24: Histology of germinoma of the pituitary gland. Pituitary germinomas share their morphological phenotype with gonadal seminomas. They consist of large anaplastic cells accompanied by a dense lymphocytic infiltrate (left). The tumour cells strongly express c-kit, PLAP (right) and Oct3/4 transcription factors.

Molecular Genetics

Intracranial germinomas are thought to arise from displaced primordial germ cells. Until recently, little was known about the molecular pathogenesis of these rare tumours. Comprehensive genomic and transcriptomic analyses revealed that pure germinomas are associated with mutually exclusive mutations in KIT and RAS in the majority of cases (138). These mutations result in the constitutive activation of the KIT-driven MAPK pathway, consistent with the observation of strong KIT expression by these tumours. Another study, employing next-generation sequencing, found additional somatic mutations in CBL, a negative regulator of KIT, as well as copy number gains at the AKT1 locus resulting in mTOR pathway activation (139) This study also found germ line variants in JMJD1C among Japanese patients, a possible explanation for the skewed incidence rates described above (139). Treatment for localised pure germinoma consists of radiotherapy; chemotherapy is an effective strategy to reduce the radiation dose (140).

 

CHORDOMA OF THE CLIVUS

Definition

A slow-growing but highly destructive neoplasm arising from remnants of the rostral notochord.

Pathology

Intracranial chordomas are almost exclusively located at the dorsum sellae. They may result in compression of the pituitary and destruction of the pituitary fossa. Tumours are soft, gelatinous lesions with a striking cytopathology. Typical tumours contain large, vacuolated (‘physaliphorous’) cells surrounded by a mucinous matrix. They are arranged in anastomosing cords or sheets. Occasional mitoses are found. Tumours express S100 and low-molecular weight keratins and epithelial membrane antigen (EMA). Brachyury, or transcription factor T, serves as a highly sensitive and specific marker for the diagnosis of chordoma, facilitating distinction from chondrosarcomas, (chordoid) meningiomas or metastases. It is physiologically expressed in undifferentiated notochord of the axial skeleton. The neoplasm slowly invades along neurovascular bundles and may be surgically incurable. Proton beam therapy is commonly applied in order to preserve neurological function (141). Dedifferentiation upon recurrence may rarely occur, resulting in a poor prognosis.

Figure 3b-25: Histology of sellar chordoma. These tumours arise from remnants of the notochord and are highly characteristic in appearance, comprising pleomorphic cells with very large vacuoles and intracellular PAS-positive mucin, floating in an alcian blue-positive matrix.

Molecular Genetics

The discovery of gene duplications involving the transcription factor T gene (brachyury) in familial chordoma strongly supported the idea of notochordal origin of these tumours (142). However, no recurrent somatic mutations in T or its promotor have been described that could explain the consistently high expression of brachyury by tumour cells. More recent analysis has revealed that a common single nucleotide variant in T (SNP rs2305089) is strongly associated with chordomas in apparently non-mendelian cases (143). The risk allele variant leads to increased expression of T, providing a plausible pathogenetic link and target for future molecular therapies.

 

SECONDARY NEOPLASMS OF THE SELLAR REGION THAT MAY MIMIC PITUITARY ADENOMAS

Definition

These lesions are here defined as neoplasms that arise at extracranial sites and colonise the pituitary, usually via haematogenous spread; i.e. pituitary metastases.

Pathology

The rich vascularity of the pituitary gland facilitates haematogenous seeding of micrometastases. Deposits from breast, lung and gastrointestinal carcinomas are most common. Autopsy series have suggested a relatively high incidence of between 3-27% in the setting of disseminated malignancy. However, many may represent asymptomatic micrometastases. In clinically manifest examples neuroimaging features can be very similar to pituitary adenomas. Even biopsy appearances can be deceptive, particularly in the setting of TTF-1-positive neuroendocrine carcinomas of the lung. However, the degree of cytological atypia and mitoses usually point to the right diagnosis. Judicial use of immunohistochemistry helps to narrow down the possible site of origin if the pituitary metastasis is the first manifestation of an occult malignancy.

 

NON-NEOPLASTIC LESIONSOF THE PITUITARY THAT MAY MIMIC ADENOMAS

LYMPHOCYTIC HYPOPHYSITIS

Definition

A rare lymphocytic inflammatory disorder of the pituitary gland of autoimmune aetiology.

Pathology

Classic lymphocytic hypophysitis consists of a dense, sometimes follicular, lymphoplasmacytoid infiltrate of the gland that in some cases may result in fibrosis and permanent hypopituitarism. All parts of the gland may be affected to variable degrees, resulting in distinction of adenohypophysitis, infundibuloneurohypophysitis or panhypophysitis. In the acute phase symmetrical swelling of the gland may lead to headaches or diabetes insipidus, a presenting symptom often associated with radiologically detectable swelling of the pituitary stalk. Hypogonadotropic hypogonadism represents a common deficit of anterior pituitary function. The inflammatory infiltrate consists predominantly of T-cells with a CD4/CD8 ratio of 2:1 or more. Lymphoid follicles may occasionally be observed.

Figure 3b-26: MRI and histology of lymphocytic hypophysitis. The MRI appearances may be mistaken for a non-functioning adenoma; however, there are distinctive features, including homogeneous enhancement with extension posteriorly and rostrally via the infundibulum (‘infundibulohypophysitis’). There is a dense, destructive lymphocytic infiltrate, leaving islands of residual anterior gland (eosinophilic cells, top right) that in this instance are lactotrophs (bottom left; serial section to top right). The lymphocytes are predominantly T-cells (CD3-positive, bottom left).

Pathogenesis

It is likely that different immunopathogenic mechanisms result in lymphocytic hypophysitis as the common endpoint. Historically, the disease was thought to be largely restricted to young females, temporally related to the late stages of pregnancy or early post-partum period. Shared placental and pituitary antigens have been implicated in these cases. An association with other autoimmune diseases has been reported in up to 50% of cases. Numerous studies have tried to pinpoint specific autoantibodies against pituitary or hypothalamic antigens (summarised in (144)). However, these assays are at present not as robust as those for other autoimmune diseases. Recent associations of lymphocytic hypophysitis with IgG4 disease and immune modulatory cancer therapies have led to novel insights. Hypophysitis in IgG4-related, multifocal systemic autoimmune disease is characterised by a relative increase of the plasma-cell population in the inflammatory infiltrate (145). These plasma cells are polyclonal and a significant proportion can be stained by monoclonal antibodies against IgG4. Patients may have raised serum IgG4 levels and coexistent lesions in other organs. The hypophysitis is exquisitely sensitive to steroids and surgery should be avoided (145). The administration of Ipilimumab, a blocker of T cell inhibitory molecule CTLA-4 that is successfully used in immunomodulatory therapy of advanced malignancies such as melanoma, induces lymphocytic hypophysitis in up to 4% of patients. Direct study of human pituitary tissue and experimental CTLA-4 blockade in mice suggested that off-target blockade of CTLA-4 expressed by pituitary endocrine cells, mostly lactotrophs and thyrotrophs, triggers inflammation (146). Specifically, binding of Ipilimumab to endocrine cells resulted in deposition of complement components, triggering a type II hypersensitivity reaction. In patients this was followed by production of anti-pituitary serum antibodies (146).

 

GRANULOMATOUS HYPOPHYSITIS

Definition

Inflammation of the pituitary gland characterised by the presence of well-formed granulomas with giant cells.

Pathology

The gland may be friable and swollen. Microscopically lymphoplasmacytic inflammation is associated with epithelioid histiocytes and multinucleated giant cells. Necrosis may or may not be present.

 

Pathogenesis

Granulomatous hypophysitis represents not a single entity and is even less common than lymphocytic hypophysitis. It may be idiopathic (primary), or a (secondary) manifestation of a systemic granulomatous disorder such as sarcoidosis or Wegener’s granulomatosis. Infectious aetiologies include tuberculosis, syphilis and fungal disease. The relationship of the idiopathic form to lymphocytic hypophysitis remains unclear. A recent review of 82 published cases noted a female sex bias but significantly later age at presentation than for lymphocytic hypophysitis (147). The authors speculate that idiopathic granulomatous hypophysitis may represent a chronic or late-stage manifestation of (initially subclinical) lymphocytic hypophysitis.

 

REFERENCES CHAPTER 3B

 

  1. Ezzat S, Asa SL, Couldwell WT, Barr CE, Dodge WE, Vance ML, McCutcheon IE. The prevalence of pituitary adenomas: a systematic review. Cancer 2004; 101:613-619
  2. Laws ER, Jr., Lopes MB. The new WHO classification of pituitary tumors: highlights and areas of controversy. Acta neuropathologica 2006; 111:80-81
  3. Zada G, Woodmansee WW, Ramkissoon S, Amadio J, Nose V, Laws ER, Jr. Atypical pituitary adenomas: incidence, clinical characteristics, and implications. Journal of neurosurgery 2011; 114:336-344
  4. Al-Shraim M, Asa SL. The 2004 World Health Organization classification of pituitary tumors: what is new? Acta Neuropathol 2006; 111:1-7
  5. Zada G, Lin N, Laws ER, Jr. Patterns of extrasellar extension in growth hormone-secreting and nonfunctional pituitary macroadenomas. Neurosurg Focus 2010; 29:E4
  6. Bakhtiar Y, Hirano H, Arita K, Yunoue S, Fujio S, Tominaga A, Sakoguchi T, Sugiyama K, Kurisu K, Yasufuku-Takano J, Takano K. Relationship between cytokeratin staining patterns and clinico-pathological features in somatotropinomae. Eur J Endocrinol 2010; 163:531-539
  7. Bhayana S, Booth GL, Asa SL, Kovacs K, Ezzat S. The implication of somatotroph adenoma phenotype to somatostatin analog responsiveness in acromegaly. J Clin Endocrinol Metab 2005; 90:6290-6295
  8. Larkin S, Reddy R, Karavitaki N, Cudlip S, Wass J, Ansorge O. Granulation pattern, but not GSP or GHR mutation, is associated with clinical characteristics in somatostatin-naive patients with somatotroph adenomas. Eur J Endocrinol 2013; 168:491-499
  9. Mazal PR, Czech T, Sedivy R, Aichholzer M, Wanschitz J, Klupp N, Budka H. Prognostic relevance of intracytoplasmic cytokeratin pattern, hormone expression profile, and cell proliferation in pituitary adenomas of akromegalic patients. Clin Neuropathol 2001; 20:163-171
  10. Obari A, Sano T, Ohyama K, Kudo E, Qian ZR, Yoneda A, Rayhan N, Mustafizur Rahman M, Yamada S. Clinicopathological features of growth hormone-producing pituitary adenomas: difference among various types defined by cytokeratin distribution pattern including a transitional form. Endocr Pathol 2008; 19:82-91
  11. Spada A, Arosio M, Bochicchio D, Bazzoni N, Vallar L, Bassetti M, Faglia G. Clinical, biochemical, and morphological correlates in patients bearing growth hormone-secreting pituitary tumors with or without constitutively active adenylyl cyclase. J Clin Endocrinol Metab 1990; 71:1421-1426
  12. Vallar L, Spada A, Giannattasio G. Altered Gs and adenylate cyclase activity in human GH-secreting pituitary adenomas. Nature 1987; 330:566-568
  13. Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, Vallar L. GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 1989; 340:692-696
  14. Adams EF, Brockmeier S, Friedmann E, Roth M, Buchfelder M, Fahlbusch R. Clinical and biochemical characteristics of acromegalic patients harboring gsp-positive and gsp-negative pituitary tumors. Neurosurgery 1993; 33:198-203; discussion 203
  15. Fougner SL, Casar-Borota O, Heck A, Berg JP, Bollerslev J. Adenoma granulation pattern correlates with clinical variables and effect of somatostatin analogue treatment in a large series of patients with acromegaly. Clin Endocrinol (Oxf) 2012; 76:96-102
  16. Lyons J, Landis CA, Harsh G, Vallar L, Grunewald K, Feichtinger H, Duh QY, Clark OH, Kawasaki E, Bourne HR, et al. Two G protein oncogenes in human endocrine tumors. Science 1990; 249:655-659
  17. Yang I, Park S, Ryu M, Woo J, Kim S, Kim J, Kim Y, Choi Y. Characteristics of gsp-positive growth hormone-secreting pituitary tumors in Korean acromegalic patients. Eur J Endocrinol 1996; 134:720-726
  18. Freda PU, Chung WK, Matsuoka N, Walsh JE, Kanibir MN, Kleinman G, Wang Y, Bruce JN, Post KD. Analysis of GNAS mutations in 60 growth hormone secreting pituitary tumors: correlation with clinical and pathological characteristics and surgical outcome based on highly sensitive GH and IGF-I criteria for remission. Pituitary 2007; 10:275-282
  19. Taboada GF, Luque RM, Neto LV, Machado Ede O, Sbaffi BC, Domingues RC, Marcondes JB, Chimelli LM, Fontes R, Niemeyer P, de Carvalho DP, Kineman RD, Gadelha MR. Quantitative analysis of somatostatin receptor subtypes (1-5) gene expression levels in somatotropinomas and correlation to in vivo hormonal and tumor volume responses to treatment with octreotide LAR. Eur J Endocrinol 2008; 158:295-303
  20. Lania A, Persani L, Ballare E, Mantovani S, Losa M, Spada A. Constitutively active Gs alpha is associated with an increased phosphodiesterase activity in human growth hormone-secreting adenomas. J Clin Endocrinol Metab 1998; 83:1624-1628
  21. Peri A, Conforti B, Baglioni-Peri S, Luciani P, Cioppi F, Buci L, Corbetta S, Ballare E, Serio M, Spada A. Expression of cyclic adenosine 3',5'-monophosphate (cAMP)-responsive element binding protein and inducible-cAMP early repressor genes in growth hormone-secreting pituitary adenomas with or without mutations of the Gsalpha gene. J Clin Endocrinol Metab 2001; 86:2111-2117
  22. Mayr B, Buslei R, Theodoropoulou M, Stalla GK, Buchfelder M, Schofl C. Molecular and functional properties of densely and sparsely granulated GH-producing pituitary adenomas. European journal of endocrinology / European Federation of Endocrine Societies 2013; 169:391-400
  23. Jeffery PL, Murray RE, Yeh AH, McNamara JF, Duncan RP, Francis GD, Herington AC, Chopin LK. Expression and function of the ghrelin axis, including a novel preproghrelin isoform, in human breast cancer tissues and cell lines. Endocr Relat Cancer 2005; 12:839-850
  24. Chen JH, Huang SM, Chen CC, Tsai CF, Yeh WL, Chou SC, Hsieh WT, Lu DY. Ghrelin induces cell migration through GHS-R, CaMKII, AMPK and NF-kappaB signaling pathway in glioma cells. J Cell Biochem 2011;
  25. Gnanapavan S, Kola B, Bustin SA, Morris DG, McGee P, Fairclough P, Bhattacharya S, Carpenter R, Grossman AB, Korbonits M. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J Clin Endocrinol Metab 2002; 87:2988
  26. Korbonits M, Bustin SA, Kojima M, Jordan S, Adams EF, Lowe DG, Kangawa K, Grossman AB. The expression of the growth hormone secretagogue receptor ligand ghrelin in normal and abnormal human pituitary and other neuroendocrine tumors. J Clin Endocrinol Metab 2001; 86:881-887
  27. Kim K, Sanno N, Arai K, Takano K, Yasufuku-Takano J, Teramoto A, Shibasaki T. Ghrelin mRNA and GH secretagogue receptor mRNA in human GH-producing pituitary adenomas is affected by mutations in the alpha subunit of G protein. Clin Endocrinol (Oxf) 2003; 59:630-636
  28. Xu T, Ye F, Wang B, Tian C, Wang S, Shu K, Guo D, Lei T. Elevation of growth hormone secretagogue receptor type 1a mRNA expression in human growth hormone-secreting pituitary adenoma harboring G protein alpha subunit mutation. Neuro Endocrinol Lett 2010; 31:147-154
  29. Plockinger U, Albrecht S, Mawrin C, Saeger W, Buchfelder M, Petersenn S, Schulz S. Selective loss of somatostatin receptor 2 in octreotide-resistant growth hormone-secreting adenomas. J Clin Endocrinol Metab 2008; 93:1203-1210
  30. Kato M, Inoshita N, Sugiyama T, Tani Y, Shichiri M, Sano T, Yamada S, Hirata Y. Differential expression of genes related to drug responsiveness between sparsely and densely granulated somatotroph adenomas. Endocr J 2011;
  31. Chinezu L, Vasiljevic A, Jouanneau E, Francois P, Borda A, Trouillas J, Raverot G. Expression of somatostatin receptors, SSTR2A and SSTR5, in 108 endocrine pituitary tumors using immunohistochemical detection with new specific monoclonal antibodies. Human pathology 2014; 45:71-77
  32. Fougner SL, Borota OC, Berg JP, Hald JK, Ramm-Pettersen J, Bollerslev J. The clinical response to somatostatin analogues in acromegaly correlates to the somatostatin receptor subtype 2a protein expression of the adenoma. Clin Endocrinol (Oxf) 2008; 68:458-465
  33. Cazabat L, Libe R, Perlemoine K, Rene-Corail F, Burnichon N, Gimenez-Roqueplo AP, Dupasquier-Fediaevsky L, Bertagna X, Clauser E, Chanson P, Bertherat J, Raffin-Sanson ML. Germline inactivating mutations of the aryl hydrocarbon receptor-interacting protein gene in a large cohort of sporadic acromegaly: mutations are found in a subset of young patients with macroadenomas. Eur J Endocrinol 2007; 157:1-8
  34. Chahal HS, Stals K, Unterlander M, Balding DJ, Thomas MG, Kumar AV, Besser GM, Atkinson AB, Morrison PJ, Howlett TA, Levy MJ, Orme SM, Akker SA, Abel RL, Grossman AB, Burger J, Ellard S, Korbonits M. AIP mutation in pituitary adenomas in the 18th century and today. N Engl J Med 2011; 364:43-50
  35. Daly AF, Vanbellinghen JF, Khoo SK, Jaffrain-Rea ML, Naves LA, Guitelman MA, Murat A, Emy P, Gimenez-Roqueplo AP, Tamburrano G, Raverot G, Barlier A, De Herder W, Penfornis A, Ciccarelli E, Estour B, Lecomte P, Gatta B, Chabre O, Sabate MI, Bertagna X, Garcia Basavilbaso N, Stalldecker G, Colao A, Ferolla P, Wemeau JL, Caron P, Sadoul JL, Oneto A, Archambeaud F, Calender A, Sinilnikova O, Montanana CF, Cavagnini F, Hana V, Solano A, Delettieres D, Luccio-Camelo DC, Basso A, Rohmer V, Brue T, Bours V, Teh BT, Beckers A. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab 2007; 92:1891-1896
  36. Georgitsi M, Raitila A, Karhu A, Tuppurainen K, Makinen MJ, Vierimaa O, Paschke R, Saeger W, van der Luijt RB, Sane T, Robledo M, De Menis E, Weil RJ, Wasik A, Zielinski G, Lucewicz O, Lubinski J, Launonen V, Vahteristo P, Aaltonen LA. Molecular diagnosis of pituitary adenoma predisposition caused by aryl hydrocarbon receptor-interacting protein gene mutations. Proc Natl Acad Sci U S A 2007; 104:4101-4105
  37. Igreja S, Chahal HS, King P, Bolger GB, Srirangalingam U, Guasti L, Chapple JP, Trivellin G, Gueorguiev M, Guegan K, Stals K, Khoo B, Kumar AV, Ellard S, Grossman AB, Korbonits M, International FC. Characterization of aryl hydrocarbon receptor interacting protein (AIP) mutations in familial isolated pituitary adenoma families. Human mutation 2010; 31:950-960
  38. Jennings JE, Georgitsi M, Holdaway I, Daly AF, Tichomirowa M, Beckers A, Aaltonen LA, Karhu A, Cameron FJ. Aggressive pituitary adenomas occurring in young patients in a large Polynesian kindred with a germline R271W mutation in the AIP gene. Eur J Endocrinol 2009; 161:799-804
  39. Leontiou CA, Gueorguiev M, van der Spuy J, Quinton R, Lolli F, Hassan S, Chahal HS, Igreja SC, Jordan S, Rowe J, Stolbrink M, Christian HC, Wray J, Bishop-Bailey D, Berney DM, Wass JA, Popovic V, Ribeiro-Oliveira A, Jr., Gadelha MR, Monson JP, Akker SA, Davis JR, Clayton RN, Yoshimoto K, Iwata T, Matsuno A, Eguchi K, Musat M, Flanagan D, Peters G, Bolger GB, Chapple JP, Frohman LA, Grossman AB, Korbonits M. The role of the aryl hydrocarbon receptor-interacting protein gene in familial and sporadic pituitary adenomas. J Clin Endocrinol Metab 2008; 93:2390-2401
  40. Vargiolu M, Fusco D, Kurelac I, Dirnberger D, Baumeister R, Morra I, Melcarne A, Rimondini R, Romeo G, Bonora E. The tyrosine kinase receptor RET interacts in vivo with aryl hydrocarbon receptor-interacting protein to alter survivin availability. J Clin Endocrinol Metab 2009; 94:2571-2578
  41. Vierimaa O, Georgitsi M, Lehtonen R, Vahteristo P, Kokko A, Raitila A, Tuppurainen K, Ebeling TM, Salmela PI, Paschke R, Gundogdu S, De Menis E, Makinen MJ, Launonen V, Karhu A, Aaltonen LA. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science 2006; 312:1228-1230
  42. Preda V, Korbonits M, Cudlip S, Karavitaki N, Grossman AB. Low rate of germline AIP mutations in patients with apparently sporadic pituitary adenomas before the age of 40: a single-centre adult cohort. Eur J Endocrinol 2014; 171:659-666
  43. Schofl C, Honegger J, Droste M, Grussendorf M, Finke R, Plockinger U, Berg C, Willenberg HS, Lammert A, Klingmuller D, Jaursch-Hancke C, Tonjes A, Schneidewind S, Flitsch J, Bullmann C, Dimopoulou C, Stalla G, Mayr B, Hoeppner W, Schopohl J. Frequency of AIP gene mutations in young patients with acromegaly: a registry-based study. J Clin Endocrinol Metab 2014:jc20142094
  44. Tichomirowa MA, Barlier A, Daly AF, Jaffrain-Rea ML, Ronchi C, Yaneva M, Urban JD, Petrossians P, Elenkova A, Tabarin A, Desailloud R, Maiter D, Schurmeyer T, Cozzi R, Theodoropoulou M, Sievers C, Bernabeu I, Naves LA, Chabre O, Montanana CF, Hana V, Halaby G, Delemer B, Aizpun JI, Sonnet E, Longas AF, Hagelstein MT, Caron P, Stalla GK, Bours V, Zacharieva S, Spada A, Brue T, Beckers A. High prevalence of AIP gene mutations following focused screening in young patients with sporadic pituitary macroadenomas. Eur J Endocrinol 2011; 165:509-515
  45. Daly AF, Tichomirowa MA, Petrossians P, Heliovaara E, Jaffrain-Rea ML, Barlier A, Naves LA, Ebeling T, Karhu A, Raappana A, Cazabat L, De Menis E, Montanana CF, Raverot G, Weil RJ, Sane T, Maiter D, Neggers S, Yaneva M, Tabarin A, Verrua E, Eloranta E, Murat A, Vierimaa O, Salmela PI, Emy P, Toledo RA, Sabate MI, Villa C, Popelier M, Salvatori R, Jennings J, Longas AF, Labarta Aizpun JI, Georgitsi M, Paschke R, Ronchi C, Valimaki M, Saloranta C, De Herder W, Cozzi R, Guitelman M, Magri F, Lagonigro MS, Halaby G, Corman V, Hagelstein MT, Vanbellinghen JF, Barra GB, Gimenez-Roqueplo AP, Cameron FJ, Borson-Chazot F, Holdaway I, Toledo SP, Stalla GK, Spada A, Zacharieva S, Bertherat J, Brue T, Bours V, Chanson P, Aaltonen LA, Beckers A. Clinical characteristics and therapeutic responses in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study. J Clin Endocrinol Metab 2010; 95:E373-383
  46. Jaffrain-Rea ML, Rotondi S, Turchi A, Occhi G, Barlier A, Peverelli E, Rostomyan L, Defilles C, Angelini M, Oliva MA, Ceccato F, Maiorani O, Daly AF, Esposito V, Buttarelli F, Figarella-Branger D, Giangaspero F, Spada A, Scaroni C, Alesse E, Beckers A. Somatostatin analogues increase AIP expression in somatotropinomas, irrespective of Gsp mutations. Endocr Relat Cancer 2013; 20:753-766
  47. Pagotto U, Arzberger T, Theodoropoulou M, Grubler Y, Pantaloni C, Saeger W, Losa M, Journot L, Stalla GK, Spengler D. The expression of the antiproliferative gene ZAC is lost or highly reduced in nonfunctioning pituitary adenomas. Cancer Res 2000; 60:6794-6799
  48. Chahal HS, Trivellin G, Leontiou CA, Alband N, Fowkes RC, Tahir A, Igreja SC, Chapple JP, Jordan S, Lupp A, Schulz S, Ansorge O, Karavitaki N, Carlsen E, Wass JA, Grossman AB, Korbonits M. Somatostatin analogs modulate AIP in somatotroph adenomas: the role of the ZAC1 pathway. J Clin Endocrinol Metab 2012; 97:E1411-1420
  49. Theodoropoulou M, Zhang J, Laupheimer S, Paez-Pereda M, Erneux C, Florio T, Pagotto U, Stalla GK. Octreotide, a somatostatin analogue, mediates its antiproliferative action in pituitary tumor cells by altering phosphatidylinositol 3-kinase signaling and inducing Zac1 expression. Cancer Res 2006; 66:1576-1582
  50. Trivellin G, Daly AF, Faucz FR, Yuan B, Rostomyan L, Larco DO, Schernthaner-Reiter MH, Szarek E, Leal LF, Caberg JH, Castermans E, Villa C, Dimopoulos A, Chittiboina P, Xekouki P, Shah N, Metzger D, Lysy PA, Ferrante E, Strebkova N, Mazerkina N, Zatelli MC, Lodish M, Horvath A, de Alexandre RB, Manning AD, Levy I, Keil MF, Sierra Mde L, Palmeira L, Coppieters W, Georges M, Naves LA, Jamar M, Bours V, Wu TJ, Choong CS, Bertherat J, Chanson P, Kamenicky P, Farrell WE, Barlier A, Quezado M, Bjelobaba I, Stojilkovic SS, Wess J, Costanzi S, Liu P, Lupski JR, Beckers A, Stratakis CA. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med 2014; 371:2363-2374
  51. Iacovazzo D, Caswell R, Bunce B, Jose S, Yuan B, Hernandez-Ramirez LC, Kapur S, Caimari F, Evanson J, Ferrau F, Dang MN, Gabrovska P, Larkin SJ, Ansorge O, Rodd C, Vance ML, Ramirez-Renteria C, Mercado M, Goldstone AP, Buchfelder M, Burren CP, Gurlek A, Dutta P, Choong CS, Cheetham T, Trivellin G, Stratakis CA, Lopes MB, Grossman AB, Trouillas J, Lupski JR, Ellard S, Sampson JR, Roncaroli F, Korbonits M. Germline or somatic GPR101 duplication leads to X-linked acrogigantism: a clinico-pathological and genetic study. Acta neuropathologica communications 2016; 4:56
  52. Kamenicky P, Bouligand J, Chanson P. Gigantism, acromegaly, and GPR101 mutations. N Engl J Med 2015; 372:1264
  53. Bottoni A, Zatelli MC, Ferracin M, Tagliati F, Piccin D, Vignali C, Calin GA, Negrini M, Croce CM, Degli Uberti EC. Identification of differentially expressed microRNAs by microarray: a possible role for microRNA genes in pituitary adenomas. J Cell Physiol 2007; 210:370-377
  54. D'Angelo D, Palmieri D, Mussnich P, Roche M, Wierinckx A, Raverot G, Fedele M, Croce CM, Trouillas J, Fusco A. Altered microRNA expression profile in human pituitary GH adenomas: down-regulation of miRNA targeting HMGA1, HMGA2, and E2F1. J Clin Endocrinol Metab 2012; 97:E1128-1138
  55. Fougner SL, Lekva T, Borota OC, Hald JK, Bollerslev J, Berg JP. The expression of E-cadherin in somatotroph pituitary adenomas is related to tumor size, invasiveness, and somatostatin analog response. J Clin Endocrinol Metab 2010; 95:2334-2342
  56. Lekva T, Berg JP, Lyle R, Heck A, Ringstad G, Olstad OK, Michelsen AE, Casar-Borota O, Bollerslev J, Ueland T. Epithelial splicing regulator protein 1 and alternative splicing in somatotroph adenomas. Endocrinology 2013; 154:3331-3343
  57. Ronchi CL, Peverelli E, Herterich S, Weigand I, Mantovani G, Schwarzmayr T, Sbiera S, Allolio B, Honegger J, Appenzeller S, Lania AG, Reincke M, Calebiro D, Spada A, Buchfelder M, Flitsch J, Strom TM, Fassnacht M. Landscape of somatic mutations in sporadic GH-secreting pituitary adenomas. Eur J Endocrinol 2016; 174:363-372
  58. Valimaki N, Demir H, Pitkanen E, Kaasinen E, Karppinen A, Kivipelto L, Schalin-Jantti C, Aaltonen LA, Karhu A. Whole-Genome Sequencing of Growth Hormone (GH)-Secreting Pituitary Adenomas. J Clin Endocrinol Metab 2015; 100:3918-3927
  59. Lv H, Li C, Gui S, Zhang Y. Expression of estrogen receptor alpha and growth factors in human prolactinoma and its correlation with clinical features and gender. J Endocrinol Invest 2012; 35:174-180
  60. Font de Mora J, Brown M. AIB1 is a conduit for kinase-mediated growth factor signaling to the estrogen receptor. Molecular and cellular biology 2000; 20:5041-5047
  61. Carretero J, Blanco EJ, Carretero M, Carretero-Hernandez M, Garcia-Barrado MJ, Iglesias-Osma MC, Burks DJ, Font de Mora J. The expression of AIB1 correlates with cellular proliferation in human prolactinomas. Ann Anat 2013; 195:253-259
  62. Akinci H, Kapucu A, Dar KA, Celik O, Tutunculer B, Sirin G, Oz B, Gazioglu N, Ince H, Aliustaoglu S, Kadioglu P. Aromatase cytochrome P450 enzyme expression in prolactinomas and its relationship to tumor behavior. Pituitary 2013; 16:386-392
  63. Leung AW, Kent Morest D, Li JY. Differential BMP signaling controls formation and differentiation of multipotent preplacodal ectoderm progenitors from human embryonic stem cells. Developmental biology 2013; 379:208-220
  64. Friedman E, Adams EF, Hoog A, Gejman PV, Carson E, Larsson C, De Marco L, Werner S, Fahlbusch R, Nordenskjold M. Normal structural dopamine type 2 receptor gene in prolactin-secreting and other pituitary tumors. The Journal of clinical endocrinology and metabolism 1994; 78:568-574
  65. Pellegrini I, Rasolonjanahary R, Gunz G, Bertrand P, Delivet S, Jedynak CP, Kordon C, Peillon F, Jaquet P, Enjalbert A. Resistance to bromocriptine in prolactinomas. The Journal of clinical endocrinology and metabolism 1989; 69:500-509
  66. Zhang W, Murao K, Imachi H, Iwama H, Chen K, Fei Z, Zhang X, Ishida T, Tamiya T. Suppression of prolactin expression by cabergoline requires prolactin regulatory element-binding protein (PREB) in GH3 cells. Horm Metab Res 2010; 42:557-561
  67. Li Q, Su Z, Liu J, Cai L, Lu J, Lin S, Xiong Z, Li W, Zheng W, Wu J, Zhuge Q, Wu Z. Dopamine receptor D2S gene transfer improves the sensitivity of GH3 rat pituitary adenoma cells to bromocriptine. Mol Cell Endocrinol 2014; 382:377-384
  68. Peverelli E, Mantovani G, Vitali E, Elli FM, Olgiati L, Ferrero S, Laws ER, Della Mina P, Villa A, Beck-Peccoz P, Spada A, Lania AG. Filamin-A is essential for dopamine d2 receptor expression and signaling in tumorous lactotrophs. The Journal of clinical endocrinology and metabolism 2012; 97:967-977
  69. Missale C, Boroni F, Losa M, Giovanelli M, Zanellato A, Dal Toso R, Balsari A, Spano P. Nerve growth factor suppresses the transforming phenotype of human prolactinomas. Proceedings of the National Academy of Sciences of the United States of America 1993; 90:7961-7965
  70. Sigala S, Martocchia A, Missale C, Falaschi P, Spano P. Increased serum concentration of nerve growth factor in patients with microprolactinoma. Neuropeptides 2004; 38:21-24
  71. Facchetti M, Uberti D, Memo M, Missale C. Nerve growth factor restores p53 function in pituitary tumor cell lines via trkA-mediated activation of phosphatidylinositol 3-kinase. Molecular endocrinology 2004; 18:162-172
  72. Cooper O, Mamelak A, Bannykh S, Carmichael J, Bonert V, Lim S, Cook-Wiens G, Ben-Shlomo A. Prolactinoma ErbB receptor expression and targeted therapy for aggressive tumors. Endocrine 2013;
  73. Hock R, Furusawa T, Ueda T, Bustin M. HMG chromosomal proteins in development and disease. Trends Cell Biol 2007; 17:72-79
  74. Newey PJ, Nesbit MA, Rimmer AJ, Head RA, Gorvin CM, Attar M, Gregory L, Wass JA, Buck D, Karavitaki N, Grossman AB, McVean G, Ansorge O, Thakker RV. Whole-exome sequencing studies of nonfunctioning pituitary adenomas. J Clin Endocrinol Metab 2013; 98:E796-800
  75. Zhou W, Song Y, Xu H, Zhou K, Zhang W, Chen J, Qin M, Yi H, Gustafsson JA, Yang H, Fan X. In nonfunctional pituitary adenomas, estrogen receptors and slug contribute to development of invasiveness. J Clin Endocrinol Metab 2011; 96:E1237-1245
  76. Palmieri D, Valentino T, De Martino I, Esposito F, Cappabianca P, Wierinckx A, Vitiello M, Lombardi G, Colao A, Trouillas J, Pierantoni GM, Fusco A, Fedele M. PIT1 upregulation by HMGA proteins has a role in pituitary tumorigenesis. Endocrine-related cancer 2012; 19:123-135
  77. Qian ZR, Li CC, Yamasaki H, Mizusawa N, Yoshimoto K, Yamada S, Tashiro T, Horiguchi H, Wakatsuki S, Hirokawa M, Sano T. Role of E-cadherin, alpha-, beta-, and gamma-catenins, and p120 (cell adhesion molecules) in prolactinoma behavior. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 2002; 15:1357-1365
  78. Gheldof A, Berx G. Cadherins and epithelial-to-mesenchymal transition. Prog Mol Biol Transl Sci 2013; 116:317-336
  79. Stemmler MP. Cadherins in development and cancer. Mol Biosyst 2008; 4:835-850
  80. Wang C, Su Z, Sanai N, Xue X, Lu L, Chen Y, Wu J, Zheng W, Zhuge Q, Wu ZB. microRNA expression profile and differentially-expressed genes in prolactinomas following bromocriptine treatment. Oncology reports 2012; 27:1312-1320
  81. Wu ZB, Li WQ, Lin SJ, Wang CD, Cai L, Lu JL, Chen YX, Su ZP, Shang HB, Yang WL, Zhao WG. MicroRNA expression profile of bromocriptine-resistant prolactinomas. Mol Cell Endocrinol 2014; 395:10-18
  82. Mindermann T, Wilson CB. Thyrotropin-producing pituitary adenomas. J Neurosurg 1993; 79:521-527
  83. Dong Q, Brucker-Davis F, Weintraub BD, Smallridge RC, Carr FE, Battey J, Spiegel AM, Shenker A. Screening of candidate oncogenes in human thyrotroph tumors: absence of activating mutations of the G alpha q, G alpha 11, G alpha s, or thyrotropin-releasing hormone receptor genes. The Journal of clinical endocrinology and metabolism 1996; 81:1134-1140
  84. Gatto F, Barbieri F, Castelletti L, Arvigo M, Pattarozzi A, Annunziata F, Saveanu A, Minuto F, Castellan L, Zona G, Florio T, Ferone D. In vivo and in vitro response to octreotide LAR in a TSH-secreting adenoma: characterization of somatostatin receptor expression and role of subtype 5. Pituitary 2011; 14:141-147
  85. Gatto F, Barbieri F, Gatti M, Wurth R, Schulz S, Ravetti JL, Zona G, Culler MD, Saveanu A, Giusti M, Minuto F, Hofland LJ, Ferone D, Florio T. Balance between somatostatin and D2 receptor expression drives TSH-secreting adenoma response to somatostatin analogues and dopastatins. Clin Endocrinol (Oxf) 2012; 76:407-414
  86. Yoshihara A, Isozaki O, Hizuka N, Nozoe Y, Harada C, Ono M, Kawamata T, Kubo O, Hori T, Takano K. Expression of type 5 somatostatin receptor in TSH-secreting pituitary adenomas: a possible marker for predicting long-term response to octreotide therapy. Endocr J 2007; 54:133-138
  87. Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald A, Beuschlein F, Meitinger T, Mizuno-Yamasaki E, Kawaguchi K, Saeki Y, Tanaka K, Wieland T, Graf E, Saeger W, Ronchi CL, Allolio B, Buchfelder M, Strom TM, Fassnacht M, Komada M. Mutations in the deubiquitinase gene USP8 cause Cushing's disease. Nature genetics 2015; 47:31-38
  88. Perez-Rivas LG, Theodoropoulou M, Ferrau F, Nusser C, Kawaguchi K, Stratakis CA, Rueda Faucz F, Wildemberg LE, Assie G, Beschorner R, Dimopoulou C, Buchfelder M, Popovic V, Berr CM, Toth M, Ardisasmita AI, Honegger J, Bertherat J, Gadelha MR, Beuschlein F, Stalla G, Komada M, Korbonits M, Reincke M. The gene of the ubiquitin-specific protease 8 is frequently mutated in adenomas causing Cushing's disease. J Clin Endocrinol Metab 2015:jc20151453
  89. Ma ZY, Song ZJ, Chen JH, Wang YF, Li SQ, Zhou LF, Mao Y, Li YM, Hu RG, Zhang ZY, Ye HY, Shen M, Shou XF, Li ZQ, Peng H, Wang QZ, Zhou DZ, Qin XL, Ji J, Zheng J, Chen H, Wang Y, Geng DY, Tang WJ, Fu CW, Shi ZF, Zhang YC, Ye Z, He WQ, Zhang QL, Tang QS, Xie R, Shen JW, Wen ZJ, Zhou J, Wang T, Huang S, Qiu HJ, Qiao ND, Zhang Y, Pan L, Bao WM, Liu YC, Huang CX, Shi YY, Zhao Y. Recurrent gain-of-function USP8 mutations in Cushing's disease. Cell research 2015; 25:306-317
  90. Hayashi K, Inoshita N, Kawaguchi K, Ibrahim Ardisasmita A, Suzuki H, Fukuhara N, Okada M, Nishioka H, Takeuchi Y, Komada M, Takeshita A, Yamada S. The USP8 mutational status may predict drug susceptibility in corticotroph adenomas of Cushing's disease. Eur J Endocrinol 2016; 174:213-226
  91. Tani Y, Inoshita N, Sugiyama T, Kato M, Yamada S, Shichiri M, Hirata Y. Upregulation of CDKN2A and suppression of cyclin D1 gene expressions in ACTH-secreting pituitary adenomas. Eur J Endocrinol 2010; 163:523-529
  92. Korbonits M, Bujalska I, Shimojo M, Nobes J, Jordan S, Grossman AB, Stewart PM. Expression of 11 beta-hydroxysteroid dehydrogenase isoenzymes in the human pituitary: induction of the type 2 enzyme in corticotropinomas and other pituitary tumors. J Clin Endocrinol Metab 2001; 86:2728-2733
  93. Ebisawa T, Tojo K, Tajima N, Kamio M, Oki Y, Ono K, Sasano H. Immunohistochemical analysis of 11-beta-hydroxysteroid dehydrogenase type 2 and glucocorticoid receptor in subclinical Cushing's disease due to pituitary macroadenoma. Endocr Pathol 2008; 19:252-260
  94. Tateno T, Kato M, Tani Y, Oyama K, Yamada S, Hirata Y. Differential expression of somatostatin and dopamine receptor subtype genes in adrenocorticotropin (ACTH)-secreting pituitary tumors and silent corticotroph adenomas. Endocrine journal 2009; 56:579-584
  95. Gentilin E, Tagliati F, Filieri C, Mole D, Minoia M, Rosaria Ambrosio M, Degli Uberti EC, Zatelli MC. miR-26a plays an important role in cell cycle regulation in ACTH-secreting pituitary adenomas by modulating protein kinase Cdelta. Endocrinology 2013; 154:1690-1700
  96. Michaelis KA, Knox AJ, Xu M, Kiseljak-Vassiliades K, Edwards MG, Geraci M, Kleinschmidt-DeMasters BK, Lillehei KO, Wierman ME. Identification of growth arrest and DNA-damage-inducible gene beta (GADD45beta) as a novel tumor suppressor in pituitary gonadotrope tumors. Endocrinology 2011; 152:3603-3613
  97. Butz H, Liko I, Czirjak S, Igaz P, Korbonits M, Racz K, Patocs A. MicroRNA profile indicates downregulation of the TGFbeta pathway in sporadic non-functioning pituitary adenomas. Pituitary 2011; 14:112-124
  98. Miao Z, Miao Y, Lin Y, Lu X. Overexpression of the Notch3 receptor in non-functioning pituitary tumours. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia 2012; 19:107-110
  99. Lu R, Gao H, Wang H, Cao L, Bai J, Zhang Y. Overexpression of the Notch3 receptor and its ligand Jagged1 in human clinically non-functioning pituitary adenomas. Oncology letters 2013; 5:845-851
  100. Karga HJ, Alexander JM, Hedley-Whyte ET, Klibanski A, Jameson JL. Ras mutations in human pituitary tumors. J Clin Endocrinol Metab 1992; 74:914-919
  101. Ruebel KH, Leontovich AA, Jin L, Stilling GA, Zhang H, Qian X, Nakamura N, Scheithauer BW, Kovacs K, Lloyd RV. Patterns of gene expression in pituitary carcinomas and adenomas analyzed by high-density oligonucleotide arrays, reverse transcriptase-quantitative PCR, and protein expression. Endocrine 2006; 29:435-444
  102. de Kock L, Sabbaghian N, Plourde F, Srivastava A, Weber E, Bouron-Dal Soglio D, Hamel N, Choi JH, Park SH, Deal CL, Kelsey MM, Dishop MK, Esbenshade A, Kuttesch JF, Jacques TS, Perry A, Leichter H, Maeder P, Brundler MA, Warner J, Neal J, Zacharin M, Korbonits M, Cole T, Traunecker H, McLean TW, Rotondo F, Lepage P, Albrecht S, Horvath E, Kovacs K, Priest JR, Foulkes WD. Pituitary blastoma: a pathognomonic feature of germ-line DICER1 mutations. Acta Neuropathol 2014; 128:111-122
  103. Scheithauer BW, Horvath E, Abel TW, Robital Y, Park SH, Osamura RY, Deal C, Lloyd RV, Kovacs K. Pituitary blastoma: a unique embryonal tumor. Pituitary 2012; 15:365-373
  104. Buslei R, Nolde M, Hofmann B, Meissner S, Eyupoglu IY, Siebzehnrubl F, Hahnen E, Kreutzer J, Fahlbusch R. Common mutations of beta-catenin in adamantinomatous craniopharyngiomas but not in other tumours originating from the sellar region. Acta Neuropathol 2005; 109:589-597
  105. Sekine S, Shibata T, Kokubu A, Morishita Y, Noguchi M, Nakanishi Y, Sakamoto M, Hirohashi S. Craniopharyngiomas of adamantinomatous type harbor beta-catenin gene mutations. Am J Pathol 2002; 161:1997-2001
  106. Brastianos PK, Taylor-Weiner A, Manley PE, Jones RT, Dias-Santagata D, Thorner AR, Lawrence MS, Rodriguez FJ, Bernardo LA, Schubert L, Sunkavalli A, Shillingford N, Calicchio ML, Lidov HG, Taha H, Martinez-Lage M, Santi M, Storm PB, Lee JY, Palmer JN, Adappa ND, Scott RM, Dunn IF, Laws ER, Jr., Stewart C, Ligon KL, Hoang MP, Van Hummelen P, Hahn WC, Louis DN, Resnick AC, Kieran MW, Getz G, Santagata S. Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas. Nature genetics 2014; 46:161-165
  107. Liu C, Li Y, Semenov M, Han C, Baeg GH, Tan Y, Zhang Z, Lin X, He X. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 2002; 108:837-847
  108. Oikonomou E, Barreto DC, Soares B, De Marco L, Buchfelder M, Adams EF. Beta-catenin mutations in craniopharyngiomas and pituitary adenomas. J Neurooncol 2005; 73:205-209
  109. Hofmann BM, Kreutzer J, Saeger W, Buchfelder M, Blumcke I, Fahlbusch R, Buslei R. Nuclear beta-catenin accumulation as reliable marker for the differentiation between cystic craniopharyngiomas and rathke cleft cysts: a clinico-pathologic approach. Am J Surg Pathol 2006; 30:1595-1603
  110. Larkin SJ, Preda V, Karavitaki N, Grossman A, Ansorge O. BRAF V600E mutations are characteristic for papillary craniopharyngioma and may coexist with CTNNB1-mutated adamantinomatous craniopharyngioma. Acta Neuropathol 2014; 127:927-929
  111. Gaston-Massuet C, Andoniadou CL, Signore M, Sajedi E, Bird S, Turner JM, Martinez-Barbera JP. Genetic interaction between the homeobox transcription factors HESX1 and SIX3 is required for normal pituitary development. Developmental biology 2008; 324:322-333
  112. Gaston-Massuet C, Andoniadou CL, Signore M, Jayakody SA, Charolidi N, Kyeyune R, Vernay B, Jacques TS, Taketo MM, Le Tissier P, Dattani MT, Martinez-Barbera JP. Increased Wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans. Proc Natl Acad Sci U S A 2011; 108:11482-11487
  113. Andoniadou CL, Gaston-Massuet C, Reddy R, Schneider RP, Blasco MA, Le Tissier P, Jacques TS, Pevny LH, Dattani MT, Martinez-Barbera JP. Identification of novel pathways involved in the pathogenesis of human adamantinomatous craniopharyngioma. Acta Neuropathol 2012;
  114. Hassanein AM, Glanz SM, Kessler HP, Eskin TA, Liu C. beta-Catenin is expressed aberrantly in tumors expressing shadow cells. Pilomatricoma, craniopharyngioma, and calcifying odontogenic cyst. Am J Clin Pathol 2003; 120:732-736
  115. Holsken A, Kreutzer J, Hofmann BM, Hans V, Oppel F, Buchfelder M, Fahlbusch R, Blumcke I, Buslei R. Target gene activation of the Wnt signaling pathway in nuclear beta-catenin accumulating cells of adamantinomatous craniopharyngiomas. Brain Pathol 2009; 19:357-364
  116. Garcia-Lavandeira M, Saez C, Diaz-Rodriguez E, Perez-Romero S, Senra A, Dieguez C, Japon MA, Alvarez CV. Craniopharyngiomas express embryonic stem cell markers (SOX2, OCT4, KLF4, and SOX9) as pituitary stem cells but do not coexpress RET/GFRA3 receptors. J Clin Endocrinol Metab 2012; 97:E80-87
  117. Holsken A, Buchfelder M, Fahlbusch R, Blumcke I, Buslei R. Tumour cell migration in adamantinomatous craniopharyngiomas is promoted by activated Wnt-signalling. Acta Neuropathol 2010; 119:631-639
  118. Holsken A, Gebhardt M, Buchfelder M, Fahlbusch R, Blumcke I, Buslei R. EGFR signaling regulates tumor cell migration in craniopharyngiomas. Clin Cancer Res 2011; 17:4367-4377
  119. Yamashiro S, Yamakita Y, Ono S, Matsumura F. Fascin, an actin-bundling protein, induces membrane protrusions and increases cell motility of epithelial cells. Molecular biology of the cell 1998; 9:993-1006
  120. Brastianos PK, Shankar GM, Gill CM, Taylor-Weiner A, Nayyar N, Panka DJ, Sullivan RJ, Frederick DT, Abedalthagafi M, Jones PS, Dunn IF, Nahed BV, Romero JM, Louis DN, Getz G, Cahill DP, Santagata S, Curry WT, Jr., Barker FG, 2nd. Dramatic Response of BRAF V600E Mutant Papillary Craniopharyngioma to Targeted Therapy. Journal of the National Cancer Institute 2016; 108
  121. Aylwin SJ, Bodi I, Beaney R. Pronounced response of papillary craniopharyngioma to treatment with vemurafenib, a BRAF inhibitor. Pituitary 2016; 19:544-546
  122. Crotty TB, Scheithauer BW, Young WF, Jr., Davis DH, Shaw EG, Miller GM, Burger PC. Papillary craniopharyngioma: a clinicopathological study of 48 cases. J Neurosurg 1995; 83:206-214
  123. Shin JL, Asa SL, Woodhouse LJ, Smyth HS, Ezzat S. Cystic lesions of the pituitary: clinicopathological features distinguishing craniopharyngioma, Rathke's cleft cyst, and arachnoid cyst. J Clin Endocrinol Metab 1999; 84:3972-3982
  124. Okada T, Fujitsu K, Ichikawa T, Mukaihara S, Miyahara K, Kaku S, Uryuu Y, Niino H, Yagishita S, Shiina T. Coexistence of adamantinomatous and squamous-papillary type craniopharyngioma: Case report and discussion of etiology and pathology. Neuropathology 2011;
  125. Harrison MJ, Morgello S, Post KD. Epithelial cystic lesions of the sellar and parasellar region: a continuum of ectodermal derivatives? J Neurosurg 1994; 80:1018-1025
  126. Oka H, Kawano N, Yagishita S, Kobayashi I, Saegusa H, Fujii K. Ciliated craniopharyngioma indicates histogenetic relationship to Rathke cleft epithelium. Clin Neuropathol 1997; 16:103-106
  127. Goodrich JT, Post KD, Duffy P. Ciliated craniopharyngioma. Surg Neurol 1985; 24:105-111
  128. Park YS, Ahn JY, Kim DS, Kim TS, Kim SH. Late development of craniopharyngioma following surgery for Rathke's cleft cyst. Clin Neuropathol 2009; 28:177-181
  129. Sato K, Oka H, Utsuki S, Kondo K, Kurata A, Fujii K. Ciliated craniopharyngioma may arise from Rathke cleft cyst. Clin Neuropathol 2006; 25:25-28
  130. Berx G, Cleton-Jansen AM, Nollet F, de Leeuw WJ, van de Vijver M, Cornelisse C, van Roy F. E-cadherin is a tumour/invasion suppressor gene mutated in human lobular breast cancers. The EMBO journal 1995; 14:6107-6115
  131. Mao J, Wang J, Liu B, Pan W, Farr GH, 3rd, Flynn C, Yuan H, Takada S, Kimelman D, Li L, Wu D. Low-density lipoprotein receptor-related protein-5 binds to Axin and regulates the canonical Wnt signaling pathway. Mol Cell 2001; 7:801-809
  132. Schweizer L, Capper D, Holsken A, Fahlbusch R, Flitsch J, Buchfelder M, Herold-Mende C, von Deimling A, Buslei R. BRAF V600E analysis for the differentiation of papillary craniopharyngiomas and Rathke's cleft cysts. Neuropathology and applied neurobiology 2015; 41:733-742
  133. Takei Y, Seyama S, Pearl GS, Tindall GT. Ultrastructural study of the human neurohypophysis. II. Cellular elements of neural parenchyma, the pituicytes. Cell and tissue research 1980; 205:273-287
  134. Brat DJ, Scheithauer BW, Staugaitis SM, Holtzman RN, Morgello S, Burger PC. Pituicytoma: a distinctive low-grade glioma of the neurohypophysis. Am J Surg Pathol 2000; 24:362-368
  135. Roncaroli F, Scheithauer BW, Cenacchi G, Horvath E, Kovacs K, Lloyd RV, Abell-Aleff P, Santi M, Yates AJ. 'Spindle cell oncocytoma' of the adenohypophysis: a tumor of folliculostellate cells? Am J Surg Pathol 2002; 26:1048-1055
  136. Phillips JJ, Misra A, Feuerstein BG, Kunwar S, Tihan T. Pituicytoma: characterization of a unique neoplasm by histology, immunohistochemistry, ultrastructure, and array-based comparative genomic hybridization. Arch Pathol Lab Med 2010; 134:1063-1069
  137. Mete O, Lopes MB, Asa SL. Spindle cell oncocytomas and granular cell tumors of the pituitary are variants of pituicytoma. Am J Surg Pathol 2013; 37:1694-1699
  138. Fukushima S, Otsuka A, Suzuki T, Yanagisawa T, Mishima K, Mukasa A, Saito N, Kumabe T, Kanamori M, Tominaga T, Narita Y, Shibui S, Kato M, Shibata T, Matsutani M, Nishikawa R, Ichimura K, Intracranial Germ Cell Tumor Genome Analysis C. Mutually exclusive mutations of KIT and RAS are associated with KIT mRNA expression and chromosomal instability in primary intracranial pure germinomas. Acta Neuropathol 2014; 127:911-925
  139. Wang L, Yamaguchi S, Burstein MD, Terashima K, Chang K, Ng HK, Nakamura H, He Z, Doddapaneni H, Lewis L, Wang M, Suzuki T, Nishikawa R, Natsume A, Terasaka S, Dauser R, Whitehead W, Adekunle A, Sun J, Qiao Y, Marth G, Muzny DM, Gibbs RA, Leal SM, Wheeler DA, Lau CC. Novel somatic and germline mutations in intracranial germ cell tumours. Nature 2014; 511:241-245
  140. Murray MJ, Bartels U, Nishikawa R, Fangusaro J, Matsutani M, Nicholson JC. Consensus on the management of intracranial germ-cell tumours. The Lancet Oncology 2015; 16:e470-477
  141. Stacchiotti S, Sommer J, Chordoma Global Consensus G. Building a global consensus approach to chordoma: a position paper from the medical and patient community. The Lancet Oncology 2015; 16:e71-83
  142. Yang XR, Ng D, Alcorta DA, Liebsch NJ, Sheridan E, Li S, Goldstein AM, Parry DM, Kelley MJ. T (brachyury) gene duplication confers major susceptibility to familial chordoma. Nature genetics 2009; 41:1176-1178
  143. Pillay N, Plagnol V, Tarpey PS, Lobo SB, Presneau N, Szuhai K, Halai D, Berisha F, Cannon SR, Mead S, Kasperaviciute D, Palmen J, Talmud PJ, Kindblom LG, Amary MF, Tirabosco R, Flanagan AM. A common single-nucleotide variant in T is strongly associated with chordoma. Nature genetics 2012; 44:1185-1187
  144. Falorni A, Minarelli V, Bartoloni E, Alunno A, Gerli R. Diagnosis and classification of autoimmune hypophysitis. Autoimmunity reviews 2014; 13:412-416
  145. Leporati P, Landek-Salgado MA, Lupi I, Chiovato L, Caturegli P. IgG4-related hypophysitis: a new addition to the hypophysitis spectrum. J Clin Endocrinol Metab 2011; 96:1971-1980
  146. Iwama S, De Remigis A, Callahan MK, Slovin SF, Wolchok JD, Caturegli P. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Science translational medicine 2014; 6:230ra245
  147. Hunn BH, Martin WG, Simpson S, Jr., McLean CA. Idiopathic granulomatous hypophysitis: a systematic review of 82 cases in the literature. Pituitary 2014; 17:357-365

Development And Microscopic Anatomy Of The Pituitary Gland

ABSTRACT

The pituitary is an organ dual of origin. The anterior lobe (adenohypophysis) is epithelial in origin, whereas the posterior lobe (neurohypophysis) derives from the neural ectoderm. Precise spatial and temporal co-ordination of transcription factor expression in both structures is critical for pituitary formation and the differentiation of hormone-producing cells. Disruption of this regulation, for instance by mutation, can lead to numerous developmental disorders. We provide an overview of the molecular drivers of pituitary organogenesis and illustrate the anatomy and histology of the mature pituitary, comprising adenohypophysis (anterior lobe), neurohypophysis (posterior lobe), pars intermedia and infundibulum (pituitary stalk). For complete coverage of this and related areas of Endocrinology, please visit our free web –book, www.endotext.org.

PITUITARY ORANOGENESIS

The pituitary is an organ of dual origin. The anterior lobe (adenohypophysis) is derived from oral ectoderm and is epithelial in origin, whereas the posterior lobe (neurohypophysis) derives from the neural ectoderm. The composite nature of the pituitary requires that the neural and oral ectoderm interact physically and developmentally. Precise spatial and temporal co-ordination and regulation of signals from both structures is critical for pituitary formation and the differentiation of the various hormone-producing cell types in the anterior lobe. The expression of transcription factors that control cell lineage commitment in the developing anterior lobe must be precisely regulated to ensure correct differentiation of hormone-producing cell types; the iterative and cumulative nature of this regulation renders it extremely sensitive to perturbation. Disruption of this process, for instance by mutation, can lead to numerous developmental disorders from congenital forms of hypopituitarism to pituitary tumours. Pituitary organogenesis is covered briefly below. For further description, please see Chapter 3c (Functional anatomy of the hypothalamus and pituitary Ronald M. Lechan, and Roberto Toni).

Pituitary organogenesis begins during week 4 of fetal development. A thickening of cells in the oral ectoderm form the hypophyseal placode, which gives rise to Rathke’s pouch, an upward evagination that extends towards the neural ectoderm. At the same time, a downward extension of the ventral diencephalon forms the posterior lobe and the two nascent lobes connect to form the composite structure of the adult pituitary. Rathke’s pouch constricts at its base and eventually separates altogether from the oral epithelium during week 6-8. The cells of the anterior wall of Rathke’s pouch undergo extensive proliferation to form the anterior lobe while the posterior wall proliferates more slowly to form the vestigial (in humans) intermediate lobe. Cell patterning and terminal differentiation occurs within the anterior lobe to form the five principal specialised endocrine cell types of the pituitary gland.

Transcriptional Control of Pituitary Organogenesis

Development of the pituitary occurs broadly in three stages:

  1. Initiation of pituitary organogenesis and formation of Rathke’s pouch
  2. Evagination of Rathke’s pouch and cell proliferation
  3. Lineage determination and cellular differentiation

Much of our understanding of the process of pituitary organogenesis comes from mouse studies, but the phenotypes associated with human disorders often share aspects with mouse models of defective pituitary development. Various transcription factors involved are presented in Table 3a-1. Reviewed extensively in (1-3).

Table 3a-1: Signalling molecules controlling pituitary organogenesis and associated dysfunction

Developmental Stage Factor Function Dysfunction Comments Reference
Initiation of pituitary organogenesis and formation of Rathke’s pouch SIX homeodomain proteins Six1-Six6

Family of six transcriptional activators/ inhibitors

Functional role difficult to determine due to redundancy and severity of mutations

  Expression persists in adult pituitary; may mediate plasticity (4,5)
Paired-like homeobox proteins Hesx1

Transcriptional repressor

Early marker of Rathke’s pouch

Downregulation essential for endocrine cell differentiation

Mutations in patients with hypopituitarism including septo-optic dysplasia, combined pituitary hormone deficiency (CPHD) and isolated growth hormone deficiency (IGHD) Expression activated by LIM homeodomain proteins (6)
Otx2 Transcription factor that regulates Hesx1

Mutations found in patients with ocular disorders (e.g. anophthalmia, microphthalmia) with or without hypopituitarism. In mice, deficiency results in craniofacial

defects and pituitary gland dysmorphology, but normal pituitary cell specification

  (7,8)
Pitx1/2/3 Interacts with various other factors to determine cell lineage Mutations in Pitx2 (R91P) found in patients with Axenfeld-Rieger syndrome. Blocks expression of LH β and FSHβ

Expressed throughout oral ectoderm and Rathke’s pouch.

 

Some functional redundancy, but all required for proper development

 

Expression maintained in the adult gland

(9)
LIM homeodomain transcription factors Isl1

Involved in cell lineage specification

 

 

No human mutations identified. Null mice do not develop Rathke’s pouch

First LIM protein to be expressed

 

Expressed in cells destined to become thyrotrophs

Reviewed in (1)
Lhx3 Expression gradient required for differentiation of endocrine cell types Heritable mutation in patients with CPHD with short, stiff neck and sensorineural hearing loss Broad temporal and spatial expression pattern with many target genes (10)
Lhx4 Expression gradient required for differentiation of endocrine cell types Heterozygous mutations in patients with CPHD. Associated with pituitary hypoplasia, small sella and Arnold-Chiari malformation Not critical for endocrine cell differentiation (11,12)
SOX2  

Expressed throughout developing Rathke’s pouch

 

Downregulation essential for endocrine cell differentiation

Mutations found in patients with an- or microphthalmia, hypogonadotrophic hypogonadism, and growth hormone deficiency (GHD)

 

Both duplications and loss of function mutations associated with hypopituitarism

 

Some expression retained in adult pituitary, confined to pituitary progenitor/stem cells (13,14)
β-catenin   Signalling activates Pitx2 expression promoting pituitary precursor proliferation. Required for Pit1 lineage determination and anterior pituitary formation

Premature activation of β-catenin results in Hesx1 repression and pituitary gland agenesis in mouse

Activating mutation of β-catenin leads to pituitary progenitor proliferation, loss of Pit1 lineage cells and adamantinomatous cranyiopharyngioma

High degree of interaction with other signalling pathways e.g. Notch. Not required for cell lineage determination (15,16)

Notch

 

 

Mediates lateral inhibition and cell lineage determination

 

Activates Hes1 expression

Dysregulaiton of the pathway associated with premature corticotroph differentiation and pituitary hypoplasia in mice Expression in the adult gland co-localises with SOX2 (17)

Migration of Rathke’s pouch cells and proliferation

 

Bone morphogenic proteins BMP4

Expressed in ventral diencephalon

 

Required for hypophyseal placode formation

Downregulation results in arrested development of Rathke’s pouch in mice   Reviewed in (1)
BMP2

Induces Isl1 expression

 

Downregulation required for cell differentiation

Prolonged expression results in hyperplastic pituitary and lack of terminal differentiation   (18)
Fibroblast growth factors FGF 8, 10, 18

Expressed in the posterior pituitary

 

Required for Lhx3 and Lhx4 expression and cell differentiation

In humans, mutations of FGF8 and its receptor are associated with Kallmann syndrome, resulting in isolated hypogonadotrophic hypogonadism   (19-21)
Shh  

Expressed in oral ectoderm and ventral diencephalon

 

Induces Lhx3 expression

Antagonism in mouse oral ectoderm results in hypoplastic Rathke’s pouch   (22)
Lineage determination and cellular differentiation Prop1  

Transcriptional activator and suppressor depending on context

 

Activates POU1F1 expression and switches developmental process from proliferation to differentiation

Mutations are most common cause of CPHD in humans   Reviewed in (1) and (23) (24-27)
POU1F1 (Pit1)  

Expressed in cells committed to somatotroph, lactotroph and thyrotroph lineage

 

Inhibits GATA2 and prevents gonadotroph cell fate

Mutations in humans associated with GH PRL, TSH deficiency and small anterior pituitary. Mutations rarely present in sporadic CPHD and more common in familial CPHD Required for GH PRL, TSHβ expression (26,28-30)
GATA2  

Specifies gonadotroph and thyrotroph lineages

 

Induces expression of Nr5a1

In mice, overexpression associated with gonadotroph and thyrotroph hypoplasia. Expression persists in adult gland (28)
Nr5a1 (SF1)  

Expressed throughout adrenal and reproductive axes

 

Regulates expression of GnRHR, LH, FSH and αGSU.

 

Expression necessary for gonadotroph differentiation

 

Mutations associated with 46XY sex reversal with adrenal failure, 46XY gonadal dysgenesis and 46XX ovarian insufficiency and premature ovarian failure in humans   (31,32)
Tbx19 (TPIT)   Activates POMC expression in association with PITX1 Mutations are commonest cause of isolated ACTH deficiency in humans Antagonists to Nr5a1 can prevent gonadotroph cell fate (33,34)

Figure 3a-1: Signalling molecules and transcription factors control pituitary development. Arrows represent regulation of expression in the direction indicated.

ANATOMY AND HISTOLOGY OF THE MATURE PITUITARY GLAND

Macroscopic Anatomy

The pituitary gland, or hypophysis cerebri, is an oval body approximately 12mm in transverse and 8mm in anterior-posterior diameter weighing approximately 500mg (Gray’s anatomy p380, 39th edition). The anterior lobe of the pituitary is generally smaller in men than women, and nullipara than multipara; during pregnancy the gland may increase by approximately 30% due to lactotroph hyperplasia. The hypophysis is connected to the brain via the infundibulum, a tubular structure arising from the tuber cinereum and median eminence of the hypothalamus. The gland rests in the sella turcica (pituitary fossa) of the sphenoid bone and is covered superiorly by the diaphragma sellae (dura), laterally by the wall of the cavernous sinus, and antero-inferiorly by the posterior wall of the sphenoid sinus, which is used as the standard route for pituitary surgery (transsphenoidal adenectomy (TSA)). Antero-superior the pituitary lies in close proximity to the optic chiasm; this explains why space-occupying lesions of the pituitary commonly present with bitemporal hemianopia.

Figure 3a-2: Coronal slice through the human brain at the level of the pituitary gland (left, MRI; right, post-mortem in situ appearance).

Figure 3a-3: Sagittal histological section through the brain illustrating the anatomical relationship of the pituitary gland to other structures at the base of the human brain (AH = adenohypophysis, NH = neurohypophysis, PS = pituitary stalk, OC = optic chiasm). This figure was published in Surgical pathology of the nervous system and its coverings 4th Edition, Burger, Scheithauer and Vogel, Chapter 9 Region of the sella turcica page 438, Copyright Elsevier (2002).

NORMAL HISTOLOGY OF THE PITUITARY

Adenohypophysis (Pars Anterior, Anterior Lobe)

The pituitary gland in adults has a distinct histological appearance, reflecting its divergent origin. The adenohypophysis (pars anterior, anterior lobe) is characterised by well-demarcated acini that usually contain a mixture of different hormone-producing cells. This nested pattern is best appreciated in reticulin preparations, which delineate the borders of the acini. The cellular heterogeneity within acini may be demonstrated histochemically in PAS-OG or preparations which were widely used before the adoption of antibody-based stains. Corticotroph cells are generally strongly basophilic (PAS-positive), somatotroph and lactotroph cells mostly acidophilic (orangeophilic), whilst gonadotrophs and thyrotrophs may be basophilic or chromophobe (reacting with neither acid nor basic stains). There is no perfect match of hormone-expression and type or degree of chromophilia; some chromophobe cells may represent degranulated chromophil cells or precursor cells (Gray’s anatomy p380, 39th edition). In a normal adult adenohypophysis approximately 10% of endocrine cells are basophils, 40% acidophils and 50% chromophobes. The PAS-OG stain is still a useful supplementary method in the differential diagnosis of some pituitary lesions (hyperplasia, corticotroph microadenoma, Crooke’s cell change or adenoma). Although most acini contain a mixture of different hormone-producing cells, there is evidence of zonation. The lateral wings of the gland mostly contain somatotrophs and lactotrophs, whilst corticotrophs are concentrated in the median mucoid wedge, which at its anterior border (the rostral tip) harbours clusters of thyrotrophs. Gonadotroph (LH/FSH) cells are diffusely scattered throughout the gland.

Figure 3a-4: Axial section of the anterior lobe of the human pituitary gland (adenohypophysis) at the level indicated by the dashed blue line in the diagram. Although there is a mixture of different hormone producing cells in most pituitary acini, the distribution of cells is not random: this is most pronounced in the ‘lateral wings’, which contain mostly somatotroph cells and the central ‘mucoid wedge’, which contains the majority of the corticotrophs. This is easily appreciated in periodic acid-Schiff / orange-G (PAS-OG) histochemistry, which stains somatotrophs yellow-orange (OG-positive) and corticotrophs purple (PAS-positive).

Follicular-stellate (FS) cells are (in the adult human pituitary) an agranular (non-hormone-producing) parenchymal component of the pars anterior. It has been postulated that they represent a stem cell capable of trans-differentiation into endocrine cells (35), but whether this is true in humans remains to be seen. FS cells are small, chromophobe, with slender processes that extend between the endocrine cells. They form small follicles at the centre of acini, comprised of apical tips of multiple FS cells. They may be visualised with S100 and GFAP antibodies, but their expression pattern is not always overlapping and may reflect different stages of maturation or function. In our hands, annexin-1 immunohistochemistry is a robust marker of FS cells. Annexin 1 (ANXA1) is a member of the annexin family of phospholipid- and calcium-binding proteins. ANXA1-positive FS cells may modulate glucocorticoid feedback loops in the anterior gland (36) or act as antigen-presenting cells.

Figure 3a-5: Non-endocrine cells of the anterior lobe include small folliculo-stellate (FS) cells with delicately branching processes that invest endocrine cells (left, annexin-1 staining) and (on the right) very rare cells that are postulated to represent adult pituitary stem cells (PSC, nestin staining).

PARS INTERMEDIA

In contrast to rodents, the pars intermedia is rudimentary in adult humans. It represents a narrow zone between the adeno- and neurohypophysis often containing microscopic remnants of Rathke’s cleft. This zone may also contain scattered intensely PAS-positive corticotrophs, which may extend from the mucoid wedge of the adenohypophysis into the neurohypophysis. This so-called “basophil invasion” must not be confused with corticotroph microadenomas; it is believed to increase with aging and it has been suggested that these basophil cells are functionally distinct from classical ACTH-producing cells of the adenohypophysis and do not respond with hyaline degeneration (“Crooke’s cell change”) in the setting of systemic hypercortisolaemia.

Figure 3a-6: Axial section of the human pituitary gland at the level of the vestigial intermediate lobe (approximately representing the boxed area in the diagram). Note the cluster of remnants of Rathke’s pouch / cleft (RC). The arcs indicate the posterior (centre) and postero-medial (left and right) edges of the mucoid wedge (with scattered basophils) and pituitary wings (with scattered somatotrophs), respectively. The asterisk indicates basophil corticotrophs ‘spilling’ into the neurohypophysis (‘basophil invasion’, see figure 3a-7).

Figure 3a-7: ‘Basophil invasion’ of corticotrophs from the vestigial pars intermedia into the neurohypophysis (pars posterior of the pituitary gland). The dashed line represents the border between pars intermedia and pars posterior.

NEUROHYPOPHYSIS (Pars Posterior, Posterior Lobe)

The neurohypophysis does not contain neuroendocrine epithelial cells. Instead, it is composed of the axons arising from groups of hypothalamic neurons, most prominently those originating from magnocellular neurons of the supraoptic and paraventricular nuclei. They form the hypothalamo-hypophyseal tract and their terminals end near the sinusoids of the posterior lobe. The neurosecretory granules mostly contain oxytocin or vasopressin and form axonal beads close to their termini. Whilst it is believed that normal astrocytes may populate (at least partially) the infundibulum, the axon terminals in the neurohypophysis are supported by so-called pituicytes, which are characterised by the expression of the TTF-1 transcription factor (absent in classic GFAP-positive astrocytes). These cells show elongated processes often running in parallel with axons, and demonstrate only patchy GFAP and S100 expression.

Figure 3a-8: The cytoarchitecture of the neurophypophysis (right) is strikingly different from the adenohypophysis, which contains the nested (inset, left) collection of endocrine cells (left). The neurohypophysis does not contain neurosecretory cell bodies; instead it is composed of specialised glial cells (pituicytes) that – unique in the human brain – express the TTF-1 (thyroid transcription factor 1) protein in their nuclei (inset, top right). The neurohypophysis contains the nerve endings of the oxytocin and vasopressin producing cells of the hypothalamus. Their large distended nerve endings can be identified on routine stains as so-called Herring-bodies (arrow), named after Percy Theodore Herring (University of Edinburgh) who described them in 1908 as the ‘physiologically active principle’ of the posterior gland.

INFUNDIBULUM (Pituitary Stalk)

The stalk is a tubular (funnel-shaped) structure divided into the anterior pars tuberalis and posterior pars infundibularis. The pars tuberalis is considered to be part of the adenohypophysis and contains a few scattered gonadotroph or corticotroph cells. It surrounds anteriorly and superficially the pars infundibularis (infundibular stem), which contains the unmyelinated axons of the magnocellular supraoptic and paraventricular neurons. Large intraaxonal accumulations of oxytocin and vasopressin may be seen as eosinophilic ovoid granular swellings along the trajectory of these axons in the infundibular stem. These structures are called “Herring bodies”. Another notable feature of the rostral portion of the stalk are tortuous capillary loops surrounding a central capillary, termed gomitoli (see example page 327, Histology for pathologists, 3rd edition). These are part of the complex hypothalamo-hypophyseal portal system.

 

REFERENCES

  1. Kelberman D, Rizzoti K, Lovell-Badge R, Robinson IC, Dattani MT. Genetic regulation of pituitary gland development in human and mouse. Endocr Rev 2009; 30:790-829
  2. Davis SW, Castinetti F, Carvalho LR, Ellsworth BS, Potok MA, Lyons RH, Brinkmeier ML, Raetzman LT, Carninci P, Mortensen AH, Hayashizaki Y, Arnhold IJ, Mendonca BB, Brue T, Camper SA. Molecular mechanisms of pituitary organogenesis: In search of novel regulatory genes. Mol Cell Endocrinol 2010; 323:4-19
  3. Zhu X, Gleiberman AS, Rosenfeld MG. Molecular physiology of pituitary development: signaling and transcriptional networks. Physiological reviews 2007; 87:933-963
  4. Gaston-Massuet C, Andoniadou CL, Signore M, Sajedi E, Bird S, Turner JM, Martinez-Barbera JP. Genetic interaction between the homeobox transcription factors HESX1 and SIX3 is required for normal pituitary development. Developmental biology 2008; 324:322-333
  5. Aijaz S, Clark BJ, Williamson K, van Heyningen V, Morrison D, Fitzpatrick D, Collin R, Ragge N, Christoforou A, Brown A, Hanson I. Absence of SIX6 mutations in microphthalmia, anophthalmia, and coloboma. Investigative ophthalmology & visual science 2004; 45:3871-3876
  6. Durmaz B, Cogulu O, Dizdarer C, Stobbe H, Pfaeffle R, Ozkinay F. A novel homozygous HESX1 mutation causes panhypopituitarism without midline defects and optic nerve anomalies. Journal of pediatric endocrinology & metabolism : JPEM 2011; 24:779-782
  7. Mortensen AH, Schade V, Lamonerie T, Camper SA. Deletion of OTX2 in neural ectoderm delays anterior pituitary development. Human molecular genetics 2015; 24:939-953
  8. Tajima T, Ishizu K, Nakamura A. Molecular and Clinical Findings in Patients with LHX4 and OTX2 Mutations. Clinical pediatric endocrinology : case reports and clinical investigations : official journal of the Japanese Society for Pediatric Endocrinology 2013; 22:15-23
  9. Suh H, Gage PJ, Drouin J, Camper SA. Pitx2 is required at multiple stages of pituitary organogenesis: pituitary primordium formation and cell specification. Development 2002; 129:329-337
  10. Pfaeffle RW, Savage JJ, Hunter CS, Palme C, Ahlmann M, Kumar P, Bellone J, Schoenau E, Korsch E, Bramswig JH, Stobbe HM, Blum WF, Rhodes SJ. Four novel mutations of the LHX3 gene cause combined pituitary hormone deficiencies with or without limited neck rotation. J Clin Endocrinol Metab 2007; 92:1909-1919
  11. Machinis K, Pantel J, Netchine I, Leger J, Camand OJ, Sobrier ML, Dastot-Le Moal F, Duquesnoy P, Abitbol M, Czernichow P, Amselem S. Syndromic short stature in patients with a germline mutation in the LIM homeobox LHX4. American journal of human genetics 2001; 69:961-968
  12. Pfaeffle RW, Hunter CS, Savage JJ, Duran-Prado M, Mullen RD, Neeb ZP, Eiholzer U, Hesse V, Haddad NG, Stobbe HM, Blum WF, Weigel JF, Rhodes SJ. Three novel missense mutations within the LHX4 gene are associated with variable pituitary hormone deficiencies. J Clin Endocrinol Metab 2008; 93:1062-1071
  13. Kelberman D, de Castro SC, Huang S, Crolla JA, Palmer R, Gregory JW, Taylor D, Cavallo L, Faienza MF, Fischetto R, Achermann JC, Martinez-Barbera JP, Rizzoti K, Lovell-Badge R, Robinson IC, Gerrelli D, Dattani MT. SOX2 plays a critical role in the pituitary, forebrain, and eye during human embryonic development. J Clin Endocrinol Metab 2008; 93:1865-1873
  14. Kelberman D, Rizzoti K, Avilion A, Bitner-Glindzicz M, Cianfarani S, Collins J, Chong WK, Kirk JM, Achermann JC, Ross R, Carmignac D, Lovell-Badge R, Robinson IC, Dattani MT. Mutations within Sox2/SOX2 are associated with abnormalities in the hypothalamo-pituitary-gonadal axis in mice and humans. J Clin Invest 2006; 116:2442-2455
  15. Kioussi C, Briata P, Baek SH, Rose DW, Hamblet NS, Herman T, Ohgi KA, Lin C, Gleiberman A, Wang J, Brault V, Ruiz-Lozano P, Nguyen HD, Kemler R, Glass CK, Wynshaw-Boris A, Rosenfeld MG. Identification of a Wnt/Dvl/beta-Catenin --> Pitx2 pathway mediating cell-type-specific proliferation during development. Cell 2002; 111:673-685
  16. Gaston-Massuet C, Andoniadou CL, Signore M, Jayakody SA, Charolidi N, Kyeyune R, Vernay B, Jacques TS, Taketo MM, Le Tissier P, Dattani MT, Martinez-Barbera JP. Increased Wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans. Proc Natl Acad Sci U S A 2011; 108:11482-11487
  17. Kita A, Imayoshi I, Hojo M, Kitagawa M, Kokubu H, Ohsawa R, Ohtsuka T, Kageyama R, Hashimoto N. Hes1 and Hes5 control the progenitor pool, intermediate lobe specification, and posterior lobe formation in the pituitary development. Mol Endocrinol 2007; 21:1458-1466
  18. Treier M, Gleiberman AS, O'Connell SM, Szeto DP, McMahon JA, McMahon AP, Rosenfeld MG. Multistep signaling requirements for pituitary organogenesis in vivo. Genes & development 1998; 12:1691-1704
  19. Falardeau J, Chung WC, Beenken A, Raivio T, Plummer L, Sidis Y, Jacobson-Dickman EE, Eliseenkova AV, Ma J, Dwyer A, Quinton R, Na S, Hall JE, Huot C, Alois N, Pearce SH, Cole LW, Hughes V, Mohammadi M, Tsai P, Pitteloud N. Decreased FGF8 signaling causes deficiency of gonadotropin-releasing hormone in humans and mice. J Clin Invest 2008; 118:2822-2831
  20. McCabe MJ, Gaston-Massuet C, Tziaferi V, Gregory LC, Alatzoglou KS, Signore M, Puelles E, Gerrelli D, Farooqi IS, Raza J, Walker J, Kavanaugh SI, Tsai PS, Pitteloud N, Martinez-Barbera JP, Dattani MT. Novel FGF8 mutations associated with recessive holoprosencephaly, craniofacial defects, and hypothalamo-pituitary dysfunction. J Clin Endocrinol Metab 2011; 96:E1709-1718
  21. Dode C, Levilliers J, Dupont JM, De Paepe A, Le Du N, Soussi-Yanicostas N, Coimbra RS, Delmaghani S, Compain-Nouaille S, Baverel F, Pecheux C, Le Tessier D, Cruaud C, Delpech M, Speleman F, Vermeulen S, Amalfitano A, Bachelot Y, Bouchard P, Cabrol S, Carel JC, Delemarre-van de Waal H, Goulet-Salmon B, Kottler ML, Richard O, Sanchez-Franco F, Saura R, Young J, Petit C, Hardelin JP. Loss-of-function mutations in FGFR1 cause autosomal dominant Kallmann syndrome. Nature genetics 2003; 33:463-465
  22. Treier M, O'Connell S, Gleiberman A, Price J, Szeto DP, Burgess R, Chuang PT, McMahon AP, Rosenfeld MG. Hedgehog signaling is required for pituitary gland development. Development 2001; 128:377-386
  23. Kelberman D, Dattani MT. Role of transcription factors in midline central nervous system and pituitary defects. Endocrine development 2009; 14:67-82
  24. Cogan JD, Wu W, Phillips JA, 3rd, Arnhold IJ, Agapito A, Fofanova OV, Osorio MG, Bircan I, Moreno A, Mendonca BB. The PROP1 2-base pair deletion is a common cause of combined pituitary hormone deficiency. J Clin Endocrinol Metab 1998; 83:3346-3349
  25. Deladoey J, Fluck C, Buyukgebiz A, Kuhlmann BV, Eble A, Hindmarsh PC, Wu W, Mullis PE. "Hot spot" in the PROP1 gene responsible for combined pituitary hormone deficiency. J Clin Endocrinol Metab 1999; 84:1645-1650
  26. Turton JP, Mehta A, Raza J, Woods KS, Tiulpakov A, Cassar J, Chong K, Thomas PQ, Eunice M, Ammini AC, Bouloux PM, Starzyk J, Hindmarsh PC, Dattani MT. Mutations within the transcription factor PROP1 are rare in a cohort of patients with sporadic combined pituitary hormone deficiency (CPHD). Clin Endocrinol (Oxf) 2005; 63:10-18
  27. Vieira TC, Boldarine VT, Abucham J. Molecular analysis of PROP1, PIT1, HESX1, LHX3, and LHX4 shows high frequency of PROP1 mutations in patients with familial forms of combined pituitary hormone deficiency. Arquivos brasileiros de endocrinologia e metabologia 2007; 51:1097-1103
  28. Dasen JS, O'Connell SM, Flynn SE, Treier M, Gleiberman AS, Szeto DP, Hooshmand F, Aggarwal AK, Rosenfeld MG. Reciprocal interactions of Pit1 and GATA2 mediate signaling gradient-induced determination of pituitary cell types. Cell 1999; 97:587-598
  29. McLennan K, Jeske Y, Cotterill A, Cowley D, Penfold J, Jones T, Howard N, Thomsett M, Choong C. Combined pituitary hormone deficiency in Australian children: clinical and genetic correlates. Clin Endocrinol (Oxf) 2003; 58:785-794
  30. Snabboon T, Plengpanich W, Buranasupkajorn P, Khwanjaipanich R, Vasinanukorn P, Suwanwalaikorn S, Khovidhunkit W, Shotelersuk V. A novel germline mutation, IVS4+1G>A, of the POU1F1 gene underlying combined pituitary hormone deficiency. Horm Res 2008; 69:60-64
  31. Ingraham HA, Lala DS, Ikeda Y, Luo X, Shen WH, Nachtigal MW, Abbud R, Nilson JH, Parker KL. The nuclear receptor steroidogenic factor 1 acts at multiple levels of the reproductive axis. Genes & development 1994; 8:2302-2312
  32. Shinoda K, Lei H, Yoshii H, Nomura M, Nagano M, Shiba H, Sasaki H, Osawa Y, Ninomiya Y, Niwa O, et al. Developmental defects of the ventromedial hypothalamic nucleus and pituitary gonadotroph in the Ftz-F1 disrupted mice. Developmental dynamics : an official publication of the American Association of Anatomists 1995; 204:22-29
  33. Pulichino AM, Vallette-Kasic S, Couture C, Gauthier Y, Brue T, David M, Malpuech G, Deal C, Van Vliet G, De Vroede M, Riepe FG, Partsch CJ, Sippell WG, Berberoglu M, Atasay B, Drouin J. Human and mouse TPIT gene mutations cause early onset pituitary ACTH deficiency. Genes & development 2003; 17:711-716
  34. Vallette-Kasic S, Brue T, Pulichino AM, Gueydan M, Barlier A, David M, Nicolino M, Malpuech G, Dechelotte P, Deal C, Van Vliet G, De Vroede M, Riepe FG, Partsch CJ, Sippell WG, Berberoglu M, Atasay B, de Zegher F, Beckers D, Kyllo J, Donohoue P, Fassnacht M, Hahner S, Allolio B, Noordam C, Dunkel L, Hero M, Pigeon B, Weill J, Yigit S, Brauner R, Heinrich JJ, Cummings E, Riddell C, Enjalbert A, Drouin J. Congenital isolated adrenocorticotropin deficiency: an underestimated cause of neonatal death, explained by TPIT gene mutations. J Clin Endocrinol Metab 2005; 90:1323-1331
  35. Horvath E, Kovacs K. Folliculo-stellate cells of the human pituitary: a type of adult stem cell? Ultrastructural pathology 2002; 26:219-228
  36. John CD, Theogaraj E, Christian HC, Morris JF, Smith SF, Buckingham JC. Time-specific effects of perinatal glucocorticoid treatment on anterior pituitary morphology, annexin 1 expression and adrenocorticotrophic hormone secretion in the adult female rat. J Neuroendocrinol 2006; 18:949-959

Skeletal Dysplasias

ABSTRACT

Skeletal dysplasias form a complex group of more than 400 conditions with extraordinary clinical and molecular heterogeneity. Their classification changes as we learn about their molecular bases. After a brief introduction to the evaluation of the short child, this chapter is structured according to the 2010 nosology and classification of genetic skeletal disorders and is not intended to detail each rare skeletal dysplasia. Rather, it aims to familiarize the reader with this classification, so that the clinician will be able to determine in which category of conditions to place an affected individual and thus establish a differential diagnosis. We then describe the clinical and radiological manifestations of some of the more common skeletal dysplasias in each group.

Introduction

Skeletal dysplasias form a complex group of more than 400 conditions with extraordinary clinical and molecular heterogeneity. Their classification changes as we learn about their molecular bases. After a brief introduction to the evaluation of the short child, this chapter is structured according to the 2010 nosology and classification of genetic skeletal disorders (1) and is not intended to detail each rare skeletal dysplasia. Rather, it aims to familiarize the reader with this classification, so that the clinician will be able to determine in which category of conditions to place an affected individual and thus establish a differential diagnosis. In the following chapter, we describe the clinical and radiological manifestations of some of the more common skeletal dysplasias in each group. The table for each section lists, when available, the inheritance pattern, the gene, and the OMIM number. General references used include OMIM (www.omim.org), Genereviews  (GR, www.ncbi.nlm.nih.gov/books/1116/), Orphanet (www.orpha.net), and chapters or manuscripts by Dr. Spranger (2, 3) and Dr. Lachman (4). For genetic testing, clinicians are encouraged to refer to the Genetic Testing Registry (http://www.ncbi.nlm.nih.gov/gtr/) and their local geneticist.

EVALUATION OF THE SHORT CHILD

The first step is to analyze the growth curve of the child, compare it to an ethnicity-appropriate reference and the growth history of the parents. After a thorough familial and clinical history and examination, treatable endocrine and common conditions should be considered. Namely, if there is proportionate short stature with increased weight-for-height ratio, one needs to consider growth hormone deficiency or insensitivity, hypothyroidism, or glucocorticoid excess. Work-up could include measuring bone age, IGF1, IGFBP3, T4, TSH. A karyotype, GH, GHBP, GHRH and ACTH may be indicated. If there is proportionate short stature with decreased weight-for-height ratio, one needs to consider undernutrition or malnutrition, malabsorption, or a chronic systemic disease. Work-up depends on history and physical examination, but may include a complete blood count with sedimentation rate (for inflammatory bowel disease) and serum tissue transglutaminase (for celiac disease), serum electrolytes and a first-void morning urinalysis (for renal tubular acidosis or nephrogenic diabetes insipidus). A more detailed discussion can be found in a review by Rose et al.(5) and other chapters in Endotext.

SKELETAL DYSPLASIA CLASSIFICATION

The first 8 groups of conditions in the 2010 nosology are separated according to the molecular basis of the disease: FGFR3, type 2 collagen, type 11 collagen, sulfation disorders, perlecan, aggrecan, filamin, and TRPV4. The other 32 groups are organized according to their clinical and radiographic presentation. The prefix acro- refers to the extremities (hands and feet), meso- to the middle portion (ulna and radius, tibia and fibula), rhizo- to the proximal portion (femur and humerus), spondylo- to the spine, epi- to the epiphyses, and meta- to the metaphyses. For example, if only the hands and feet are shorter, one would consult the acromelic group of conditions, whereas if the spine and metaphyses are affected, one would consult the spondylometaphyseal dysplasias. Listed below are the 40 groups of conditions to be detailed in this chapter.

Groups of conditions organized according to their molecular bases

  1. FGFR3 chondrodysplasia group
  2. Type 2 collagen group and similar disorders
  3. Type 11 collagen group
  4. Sulfation disorders group
  5. Perlecan group
  6. Aggrecan group
  7. Filamin group and related disorders
  8. TRPV4 group

Groups of conditions organized according to their clinical presentations

  1. Short-ribs dysplasias (with or without polydactyly) group
  2. Multiple epiphyseal dysplasia and pseudoachondroplasia group
  3. Metaphyseal dysplasias
  4. Spondylometaphyseal dysplasias (SMD)
  5. Spondylo-epi-(meta)-physeal dysplasias (SE(M)D)
  6. Severe spondylodysplastic dysplasias
  7. Acromelic dysplasias (extremities of the limbs)
  8. Acromesomelic dysplasias (extremities and middle portion of the limbs)
  9. Mesomelic and rhizo-mesomelic dysplasias (proximal and middle portions of the limbs)
  10. Bent bones dysplasias
  11. Slender bone dysplasia group
  12. Dysplasias with multiple joint dislocations
  13. Chondrodysplasia punctata (CDP) group
  14. Neonatal osteosclerotic dysplasias
  15. Increased bone density group (without modification of bone shape)
  16. Increased bone density group with metaphyseal and/or diaphyseal involvement
  17. Osteogenesis imperfecta and decreased bone density group
  18. Abnormal mineralization group
  19. Lysosomal storage diseases with skeletal involvement (dysostosis multiplex group)
  20. Osteolysis group
  21. Disorganized development of skeletal components group
  22. Overgrowth syndromes with skeletal involvement
  23. Genetic inflammatory/rheumatoid-like osteoarthropathies
  24. Cleidocranial dysplasia and isolated cranial ossification defects group
  25. Craniosynostosis syndromes
  26. Dysostoses with predominant craniofacial involvement
  27. Dysostoses with predominant vertebral with and without costal involvement
  28. Patellar dysostoses
  29. Brachydactylies (with or without extraskeletal manifestations)
  30. Limb hypoplasia—reduction defects group
  31. Polydactyly-Syndactyly-Triphalangism group
  32. Defects in joint formation and synostoses

    1. FGFR3 chondrodysplasia group

Thanatophoric dysplasia (thus named because it often results in early death) is characterized by micromelia with bowed femurs, short ribs, narrow thorax, macrocephaly, distinctive facial features, brachydactyly, hypotonia. Radiographically, there is rhizomelic shortening of the long bones with irregular metaphyses, platyspondyly, small foramen magnum with brain stem compression, bowed femurs (TD type I) and cloverleaf skull (always in TD type II; sometimes in TD type I). CNS abnormalities include temporal lobe malformations, hydrocephaly, brain stem hypoplasia and neuronal migration abnormalities.

Figure 1. Thanatophoric dysplasia type 1. Severe platyspondyly, very short ribs narrow thorax, short broad pelvis, large skull, very short and bent long bones.

Achondroplasia is characterized by small stature with rhizomelia and redundant skin folds, limitation of elbow extension and genu varum, short fingers with trident configuration of the hands. Craniocervical junction compression is a major complication which may occur and requires surveillance for early detection and management. There is also thoracolumbar kyphosis, lumbar lordosis, and a large head with frontal bossing with midface hypoplasia. The radiographic findings include short tubular bones with metaphyseal flaring, narrowing of the interpediculate distance of the lumbar spine, rounded ilia and horizontal acetabula, narrow sacrosciatic notch and proximal femoral radiolucency. In hypochondroplasia, there are similar but milder clinical and radiological findings, the head is large but there is no midface hypoplasia.

Figure 2. Achondroplasia. Small rounded iliac bones, horizontal acetabula, decreasing interpediculate distance, normal vertebral body height, short ribs.

Figure 3. Hypochondroplasia. decreased interpediculate distance, short broad long bones , short wide femoral necks, relative elongation of the distal fibula compare to tibia.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Thanatophoric dysplasia type 1 (TD1) AD 187600 1366 1860 FGFR3
 Thanatophoric dysplasia type 2 (TD2) AD 187601 1366 93274 FGFR3
 Severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN) AD 187600 1455 85165 FGFR3
 Achondroplasia AD 100800 1152 15 FGFR3
 Hypochondroplasia AD 146000 1477 429 FGFR3
 Camptodactyly, tall stature, and hearing loss syndrome (CATSHL) AD 610474     FGFR3

Please also refer to group 33 for craniosynostoses syndromes linked to FGFR3 mutations, as well as LADD syndrome in group 39 for another FGFR3-related phenotype.

2. TYPE 2 COLLAGEN GROUP

Stickler syndrome is characterized by ocular findings of myopia, cataract, and retinal detachment, sensorineural and conductive hearing loss, flat mala and cleft palate (alone or as part of the Robin sequence), mild spondyloepiphyseal dysplasia and early-onset arthritis (6).

Figure 4. Stickler syndrome. small epiphyses, wide femoral neck, hypoplastic iliac wings, flat epiphyses, schmorl’s nodules.

Spondyloepiphyseal dysplasia congenita (SEDC) presents with disproportionate short stature (short trunk), abnormal epiphyses, and flattened vertebral bodies. Some features of Stickler syndrome include myopia and/or retinal degeneration with retinal detachment and cleft palate.

Figure 5. sed congenita. platyspondyly, delayed epiphyseal ossification (especially femoral heads), dens hypoplasia.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Achondrogenesis type 2 (ACG2; Langer-Saldino) AD 200610   93296 COL2A1
 Platyspondylic dysplasia, Torrance type AD 151210   85166 COL2A1
 Hypochondrogenesis AD 200610   93296 COL2A1
 Spondyloepiphyseal dysplasia congenita (SEDC) AD 183900   94068 COL2A1
 Spondyloepimetaphyseal dysplasia (SEMD) Strudwick type AD 184250   93346 COL2A1
 Kniest dysplasia AD 156550   485 COL2A1
 Spondyloperipheral dysplasia AD 271700   1856 COL2A1
 Mild SED with premature onset arthrosis AD       COL2A1
 SED with metatarsal shortening (formerly Czech dysplasia) AD 609162   137678 COL2A1
 Stickler syndrome type 1 AD 108300 1302 828 COL2A1

3. TYPE 11 COLLAGEN GROUP

Marshall syndrome resembles Stickler syndrome but is characterized by a flat or retracted midface, thick calvaria, abnormal frontal sinuses with shallow orbits, intracranial calcifications, and ectodermal abnormalities including abnormal sweating and teeth.

Otospondylomegaepiphyseal dysplasia (OSMED) is characterized by sensorineural hearing loss, enlarged epiphyses, skeletal dysplasia with disproportionately short limbs, vertebral body anomalies, midface hypoplasia, a short nose with anteverted nares and a flat nasal bridge, a long philtrum, cleft palate/bifid uvula, micrognathia, and hypertelorism.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Stickler syndrome type 2 AD 604841 1302 90654 COL11A1
 Marshall syndrome AD 154780   560 COL11A1
 Fibrochondrogenesis AR 228520   2021 COL11A1
 Otospondylomegaepiphyseal dysplasia (OSMED), recessive type AR 215150   1427 COL11A2
 Otospondylomegaepiphyseal dysplasia (OSMED), dominant type (Weissenbacher-Zweymüller syndrome, Stickler syndrome type 3) AD 215150   1427 COL11A2

Please also refer to Stickler syndrome type 1 in group 2

4.  SULFATION DISORDERS GROUP

Achondrogenesis type 1B (ACG1B) is characterized extremely short limbs with short fingers and toes, hypoplasia of the thorax, protuberant abdomen, and hydropic fetal appearance. There is a normal-sized skull with a flat facies. There is a lack of ossification of the vertebral bodies (except for pedicles), short and thin ribs, and ossification of the upper part of iliac bones giving crescent-shaped appearance. Shortening of the tubular bones with metaphyseal spurring ("thorn apple" appearance) is seen.

The clinical features of diastrophic dysplasia (DTD) include limb shortening with hitchhiker thumbs, ulnar deviation of the fingers, a gap between the first and second toes, clubfeet, contractures of large joints, early-onset osteoarthritis and radial dislocation. The skull is normal-sized. There is some trunk shortening, a small chest with a protuberant abdomen and spinal deformities (scoliosis, exaggerated lumbar lordosis, cervical kyphosis). Non-skeletal findings include a cleft palate, cystic ear swelling in the neonatal period, and flat hemangiomas of the forehead.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Achondrogenesis type 1B (ACG1B) AR 600972 1516 93298 SLC26A2
 Atelosteogenesis type 2 (AO2) AR 256050 1317 56304 SLC26A2
 Diastrophic dysplasia (DTD) AR 222600 1350 628 SLC26A2
 MED, autosomal recessive type (rMED; EDM4) AR 226900 1306 93307 SLC26A2
 SEMD, PAPSS2 type AR 603005   93282 PAPSS2
 Chondrodysplasia with congenital joint dislocations, CHST3 type (recessive Larsen syndrome) AR 608637 62112 263463 CHST3
 Ehlers-Danlos syndrome, CHST14 type (“musculo-skeletal variant”) AR 601776   2953 CHST14

Please also refer to groups 7 and 26 for other conditions with multiple dislocations

5. PERLECAN GROUP

Schwartz-Jampel syndrome manifests with myotonia (characteristic facies with blepharophimosis and a puckered facial appearance) and osteoarticular abnormalities with progressive joint stiffness. There is also a flattening of the vertebral bodies, short stature, hip dysplasia, bowing of the diaphyses and irregular epiphyses.

Group/name of disorder Inher. OMIM Orpha Gene
 Dyssegmental dysplasia, Silverman-Handmaker type AR 224410 1865 HSPG2
 Dyssegmental dysplasia, Rolland-Desbuquois type AR 224400 156731 HSPG2
 Schwartz-Jampel syndrome (myotonic chondrodystrophy) AR 255800 800 HSPG2Aggrecan group

6. AGGRECAN GROUP

These conditions have been each described in one family and will not be discussed in detail here.

Group/name of disorder Inher. OMIM Orpha Gene
 SED, Kimberley type AD 608361 93283 ACAN
 SEMD, Aggrecan type AR 612813 171866 ACAN
 Familial osteochondritis dissecans AD 165800 251262 ACAN

The otopalatodigital (OPD) spectrum disorders caused by FLNA mutations include Otopalatodigital syndromes type I and II, frontometaphyseal dysplasia, Melnick-Needles syndrome and terminal osseous dysplasia with pigmentary skin defects (TODPD). Manifestations include abnormal facial features (such as widely spaced eyes), hypoplasia of the thorax, scoliosis, shortened digits, bowed long bones and joint movement limitations.

Larsen syndrome is characterized by large-joint dislocations (hip, knee, and elbow) and characteristic craniofacial abnormalities (prominent forehead, depressed nasal bridge, flattened midface, and ocular hypertelorism).  There can also be club feet (equinovarus or equinovalgus foot deformities); scoliosis and cervical kyphosis, cervical myelopathy; and spatula-shaped fingers, most marked in the thumb.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Frontometaphyseal dysplasia XLD 305620 1393 1826 FLNA
 Osteodysplasty Melnick-Needles XLD 309350 1393 2484 FLNA
 Otopalatodigital syndrome type 1 (OPD1) XLD 311300 1393 90650 FLNA
 Otopalatodigital syndrome type 2 (OPD2) XLD 304120 1393 90652 FLNA
 Terminal osseous dysplasia with pigmentary defects (TODPD) XLD 300244 1393 88630 FLNA
 Atelosteogenesis type 1 (AO1) AD 108720 2534 1190 FLNB
 Atelosteogenesis type 3 (AO3) AD 108721 2534 56305 FLNB
 Larsen syndrome (dominant) AD 150250 2534 503 FLNB
 Spondylo-carpal-tarsal dysplasia AR 272460 2534 3275 FLNB
 Spondylo-carpal-tarsal dysplasia AR 272460   3275  
 Franck-ter Haar syndrome AR 249420   137834 SH3PXD2B

Please also refer to group 4 for recessive Larsen syndrome and group 26 for conditions with multiple dislocations.

8. TRPV4 group

Metatropic dysplasia is a severe spondyloepimetaphyseal dysplasia characterized in infancy by a long trunk and short limbs with limitation and enlargement of joints and usually severe kyphoscoliosis. The term metatropic comes from the Greek metatropos, and refers to the changing pattern of the skeletal anomalies. Indeed, there is progressive kyphoscoliosis which leads to a shortened trunk. Radiologic features include platyspondyly, metaphyseal enlargement, and shortening of long bones.

Spondylometaphyseal dysplasia, Kozlowski type is characterized by short-trunked short stature, metaphyseal abnormalities in the femur (prominent in the femoral neck and trochanteric area) with coxa vara, scoliosis and platyspondyly.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Metatropic dysplasia AD 156530   2635 TRPV4
 Spondyloepimetaphyseal dysplasia, Maroteaux type (Pseudo-Morquio syndrome type 2) AD 184095   263482 TRPV4
 Spondylometaphyseal dysplasia, Kozlowski type AD 184252   93314 TRPV4
 Brachyolmia, autosomal dominant type AD 113500   93304 TRPV4
 Familial digital arthropathy with brachydactyly AD 606835   85169 TRPV4

9.Short-ribs dysplasias (with or without polydactyly) group

The short rib-polydactyly syndromes (SRPS) are ciliopathies characterized by short ribs, short limbs, polydactyly, and multiple anomalies of major organs, including heart, intestines, genitalia, kidney, liver, and pancreas. In SRPS I (Saldino-Noonan type), the long bones are torpedo-shaped; in SRPS III (Verma-Naumoff type) they are banana-peel shaped. In SRPS II (Majewski syndrome) the tibiae are short and oval, and in SRPS VI (Beemer type), the tibiae are not as short and polydactyly is rare (7).

In asphyxiating thoracic dystrophy (Jeune syndrome), there is a severely constricted thoracic cage, short-limbed short stature, polydactyly, retinal degeneration and pancreatic cysts.

Figure 6. asphyxiating thoracic dystrophy. short ribs long and narrow chest, small pelvis, trident acetabula, no platyspondyly (helps differentiate from thanatophoric dysplasia), cystic renal disease.

Ellis-van Creveld syndrome is characterized by short limbs, short ribs, postaxial polydactyly, and dysplastic nails and teeth.

Figure 7. chondroectodermal dysplasia (or Ellis-van Creveld syndrome). short ribs, early ossification of femoral head, polydactyly cone-shaped epiphyses, no platyspondyly (helps differentiate from thanatophoric dysplasia), flatening of lateral aspect of proximal tibial epiphysis.

In uniparental disomy of paternal chromosome 14, there is a narrow, bell-shaped thorax with caudal bowing of the anterior ribs and cranial bowing of the posterior ribs (coat hanger appearance) (8), and flaring of the iliac wings. There are also joint contractures, dysmorphic facial features, and developmental delay/intellectual deficiency.

Group/name of disorder Inher. OMIM Orpha Gene
 Chondroectodermal dysplasia (Ellis-van Creveld) AR 225500 289 EVC1, ECV2, LBN
Short rib—polydactyly syndrome (SRPS) type 1/3 (Saldino-Noonan/Verma-Naumoff) AR 263510 93271 DYNC2H1
 SRPS type 1/3 (Saldino-Noonan) AR 263510 93271  IFT80
 SRPS type 2 A AR 263520 93269 NEK1
SRPS type 2B AR 615087 93269 DYNC2H1
SRPS type 3 Verma-Naumoff AR 263510 93271 DYNC2H1
 SRPS type 4 (Beemer) AR 269860 93268  
SRPS type 5 AR 614091   WDR35
Uniparental disomy of paternal chromosome 14 (UPD14)   608149 96334 Complete chromosome 14
 Cerebrocostomandibular syndrome AR/AD 117650 1393 SNRPB
 Oral-facial-digital syndrome type 4 (Mohr-Majewski) AR 258860 2753 TCTN3
 Asphyxiating thoracic dysplasia (ATD; Jeune) AR 208500 474 TTC21B, IFT80, WDR19, DYNC2H1,  ATD
 Thoracolaryngopelvic dysplasia (Barnes) AD 187760 3317  

10. Multiple epiphyseal dysplasia and pseudoachondroplasia group

Multiple epiphyseal dysplasia is usually not recognizable before 1-2 years of age (9). Then, joint pain at the hips and knees is noted after physical exercise. Mild to moderate short stature is seen by 5-6 years of age.  Radiologically, there is bilateral necrosis of the femoral heads, and the epiphyses of tubular bones, (including metacarpals, metatarsals and phalanges) show maturational delay. Femoral and phalangeal epiphyses are rounded (COMP) or flat (SCL26A2, see group 4). Double-layered patellae can be seen (SCL26A2). The most frequently mutated genes are COMP and SCL26A2, then the genes encoding type 9 collagen and Matrillin 3.

Figure 8. Multiple epiphyseal dysplasia. Flattened epiphyses, normal spine (no platyspondyly).

Figure 9. pseudoachondroplasia. small femoral head, irregular epiphyses, platyspondyly with anterior tongues of vertebral bodies, irregular acetabula.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Pseudoachondroplasia (PSACH) AD 177170 1487 750 COMP
 Multiple epiphyseal dysplasia (MED) type 1 (EDM1) AD 132400 1123 93308 COMP
 Multiple epiphyseal dysplasia (MED) type 2 (EDM2) AD 600204 1123 166002 COL9A2
 Multiple epiphyseal dysplasia (MED) type 3 (EDM3) AD 600969 1123 166002 COL9A3
 Multiple epiphyseal dysplasia (MED) type 5 (EDM5) AD 607078 1123 93311 MATN3
 Multiple epiphyseal dysplasia (MED) type 6 (EDM6) AD 614135 1123 166002 COL9A1
 Multiple epiphyseal dysplasia (MED), other types     1123    
 Stickler syndrome, recessive type AR 614134 1302 250984 COL9A1
 Familial hip dysplasia (Beukes) AD 142669 1123 2114 UFSP2
 Multiple epiphyseal dysplasia with microcephaly and nystagmus (Lowry-Wood) AR 226960   1824  

Please also refer to multiple epiphyseal dysplasia, recessive type (rMED; EDM4) in sulfation disorders (group 4), familial osteochondritis dissecans in the aggrecan group, as well as ASPED in the Acromelic group

11. Metaphyseal dysplasias

Cartilage-hair hypoplasia manifests with severe disproportionate short-limbed short stature with short hands, bowed femorae and tibiae, joint hypermobility and often metaphyseal dysplasia and large, round epiphyses during childhood, bullet-shaped middle phalanges and vertebral dysplasia. Non-skeletal findings include fine silky slow growing hair, immunodeficiency manifested by an increased rate of infections, anemia, gastrointestinal dysfunction, and an increased risk for malignancy.

Figure 10. cartilage-hair hypoplasia. widening of the growth plate (often focal), metaphyseal cupping and irregularity with cyst-like lucencies, short metacarpals and phalanges with cupping and cone-shaped epiphyses.

Shwachman-Diamond syndrome manifests with exocrine pancreatic insufficiency with malabsorption, malnutrition, and growth failure, hematologic abnormalities, including increased risk of malignant transformation, and skeletal abnormalities which include short stature, generalized osteopenia, with delayed appearance of secondary ossification centers (delayed bone age) metaphyseal chondrodysplasia (metaphyses wide and irregular) and finally thickening and irregularity of the growth plates.

Schmid type of metaphyseal chondrodyplasia manifests with short stature, widened growth plates, bowing of the long bones and resembles a milder form of Jansen type metaphyseal chondrodysplasia. Radiological signs include enlarged capital femoral epiphysis in early childhood, coxa vara, greater involvement of the distal femoral metaphysis than the proximal (these disappear after epiphyseal fusion), anterior rib changes and a normal spine.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Metaphyseal dysplasia, Schmid type (MCS) AD 156500   174 COL10A1
 Cartilage-hair hypoplasia (CHH; metaphyseal dysplasia, McKusick type) AR 250250 84550 175 RMRP
 Metaphyseal dysplasia, Jansen type AD 156400   33067 PTHR1
 Eiken dysplasia AR 600002   79106 PTHR1
 Metaphyseal dysplasia with pancreatic insufficiency and cyclic neutropenia (Shwachman-Diamond syndrome) AR 260400 1756 811 SBDS
 Metaphyseal anadysplasia type 1 AD, AR 602111   1040 MMP13
 Metaphyseal anadysplasia type 2 AR 613073   1040 MMP9
 Metaphyseal dysplasia, Spahr type AR 250400   2501 MMP13
 Metaphyseal acroscyphodysplasia (various types) AR 250215   1240  
 Genochondromatosis (type 1/type 2) AD/SP 137360   85197  
 Metaphyseal chondromatosis with d-2-hydroxyglutaric aciduria AR/SP 614875   99646  IDH1

12. Spondylometaphyseal dysplasias (SMDSpondylometaphyseal dysplasias (SMD)

SMD Sutcliffe type presents with proportional mild short stature. The spine shows odontoid hypoplasia, hyperconvex vertebral body endplates (lower thoracic and upper lumbar) with an appearance of anterior wedging and no platyspondyly or kyphoscoliosis. Hips show progressive coxa vara with short femoral necks leading to a waddling gait. Metaphyseal abnormalities include flakelike, triangular, or curvilinear ossification centers at the edges of the metaphyses simulating “corner fractures” of long tubular bones, distal tibial metaphyses on the ulnar aspect of the distal radius and in the proximal humerus. Some patients have been reported to have COL2A1 mutations.

Group/name of disorder Inher. OMIM Orpha Gene
 Spondyloenchondrodysplasia (SPENCD) AR 271550 1855 ACP5
 Odontochondrodysplasia (ODCD) AR 184260 166272  
 Spondylometaphyseal dysplasia, Sutcliffe type or corner fractures type AD 184255 93315 COL2A1
 SMD with severe genu valgum AD 184253 93316  
 SMD with cone-rod dystrophy AR 608940 85167 PCYT1A
 SMD with retinal degeneration, axial type AR 602271 168549  
 Dysspondyloenchondromatosis SP   85198 COL2A1
 Cheiro-spondyloenchondromatosis SP   99647  

Please also refer to SMD Kozlowski (group TRPV4) disorders in group 11 as well as SMD Sedaghatian type in group 12; there are many individual reports of SMD variants

13. Spondylo-epi-(meta)-physeal dysplasias (SE(M)D)

Spondyloepiphyseal dysplasia tarda manifests with disproportionately short stature and a short trunk. Affected males exhibit retarded growth from about six years of age. Progressive joint and back pain with osteoarthritis follows, involving the larger joints more than the small joints. Radiologically, there are multiple epiphyseal abnormalities, platyspondyly, narrow disc spaces, scoliosis, hypoplastic odontoid process, short femoral necks and coxa vara.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Dyggve-Melchior-Clausen dysplasia (DMC) AR 223800   239 DYM
 Immuno-osseous dysplasia (Schimke) AR 242900 1376 1830 SMARCAL1
 SED, Wolcott-Rallison type AR 226980   1667 EIF2AK3
 SEMD, Matrilin type AR 608728   156728 MATN3
 SEMD, short limb—abnormal calcification type AR 271665   93358 DDR2
 SED tarda, X-linked (SED-XL) XLR 313400 1145 93284 SEDL
 Spondylo-megaepiphyseal-metaphyseal dysplasia (SMMD) AR 613330   228387 NKX3-2
 Spondylodysplastic Ehlers-Danlos syndrome AR 612350   157965 SLC39A13
 SPONASTRIME dysplasia AR 271510   93357  
 SEMD with joint laxity (SEMD-JL) leptodactylic or Hall type AD 603546   93360 KIF22
 SEMD with joint laxity (SEMD-JL) Beighton type AR 271640   93359 B3GALT6
 Platyspondyly (brachyolmia) with amelogenesis imperfecta AR 601216   2899 LTBP3
 Late onset SED, autosomal recessive type AR 609223   93284  
 Brachyolmia, Hobaek type AR 271530   93301 PAPSS2
 Brachyolmia, Toledo type AR 271630   93303 PAPSS2

Please also refer to Brachyolmia (group 8), Opsismodysplasia (group 14), SEMDs (group 11), mucopolysaccharidosis type 4 (Morquio syndrome) and other conditions in group 26, as well as PPRD (SED with progressive arthropathy) in group 31

14. Severe spondylodysplastic dysplasias

In opsismodysplasia, there is a large anterior fontanelle, anteverted nostrils, pelvic bone anomalies, metaphyseal cupping, delayed ossification, shortened digits, hypotonia, and early death.

Group/name of disorder Inher. OMIM Orpha Gene
 Achondrogenesis type 1A (ACG1A) AR 200600 93299 TRIP11
 Schneckenbecken dysplasia AR 269250 3144 SLC35D1
 Spondylometaphyseal dysplasia, Sedaghatian type AR 250220 93317 GPX4
 Severe spondylometaphyseal dysplasia (SMD Sedaghatian-like) AR     SBDS
 Opsismodysplasia AR 258480 2746 INPPL1

Please also refer to Thanatophoric dysplasia, types 1 and 2 (group 1); ACG2 and Torrance dysplasia (group 2); Fibrochondrogenesis (group 3); Achondrogenesis type 1B (ACG1B, group 4); and Metatropic dysplasia (TRPV4 group).

15. Acromelic dysplasias

In Trichorhinophalangeal syndromes, skeletal abnormalities include a short stature, cone-shaped epiphyses at the phalanges, hip malformations, and short stature. All phalanges, metacarpals and metatarsal bones are shortened. Non-skeletal features include sparse scalp hair, bulbous tip of the nose, long flat philtrum, thin upper vermilion border, and protruding ears.

Figure 11. Trichorhinophalangeal syndrome. shortened phalanges and metacarpals, cone-shaped epiphyses.

In Geleophysic dysplasia, there is short stature, short hands and feet, progressive joint limitation and contractures, distinctive facial features ("smiling" round and full face, small nose with anteverted nostrils, a broad nasal bridge, hypertelorism, long flat philtrum, and a thin upper lip), progressive cardiac valvular disease, and thickened skin.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Trichorhinophalangeal dysplasia types 1/3 AD 190350   77258 TRPS1
 Trichorhinophalangeal dysplasia type 2 (Langer-Giedion) AD 150230   502 TRPS1 andEXT1
 Acrocapitofemoral dysplasia AR 607778   63446 IHH
 Cranioectodermal dysplasia (Levin-Sensenbrenner) type 1 AR 218330   1515 IFT122
 Cranioectodermal dysplasia (Levin-Sensenbrenner) type 2 AR 613610   1515 WDR35
 Geleophysic dysplasia AR 231050 11168 2623 ADAMTSL2
 Geleophysic dysplasia, other types AR 614185 11168 2623 FBN1
 Acromicric dysplasia AD 102370   969 SMAD4
 Acrodysostosis type 1 AD 101800   950  PRKAR1A
 Acrodysostosis type 2 AD 614613   950 PDE4D
 Angel-shaped phalango-epiphyseal dysplasia (ASPED) AD 105835   63442  
 Saldino-Mainzer dysplasia AR 266920   140969 IFT140
Myhre syndrome AD 139210   2588 SMAD4
Weill-Marchesani syndrome type 1 AR 277600 1114 3449 ADAMTS10
Weill-Marchesani syndrome type 2 AD 608328 1114 2084 FBN1

Please also refer to the short rib dysplasias group

16. Acromesomelic dysplasias

In Acromesomelic dysplasia, type Maroteaux, there is disproportionate shortening the middle segments (forearms and forelegs) and distal segments (hands and feet) of the appendicular skeleton. There are short broad fingers, shortening of the middle long bones with a bowed radius, and wedging of vertebral bodies.

Group/name of disorder Inher. OMIM Orpha Gene
 Acromesomelic dysplasia type Maroteaux (AMDM) AR 602875 40 NPR2
 Grebe dysplasia AR 200700 2098 GDF5
 Fibular hypoplasia and complex brachydactyly (Du Pan) AR 228900 2639 GDF5
 Acromesomelic dysplasia with genital anomalies AR 609441   BMPR1B
 Acromesomelic dysplasia, Osebold-Remondini type AD 112910 93382  
Acromesomelic dysplasia, Hunter-Thomson type AR 201250 968 GDF5

17. Mesomelic and rhizo-mesomelic dysplasias

Leri-Weill dyschondrosteosis is characterized by short stature, mesomelia, and Madelung wrist deformity (abnormal alignment of the radius, ulna, and carpal bones at the wrist - more common and severe in females).

Group/name of disorder Inher. OMIM GR Orpha Gene
 Dyschondrosteosis (Leri-Weill) Pseudo-AD 127300 1215 240 SHOX
 Langer type (homozygous dyschondrosteosis) Pseudo-AR 249700 1215 2632 SHOX
 Omodysplasia AR 258315   93329 GPC6
 Robinow syndrome, recessive type AR 268310 1240 1507 ROR2
 Robinow syndrome, dominant type AD 180700   3107 WNT5A
 Mesomelic dysplasia, Korean type AD        
 Mesomelic dysplasia, Kantaputra type AD 156232   1836  
 Mesomelic dysplasia, Nievergelt type AD 163400   2633  
 Mesomelic dysplasia, Kozlowski-Reardon type AR 249710   2631  
 Mesomelic dysplasia with acral synostoses (Verloes-David-Pfeiffer type) AD 600383   2496 SULF1, SLCO5A1
 Mesomelic dysplasia, Savarirayan type (Triangular Tibia-Fibular Aplasia) SP 605274   85170  

18. Bent bones dysplasias

Campomelic dysplasia is characterized by bowed, short and fragile long bones, clubfeet, pelvis and chest abnormalities and eleven pairs of ribs. Non-skeletal anomalies include a flat face, laryngotracheomalacia, Pierre Robin sequence with cleft palate, ambiguous genitalia in males, and brain, heart and kidney malformations.

Figure 12. Campomelic dysplasia. bell-shaped thorax, hypoplastic scapula, bowed femurs, widely-spaced ischial bones.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Campomelic dysplasia (CD) AD 114290 1760 140 SOX9
 Stüve-Wiedemann dysplasia AR 601559   3206 LIFR
 Kyphomelic dysplasia, several forms   211350   1801  

Bent bones at birth can be seen in osteogenesis imperfecta, Antley-Bixler syndrome, cartilage-hair hypoplasia, Cummings syndrome, hypophosphatasia, dyssegmental dysplasia, TD, ATD, and other conditions.

19. Slender bone dysplasia group

In Three M (3M) syndrome, there is severe prenatal and postnatal growth retardation, distinctive facial features (large head, triangular face, hypoplastic midface, full eyebrows, fleshy nose tip, long philtrum, prominent mouth and lips, and pointed chin),  and normal mental development. The main skeletal anomalies are slender long bones and ribs, foreshortened vertebral bodies, and delayed bone age. Joint hypermobility, joint dislocation, winged scapulae, and pes planus can also be seen.

Group/name of disorder Inher. OMIM GR Orpha Gene
 3-M syndrome (3M1) AR 273750 1481 2616 CUL7
 3-M syndrome (3M2) AR 612921 1481 2616 OBSL1
 Kenny-Caffey dysplasia type 1 AR 244460   93324 TBCE
 Kenny-Caffey dysplasia type 2 AD 127000   93325 FAM111A
 Microcephalic osteodysplastic primordial dwarfism type 1/3 (MOPD1) AR 210710   2636 RNU4ATAC
 Microcephalic osteodysplastic primordial dwarfism type 2 (MOPD2; Majewski type) AR 210720   2637 PCNT2
 IMAGE syndrome (intrauterine growth retardation, metaphyseal dysplasia, adrenal hypoplasia, and genital anomalies) XL/AD 614732   85173 CDKN1C
 Osteocraniostenosis SP 602361   2763 FAM111A
 Hallermann-Streiff syndrome AR 234100   2108 GJA1

Please also see Cerebro-arthro-digital dysplasia.

20. Dysplasias with multiple joint dislocations

Desbuquois dysplasia is characterized by short stature of prenatal onset affecting the rhizomelic and mesomelic portion of the limbs, marked joint laxity, kyphoscoliosis and facial dysmorphisms (round flat face, prominent eyes, and midface hypoplasia)

Group/name of disorder Inher. OMIM Orpha Gene
 Desbuquois dysplasia (with accessory ossification center in digit 2) AR 251450 1425 CANT1
 Desbuquois dysplasia with short metacarpals and elongated phalanges (Kim type) AR 251450 1425 CANT1
 Desbuquois dysplasia (other variants with or without accessory ossification center) AR      
 Pseudodiastrophic dysplasia AR 264180 85174  

Please also refer to SED with congenital dislocations, CHST3 type (group 4); Atelosteogenesis type 3 and Larsen syndrome (group 6); SEMDs with joint laxity (group 11)

21. Chondrodysplasia punctata (CDP) group

The more severe, classic rhizomelic chondrodysplasia punctata type 1 can manifest in neonates with cataracts, rhizomelia, metaphyseal abnormalities, and punctate calcifications in the epiphyseal cartilage at the knee, hip, elbow, and shoulder, involving the hyoid bone, larynx, costochondral junctions, and vertebrae (chondrodysplasia punctata). In addition, unossified cartilage in the vertebral bodies show as radiolucent coronal clefts.

Figure 13. rhizomelic chondrodysplasia punctate type 1. punctate epitphyses, very small humeri less shortening of femurs, coronal clefts in vertebral bodies.

Group/name of disorder Inher. OMIM GR Orpha Gene
 CDP, X-linked dominant, Conradi-Hünermann type (CDPX2) XLD 302960 55062 35173 EBP
 CDP, X-linked recessive, brachytelephalangic type (CDPX1) XLR 302950 1544 79345 ARSE
 Congenital hemidysplasia, ichthyosis, limb defects (CHILD) XLD 308050 51754 139 NSDHL
 Congenital hemidysplasia, ichthyosis, limb defects (CHILD) XLD 308050   139 EBP
 Greenberg dysplasia AR 215140   1426 LBR
 Rhizomelic CDP type 1 AR 215100 1270 177 PEX7
 Rhizomelic CDP type 2 AR 222765   177 DHPAT
 Rhizomelic CDP type 3 AR 600121   177 AGPS
 CDP tibial-metacarpal type AD/AR 118651   79346  
 Astley-Kendall dysplasia AR?     85175  

Note that stippling can occur in several syndromes such as Zellweger, Smith-Lemli-Opitz and others. Please also refer to desmosterolosis as well as SEMD short limb—abnormal calcification type in group 11.

22. Neonatal osteosclerotic dysplasias

Caffey disease manifests with subperiosteal new bone formation (long bones, ribs, mandible, scapulae, and clavicles) associated with fever, joint swelling and pain. Onset is around age two months and the episodes stop by age two years.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Blomstrand dysplasia AR 215045   50945 PTHR1
 Desmosterolosis AR 602398   35107 DHCR24
 Caffey disease (including infantile and attenuated forms) AD 114000 99168 1310 COL1A1
 Caffey disease (severe variants with prenatal onset) AR 114000 99168 1310 COL1A1
 Raine dysplasia (lethal and non-lethal forms) AR 259775   1832 FAM20C

Please also refer to Astley-Kendall dysplasia and CDPs in group 21

23. Increased bone density group (without modification of bone shape)

Osteopetrosis can manifest with increased bone density, diffuse and focal sclerosis, modelling defects at metaphyses, pathological fractures, osteomyelitis, tooth eruption defects and dental caries. Other complications include cranial nerve compression, hydrocephalus, pancytopaenia, extramedullary haematopoiesis, hepatosplenomegaly, and hypocalcaemia (10).

Figure 14. osteopetrosis. thick dense bones, alternating bands of sclerosis and normal density bone in long bones, rugger jersey spine, dense base of skull.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Osteopetrosis, severe neonatal or infantile forms (OPTB1) AR 259700   667 TCIRG1
 Osteopetrosis, severe neonatal or infantile forms (OPTB4) AR 611490 1127 667 CLCN7
 Osteopetrosis, infantile form, with nervous system involvement (OPTB5) AR 259720   667 OSTM1
 Osteopetrosis, intermediate form, osteoclast-poor (OPTB2) AR 259710   667 TNFSF11
 Osteopetrosis, infantile form, osteoclast-poor with immunoglobulin deficiency (OPTB7) AR 612301   667 TNFRSF11A
 Osteopetrosis, intermediate form (OPTB6) AR 611497   210110 PLEKHM1
 Osteopetrosis, intermediate form (OPTA2) AR 259710 1127 667 CLCN7
 Osteopetrosis with renal tubular acidosis (OPTB3) AR 259730   2785 CA2
 Osteopetrosis, late-onset form type 1 (OPTA1) AD 607634   2783 LRP5
 Osteopetrosis, late-onset form type 2 (OPTA2) AD 166600   53 CLCN7
 Osteopetrosis with ectodermal dysplasia and immune defect (OLEDAID) XL 300301   69088 IKBKG
 Osteopetrosis, moderate form with defective leucocyte adhesion (LAD3) AR 612840   2968 KIND3
 Pyknodysostosis AR 265800   763 CTSK
 Osteopoikilosis AD 155950   2485 LEMD3
 Melorheostosis with osteopoikilosis AD 155950   2485 LEMD3
 Osteopathia striata with cranial sclerosis (OSCS) XLD 300373   2780 WTX
 Melorheostosis SP 155950   2485 LEMD3
 Dysosteosclerosis AR 224300   1782 SLC29A3
 Osteomesopyknosis AD 166450   2777  
 Osteopetrosis with infantile neuroaxonal dysplasia AR? 600329   85179  

24. Increased bone density group with metaphyseal and/or diaphyseal involvement

Camurati-Engelmann manifests with bilateral cortical thickening (hyperostosis) of the diaphyses of the long bones starting with the femora and tibiae. The metaphyses and the skull base may be affected as well, but the epiphyses are spared. Limb pain, muscle weakness, a waddling gait, and easy fatigability can also occur.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Craniometaphyseal dysplasia, autosomal dominant type AD 123000 1461 1522 ANKH
 Diaphyseal dysplasia Camurati-Engelmann AD 131300 1156 1328 TGFB1
 Hematodiaphyseal dysplasia Ghosal AR 231095   1802 TBXAS1
 Hypertrophic osteoarthropathy AR 259100   1525 HPGD
 Pachydermoperiostosis (hypertrophic osteoarthropathy, primary, autosomal dominant) AD 167100   2796  
 Oculodentoosseous dysplasia (ODOD) mild type AD 164200   2710 GJA1
 Oculodentoosseous dysplasia (ODOD) severe type AR 257850   2710 GJA1
 Osteoectasia with hyperphosphatasia (juvenile Paget disease) AR 239000   2801 OPG
 Sclerosteosis AR 269500 1228 3152 SOST
 Endosteal hyperostosis, van Buchem type AR 239100 1228 3416 SOST
 Trichodentoosseous dysplasia AD 190320   3352 DLX3
 Craniometaphyseal dysplasia, autosomal recessive type AR 218400   1522 GJA1
 Diaphyseal medullary stenosis with bone malignancy AD 112250   85182 MTAP
 Craniodiaphyseal dysplasia AD 122860 1228 1513 SOST
 Craniometadiaphyseal dysplasia, Wormian bone type AR 615118   85184  
 Endosteal sclerosis with cerebellar hypoplasia AR 213002   85186  
 Lenz-Majewski hyperostotic dysplasia SP 151050   2658 PTDSS1
 Metaphyseal dysplasia, Braun-Tinschert type XL 605946   85188  
 Pyle disease AR 265900   3005 SFRP4
      1. 25. Osteogenesis imperfecta and decreased bone density

 

Osteogenesis imperfect (OI) manifests with low bone mineral density and bone fragility with frequent fractures, bone deformities and short stature, dentinogenesis imperfecta (fragile grey or brown somewhat translucent teeth), and progressive hearing loss. In type I, stature is normal or slightly short, there is no bone deformity, the sclerae can be blue and there is no dentinogenesis imperfecta. Type II is the most severe with multiple rib and long bone fractures at or before birth, marked deformities, broad long bones, low density on skull X-rays, and dark sclera. OI type III presents with very short stature, a triangular face, severe scoliosis, gray sclera, and dentinogenesis imperfecta. In Type IV, the phenotype is milder with moderately short stature, mild to moderate scoliosis, grayish or white sclera, and dentinogenesis imperfecta. Type V is characterized by mild to moderate short stature, calcification of the forearm interosseous membrane, radial head dislocation and hyperplastic callus formation following fractures, and no dentinogenesis imperfecta.

Figure 15. oi type ii. wormian bones, thick short crumpled long bones, rectangular wavy femora, thick beaded ribs.

 

Group/name of disorder Inher. OMIM GR Orpha Gene
Osteogenesis imperfecta, non-deforming form (OI type I) AD 166200 1295 216796 COL1A1,COL1A2
Osteogenesis imperfecta, perinatal lethal form (OI type II) AD, AR 166210 1295 216804 COL1A1,COL1A2,CRTAP,LEPRE1,PPIB
Osteogenesis imperfecta, progressively deforming type (OI type III) AD, AR 259420 1295 216812 COL1A1,COL1A2,CRTAP,LEPRE1,PPIB,FKBP10,SERPINH1 , WNT1, TMEM38B
Osteogenesis imperfecta, moderate form (OI type IV) AD, AR 166220 1295 216820 COL1A1,COL1A2,CRTAP,FKBP10,SP7
Osteogenesis imperfecta with calcification of the interosseous membranes and/or hypertrophic callus (OI type V) AD 610967   216828  IFITM5
Osteogenesis imperfecta, type VI AR 613982   216812 SERPINF1
Osteogenesis imperfecta, type VII AR 610682   216804 CRTAP
Bruck syndrome type 1 (BS1) AR 259450   2771 FKBP10
Bruck syndrome type 2 (BS2) AR 609220   2771 PLOD2
Osteoporosis-pseudoglioma syndrome AR 259770   2788 LRP5
Calvarial doughnut lesions with bone fragility AD 126550   85192  
Idiopathic juvenile osteoporosis SP 259750   85193  
Cole-Carpenter dysplasia (bone fragility with craniosynostosis) SP 112240   2050 P4HB
Spondylo-ocular dysplasia AR 605822   85194 XYLT2
Osteopenia with radiolucent lesions of the mandible AD 166260   53697 ANO5
Ehlers-Danlos syndrome, progeroid form AR 130070   75496 B4GALT7
Geroderma osteodysplasticum AR 231070   2078 GORAB
Cutis laxa, autosomal recessive form, type 2B (ARCL2B) AR 612940   90350 PYCR1
Cutis laxa, autosomal recessive form, type 2A (ARCL2A) (Wrinkly skin syndrome) AR 219200 5200 90350 ATP6VOA2
Wrinkly skin syndrome AR 278250 5200 2834 ATP6VOA2
Singleton-Merten dysplasia AD 182250   85191 IFIH1

26. Abnormal mineralization group

Hypophosphatasia results from low alkaline phosphatase (TNSALP) activity. Inorganic pyrophosphate (PPi), an inhibitor of mineralization, and pyridoxal 5′-phosphate (PLP), are substrates that accumulate. The types include the prenatal benign form which spontaneously improves, perinatal (lethal), infantile (respiratory complications, premature craniosynostosis, widespread demineralization and rachitic changes in the metaphyses), childhood (skeletal deformities, short stature, and waddling gait), and adult (stress fractures, thigh pain, chondrocalcinosis and marked osteoarthropathy). Two other forms include odontohypophosphatasia (no clinical changes in long bones are present, only biochemical and dental manifestations such as premature exfoliation of fully rooted primary teeth and/or severe dental caries) and pseudohypophosphatasia (indistinguishable from infantile hypophosphatasia, but serum alkaline phosphatase activity is normal). Enzyme replacement is now available.

Hypophosphatemic rickets is discussed in detail in the section on bone and mineral metabolism of Endotext. Rickets manifests with bowing of the weight bearing bones. Other frequent manifestations are growth failure with disproportionate short stature, frontal bossing, and swelling of wrists, knees, and ankles. A rachitic rosary arises due to expansion of the costo-chondral junctions, and an inward diaphragmatic pull of soft rib cage leads to Harrison's sulcus (groove). Dentition may be delayed and enamel development can be impaired.

Figure 16. rickets. widened growth plates, cupping fraying of metaphyses, demineralization , widened anterior rib ends.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Hypophosphatasia, perinatal lethal and infantile forms AR 241500 1150 436 ALPL
 Hypophosphatasia, adult form AD 146300 1150 436 ALPL
 Hypophosphatemic rickets, X-linked dominant XLD 307800 83985 89936 PHEX
 Hypophosphatemic rickets, autosomal dominant AD 193100   89937 FGF23
 Hypophosphatemic rickets, autosomal recessive, type 1 (ARHR1) AR 241520   289176 DMP1
 Hypophosphatemic rickets, autosomal recessive, type 2 (ARHR2) AR 613312   289176 ENPP1
 Hypophosphatemic rickets with hypercalciuria, X-linked recessive XLR 300554   1652 ClCN5
 Hypophosphatemic rickets with hypercalciuria, autosomal recessive (HHRH) AR 241530   157215 SLC34A3
 Neonatal hyperparathyroidism, severe form AR 239200   417 CASR
 Familial hypocalciuric hypercalcemia with transient neonatal hyperparathyroidism AD 145980   405 CASR
 Calcium pyrophosphate deposition disease (familial chondrocalcinosis) type 2 AD 118600   1416 ANKH
    1. 27. Lysosomal storage diseases with skeletal involvement (dysostosis multiplex group).

Several lysosomal storage diseases manifest with dysostosis multiplex (11). Clinically, there is evolving joint contractures without inflammation. Radiologically, the skull shows an abnormal J-shaped sella turcica and a thickened diploic space. The ribs are oar-shaped ribs (widened anteriorly and tapered posteriorly) and clavicles are short and thickened. The spine shows multiple superiorly notched (inferiorly beaked) vertebrae and posterior scalloping. The pelvis shows rounded iliac wings and inferior tapering of the ilea. The long bones can have mildly hypoplastic epiphyses. The capital femoral epiphyses can be fragmented, and there can be proximal humeral notching, long and narrow femoral necks, hypoplastic distal ulnae, and thickened short diaphyses. In the hands, proximally pointed metacarpals are short and thick with thin cortices.

Figure 17. mucopolysaccharidoses. wide ribs, glenoid hypoplasia, steep acetabula with constricted iliac wings, flat/irregular femoral head , spearhead metacarpals, platyspondyly, central anterior vertebral body beaking, hypoplastic odontoid.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Mucopolysaccharidosis type 1H/1S AR 607014 1162 93473 IDUA
 Mucopolysaccharidosis type 2 XLR 309900 1274 580 IDS
 Mucopolysaccharidosis type 3A AR 252900   581 SGSH
 Mucopolysaccharidosis type 3B AR 252920   581 NAGLU
 Mucopolysaccharidosis type 3C AR 252930   581 HSGNAT
 Mucopolysaccharidosis type 3D AR 252940   581 GNS
 Mucopolysaccharidosis type 4A AR 253000 148668 582 GALNS
 Mucopolysaccharidosis type 4B AR 253010   582 GLB1
 Mucopolysaccharidosis type 6 AR 253200   583 ARSB
 Mucopolysaccharidosis type 7 AR 253220   584 GUSB
 Fucosidosis AR 230000   349 FUCA1
 alpha-Mannosidosis AR 248500 1396 61 MAN2B1
 beta-Mannosidosis AR 248510   118 MANBA
 Aspartylglucosaminuria AR 208400   93 AGA
 GMI Gangliosidosis, several forms AR 230500   354 GLB1
 Sialidosis, several forms AR 256550   812 NEU1
 Sialic acid storage disease (SIASD) AR 269920   834 SLC17A5
 Galactosialidosis, several forms AR 256540   351 CTSA
 Multiple sulfatase deficiency AR 272200   585 SUMF1
 Mucolipidosis II (I-cell disease), alpha/beta type AR 252500 1828 576 GNPTAB
 Mucolipidosis III (Pseudo-Hurler polydystrophy), alpha/beta type AR 252600 1875 577 GNPTAB
 Mucolipidosis III (Pseudo-Hurler polydystrophy), gamma type AR 252605 24701 577 GNPTG
    1. 28. Osteolysis group

Hajdu-Cheney syndrome is characterized by short stature, bowing of the long bones, vertebral anomalies, progressive focal bone destruction, acroosteolysis and generalized osteoporosis. Facial features are coarse and can include hypertelorism, bushy eyebrows, micrognathia, a small mouth with dental anomalies, low-set ears, and short neck.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Familial expansile osteolysis AD 174810   85195 TNFRSF11A
 Mandibuloacral dysplasia type A AD 248370   90153 LMNA
 Mandibuloacral dysplasia type B AR 608612   90154 ZMPSTE24
 Progeria, Hutchinson-Gilford type AD 176670 1121 740 LMNA
 Torg-Winchester syndrome AR 259600   3460 MMP2
 Hajdu-Cheney syndrome AD 102500   955  NOTCH2
 Multicentric carpal-tarsal osteolysis with and without nephropathy AD 166300   2774  MAFB
 Lipomembraneous osteodystrophy with leukoencephalopathy (presenile dementia with bone cysts; Nasu-Hakola) AR 221770 1197 2770 TREM2
 Lipomembraneous osteodystrophy with leukoencephalopathy (presenile dementia with bone cysts; Nasu-Hakola) AR 221770 1197 2770 TYROBP

Please also refer to Pycnodysostosis, cleidocranial dysplasia, and Singleton-Merten syndrome. Note: several neurologic conditions may cause acroosteolysis

  1. Disorganized development of skeletal components group

Multiple hereditary exostoses are characterized by projections of bone capped by cartilage, in the metaphyses and the diaphyses of long bones.

Fibrodysplasia ossificans progressiva (FOP) is characterized by malformation of the hallux during embryonic skeletal development and by progressive heterotopic endochondral ossification later in life. In the first decade, episodes of painful soft tissue swellings precipitated by soft tissue injury, intramuscular injections, viral infection, muscular stretching, falls or fatigue lead to heterotopic bone formation. The heterotopic bone forms in the skeletal muscles, tendons, ligaments, fascia, and aponeuroses. This phenomenon is seen first in the dorsal, axial, cranial and proximal regions of the body, and later in the ventral, appendicular, caudal and distal regions.

Figure 18. fibrodysplasia ossificans progressive. trapezoid-shaped proximal phalanx of the great toe, soft tissue ossification, exostosis-like structures at sites of ligamentous attachment.

Fibrous dysplasia, polyostotic form, or McCune-Albright syndrome is characterized by polyostotic fibrous dysplasia, cafe au lait cutaneous spots and endocrinopathies (peripheral precocious puberty, thyroidopathies, acromegaly, etc.). The skeletal manifestations are asymmetric fibrous dysplasia affecting any bone. Pathologic fracture, pseudarthrosis, bone deformity such as the shepherd's crook of the proximal femurs are characteristic.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Multiple cartilaginous exostoses 1 AD 133700 1235 321 EXT1
 Multiple cartilaginous exostoses 2 AD 133701 1235 321 EXT2
 Multiple cartilaginous exostoses 3 AD 600209   321  
 Cherubism AD 118400 1137 184 SH3BP2
 Fibrous dysplasia, polyostotic form,
McCune-Albright syndrome
SP 174800   562 GNAS
 Progressive osseous heteroplasia AD 166350   2762 GNAS
 Gnathodiaphyseal dysplasia AD 166260   53697 TMEM16E
 Metachondromatosis AD 156250   2499 PTPN11
 Osteoglophonic dysplasia AD 166250 1455 2645 FGFR1
 Fibrodysplasia ossificans progressiva (FOP) AD, SP 135100   337 ACVR1
 Neurofibromatosis type 1 (NF1) AD 162200 1109 636 NF1
 Carpotarsal osteochondromatosis AD 127820   2767  
 Cherubism with gingival fibromatosis (Ramon syndrome) AR 266270   3019  
 Dysplasia epiphysealis hemimelica (Trevor) SP 127800   1822  
 Enchondromatosis (Ollier) SP 166000   296 IDH1, IDH2, and PTH1R
 Enchondromatosis with hemangiomata (Maffucci) SP 166000   296 DH1, IDH2, and PTH1R

Please also refer to Proteus syndrome in group 30.

  1. Overgrowth syndromes with skeletal involvement

Marfan syndrome manifests with skeletal, ocular and cardiovascular features. Skeletal features include joint laxity, scoliosis and extremities that are disproportionately long for the size of the trunk. Overgrowth of the ribs can cause pectus excavatum or carinatum. Ocular features include myopia and displacement of the lens from the center of the pupil. Cardiovascular features include dilatation of the aorta, susceptibility to aortic tear and rupture, mitral or tricuspid valve prolapse, and enlargement of the proximal pulmonary artery.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Weaver syndrome SP/AD 277590   3447 EZH2
 Sotos syndrome AD 117550 1479 821 NSD1
 Marshall-Smith syndrome SP 602535   561 NFIX
 Proteus syndrome SP 176920 99495 744 AKT1
 Marfan syndrome AD 154700 1335 558 FBN1
 Congenital contractural arachnodactyly AD 121050 1386 115 FBN2
 Loeys-Dietz syndrome types 1A and 2A AD 609192,610168, 1133   TGFBR1
 Loeys-Dietz syndrome types 1B and 2B AD 608967, 610380 1133   TGFBR2
Loeys-Dietz syndrome, type 3 AD 613795 1133 284984 SMAD3
Loeys-Dietz syndrome, type 4 AD 614816 1133 91387 TGFB2
 Overgrowth syndrome with 2q37 translocations SP       NPPC
 Overgrowth syndrome with skeletal dysplasia (Nishimura-Schmidt, endochondral gigantism) SP?        

 

Please also refer to Shprintzen-Goldberg syndrome in Craniosynostosis group

  1. Genetic inflammatory/rheumatoid-like osteoarthropathies

Familial hyperphosphatemic tumoral calcinosis is characterized by the progressive deposition of calcium phosphate crystals in periarticular spaces, soft tissues, and bones (periosteal reaction and cortical hyperostosis). It is caused by increased renal absorption of phosphate secondary to loss-of-function mutations in FGF23 or GALNT3.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Progressive pseudorheumatoid dysplasia (PPRD; SED with progressive arthropathy) AR 208230   1159 WISP3
 Chronic infantile neurologic cutaneous articular syndrome (CINCA)/neonatal onset multisystem inflammatory disease (NOMID) AD 607115   1451 CIAS1
 Sterile multifocal osteomyelitis, periostitis, and pustulosis (CINCA/NOMID-like) AR 612852   210115 IL1RN
 Chronic recurrent multifocal osteomyelitis with congenital dyserythropoietic anemia (CRMO with CDA; Majeed syndrome) AR 609628 1974 77297 LPIN2
 Tumoral calcinosis, hyperphosphatemic, familial AR 211900   53715 GALNT3, FGF23, KL
 Infantile systemic hyalinosis/Juvenile hyaline fibromatosis (ISH/JHF) AR 236490 1525 2176 ANTXR2
camptodactyly-arthropathy-coxa vara-pericarditis syndrome (non-inflammatory) AR 208250   2848 PRG4
  1. Cleidocranial dysplasia and isolated cranial ossification defects group

Cleidocranial dysplasia manifests with large, wide-open fontanels at birth which may remain open with bulging calvaria, mid-face hypoplasia, hypoplasia or aplasia of the clavicles permitting apposition of the shoulders, wide pubic symphysis, brachydactyly, tapering fingers, and short, broad thumbs, dental anomalies (delayed eruption of secondary dentition, failure to shed the primary teeth, supernumerary teeth with dental crowding, and malocclusion).

Figure 19. Cleidocranial dysplasia. wormian bones, partial (or rarely complete) absence of clavicle, widened symphysis pubis, tall femoral head ossification centers, cone-shaped epiphyses.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Cleidocranial dysplasia AD 119600 1513 1452 RUNX2
 CDAGS syndrome (craniosynostosis, delayed fontanel closure, parietal foramina, imperforate anus, genital anomalies, skin eruption) AR 603116   85199  
 Yunis-Varon syndrome AR 216340   3472 FIG4
 Parietal foramina (isolated) AD 168500 1128 60015 ALX4
 Parietal foramina (isolated) AD 168500 1128 60015 MSX2

Please also refer to pycnodysostosis, wrinkly skin syndrome, and several others

  1. Craniosynostosis syndromes

Craniosynostosis is often secondary to mutations in one of the FGFR genes (12). In Apert syndrome (FGFR2) there is midface hypoplasia and symmetrical syndactyly of hands and feet. In Crouzon syndrome there is maxillary hypoplasia, shallow orbits, ocular proptosis, and normal extremities. It is caused by FGFR2 mutations unless there is acanthosis nigricans (FGFR3). In Muenke syndrome (FGFR3), there is unilateral or bilateral coronal synostosis, and absent or minimal hand/foot anomalies. In Pfeiffer syndrome there is  high forehead, maxillary hypoplasia, mild syndactyly of hands and/or feet, broad thumbs and/or great toe (FGFR2, rarely FGFR1). In Saethre-Chotzen syndrome there is brachycephaly/plagiocephaly, a high forehead, facial asymmetry, maxillary hypoplasia, brachydactyly, partial cutaneous syndactyly in some cases, and thumb/great toe anomalies (TWIST gene, occasionally FGFR3).

Group/name of disorder Inher. OMIM GR Orpha Gene
 Pfeiffer syndrome (FGFR1-related) AD 101600 1455 710 FGFR1
 Pfeiffer syndrome (FGFR2-related) AD 101600 1455 710 FGFR2
 Apert syndrome AD 101200 1455 87 FGFR2
 Craniosynostosis with cutis gyrata (Beare-Stevenson) AD 123790 1455 1555 FGFR2
 Crouzon syndrome AD 123500 1455 207 FGFR2
 Crouzon-like craniosynostosis with acanthosis nigricans (Crouzonodermoskeletal syndrome) AD 612247 1455 93262 FGFR3
 Craniosynostosis, Muenke type AD 602849 1455 53271 FGFR3
 Antley-Bixler syndrome AR 201750 1419 63269 POR
 Craniosynostosis Boston type AD 604757   1541 MSX2
 Saethre-Chotzen syndrome AD 101400 1189 794 TWIST1
 Shprintzen-Goldberg syndrome AD 182212 1277 2462 SKI
 Baller-Gerold syndrome AR 218600 1204 1225 RECQL4
 Carpenter syndrome AR 201000   65759 RAB23

Please also refer to Cole-Carpenter syndrome in group 24, CDAGS syndrome in group 29, and Craniofrontonasal syndrome in group 34

  1. Dysostoses with predominant craniofacial involvement

Treacher Collins syndrome manifests with fdownslanting eyes, coloboma of the eyelids, micrognathia, microtia and other deformity of the ears, hypoplastic zygomatic arches, macrostomia, conductive hearing loss and cleft palate.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Mandibulo-facial dysostosis (Treacher Collins, Franceschetti-Klein) AD 154500 1532 861 TCOF1
 Mandibulo-facial dysostosis (Treacher-Collins, Franceschetti-Klein) AD 154500 1532 861 POLR1D
 Mandibulo-facial dysostosis (Treacher-Collins, Franceschetti-Klein) AR 154500 1532 861 POLR1C
 Oral-facial-digital syndrome type I (OFD1) XLR 311200   2750 CXORF5
 Weyer acrofacial (acrodental) dysostosis AD 193530   952 EVC1
 Endocrine-cerebro-osteodysplasia (ECO) AR 612651   199332 ICK
 Craniofrontonasal syndrome XLD 304110   1520 EFNB1
 Frontonasal dysplasia, type 1 AR 136760   250 ALX3
 Frontonasal dysplasia, type 2 AR 613451   228390 ALX4
 Frontonasal dysplasia, type 3 AR 613456   306542 ALX1
 Hemifacial microsomia SP/AD 164210 5199 374  
 Miller syndrome (postaxial acrofacial dysostosis) AR 263750   246 DHODH
 Acrofacial dysostosis, Nager type AD/AR 154400   245 SF3B4
 Acrofacial dysostosis, Rodriguez type AR 201170   1788  

Please also refer to Oral-facial-digital syndrome type IV in the Short Rib Dysplasias group

  1. Dysostoses with predominant vertebral with and without costal involvement

In spondylocostal dysostosis, there are multiple segmentation defects of the vertebrae, malalignment of the ribs with variable points of intercostal fusion, and a reduction in rib number. Clinically there is scoliosis, a short neck and trunk.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Currarino triad AD 176450   1552 HLXB9
 Spondylocostal dysostosis type 1 (SCD1) AR 277300 8828 2311 DLL3
 Spondylocostal dysostosis type 2 (SCD2) AR 608681 8828 2311 MESP2
 Spondylocostal dysostosis type 3 (SCD3) AR? 609813 8828 2311 LFNG
 Spondylocostal dysostosis type 4 (SCD4) AR 613686 8828 2311 HES7
 Spondylothoracic dysostosis AR 122600 8828 1797 MESP2
 Klippel-Feil anomaly with laryngeal malformation AD 118100   2345 GDF6
 Spondylocostal/thoracic dysostosis, other forms AD/AR        
 Cerebro-costo-mandibular syndrome (rib gap syndrome) AD/AR 117650   1393 SNRPB
 Cerebro-costo-mandibular-like syndrome with vertebral defects AR 611209   263508 COG1
 Diaphanospondylodysostosis AR 608022   66637 BMPER

Please also refer to Spondylocarpotarsal dysplasia in group 7 and spondylo-metaphyseal-megaepiphyseal dysplasia in group 13

  1. Patellar dysostoses

Nail-patella syndrome presents with patella hypoplasia, nail hypoplasia or dystrophy, elbow and knee deformities (limitation of elbow extension, pronation, and supination; cubitus valgus; and antecubital pterygia), iliac horns (bilateral, conical bony processes projecting posteriorly and laterally from the central part of the iliac bones of the pelvis), nephropathy (nephrotic syndrome which may progress to end-stage renal disease), and ocular defects (cloverleaf appearance of the iris, primary open angle glaucoma).

Figure 20. Nail-patella syndrome. absent patella, iliac horns, radial head dislocation, spondylolysthesis.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Ischiopatellar dysplasia (small patella syndrome) AD 147891   1509 TBX4
 Small patella—like syndrome with clubfoot AD 119800   293150 PITX1
 Nail-patella syndrome AD 161200 1132 2614 LMX1B
 Genitopatellar syndrome AR? 606170 114806 85201 KAT6B
 Ear-patella-short stature syndrome (Meier-Gorlin) AR 224690   2554 ORC1, ORC1L, ORC4, ORC4L, ORC6, ORC6L, CDT1, CDC6, CDC18L

Please also refer to MED group for conditions with patellar changes as well as ischio-pubic-patellar dysplasia as mild expression of campomelic dysplasia

  1. Brachydactylies (with or without extraskeletal manifestations)

Coffin-Siris syndrome (CSS) is characterized by aplasia or hypoplasia of the distal phalanx or nail of the fifth digit (or more digits), distinctive facial features (wide mouth with thick, everted upper and lower lips, broad nasal bridge with broad nasal tip, thick eyebrows and long eyelashes), and moderate to severe developmental/cognitive delay.

Thorough discourses on the genes involved in each condition can be found in papers by Schwabe and Mundlos (13), Temtamy and Aglan (14), and Mundlos (15).

Group/name of disorder Inher. OMIM GR Orpha Gene
 Brachydactyly type A1 AD 112500   93388 IHH
 Brachydactyly type A1 AD       5p13.3-p13.2
 Brachydactyly type A2 AD 112600   93396 BMPR1B
 Brachydactyly type A2 AD 112600   93396 BMP2
 Brachydactyly type A2 AD 112600   93396 GDF5
 Brachydactyly type A3 AD 112700   93393  
 Brachydactyly type B AD 113000   93383 ROR2
 Brachydactyly type B2 AD 611377   140908 NOG
 Brachydactyly type C AD, AR 113100   93384 GDF5
 Brachydactyly type D AD 113200   93385 HOXD13
 Brachydactyly type E AD 113300   93387 PTHLH
 Brachydactyly type E AD 113300   93387 HOXD13
 Brachydactyly—mental retardation syndrome AD 600430   1001 HDAC4
 Hyperphosphatasia with mental retardation, brachytelephalangy, and distinct face AR 239300   247262 PIGV
 Brachydactyly-hypertension syndrome (Bilginturian) AD 112410   1276  
 Brachydactyly with anonychia (Cooks syndrome) AD 106995   1487 SOX9
 Microcephaly-oculo-digito-esophageal-duodenal syndrome (Feingold syndrome) AD 164280 7050 1305 MYCN
 Hand-foot-genital syndrome AD 140000 1423 2438 HOXA13
 Brachydactyly with elbow dysplasia (Liebenberg syndrome) AD 186550   1275 PITX1
 Keutel syndrome AR 245150   85202 MGP
 Albright hereditary osteodystrophy (AHO) AD 103580   665 GNAS1
 Rubinstein-Taybi syndrome AD 180849 1526 783 CREBBP
 Rubinstein-Taybi syndrome AD 180849 1526 783 EP300
 Catel-Manzke syndrome XLR? 302380   1388  
 Brachydactyly, Temtamy type AR 605282     CHSY1
 Christian type brachydactyly AD 112450   1278  
 Coffin-Siris syndrome AR 135900 131811 1465 SMARCA2, SMARCA4, SMARCB1, SMARCE1, ARID1A, ARID1B
 Mononen type brachydactyly XLD? 301940   2565  
 Poland anomaly SP 173800   2911  

Please also refer to group 20 for other conditions with brachydactyly as well as brachytelephalangic CDP

  1. Limb hypoplasia—reduction defects group

Fanconi anemia can present with bone marrow failure, developmental delay and central nervous system malformation, short stature, skeletal anomalies often involving the radial ray, anomalies of the eyes, kidneys and urinary tract, ears (including deafness), heart, gastrointestinal system, abnormal skin pigmentation, and hypogonadism. There is an increased risk of malignancy.

 

Group/name of disorder Inher. OMIM GR Orpha Gene
 Ulnar-mammary syndrome AD 181450   3138 TBX3
 de Lange syndrome AD 122470 1104 199 NIPBL
 Fanconi anemia AR 227650 1401 84 Several genes, see OMIM
 Thrombocytopenia-absent radius (TAR) AR?/AD? 274000 23758 3320 Several
 Thrombocythemia with distal limb defects AD     329319 THPO
 Holt-Oram syndrome AD 142900 1111 392 TBX5
 Okihiro syndrome (Duane—radial ray anomaly) AD 607323 1373 959 SALL4
 Cousin syndrome AR 260660   93333 TBX15
 Roberts syndrome AR 268300 1153 3103 ESCO2
 Split-hand-foot malformation with long bone deficiency (SHFLD1) AD 119100   3329  
 Split-hand-foot malformation with long bone deficiency (SHFLD2) AD 610685   3329  
 Split-hand-foot malformation with long bone deficiency (SHFLD3) AD 612576   3329  
 Tibial hemimelia AR 275220   93322  
 Tibial hemimelia-polysyndactyly-triphalangeal thumb AD 188770   3332  
 Acheiropodia AR 200500   931 LMBR1
 Tetra-amelia XL 301090 1276 3301  
 Tetra-amelia AR 273395 1276 3301 WNT3
 Ankyloblepharon-ectodermal dysplasia-cleft lip/palate (AEC) AD 106260 43797 1071 TP63
 Ectrodactyly-ectodermal dysplasia cleft-palate syndrome Type 3 (EEC3) AD 604292   1896 TP63
 Ectrodactyly-ectodermal dysplasia cleft-palate syndrome type 1 (EEC1) AD 129900   1896  
 Ectrodactyly-ectodermal dysplasia-macular dystrophy syndrome (EEM) AR 225280   1897 CDH3
 Limb-mammary syndrome (including ADULT syndrome) AD 603543 43797 69085 TP63
 Split hand-foot malformation, isolated form, type 4 (SHFM4) AD 605289 43797 2440 TP63
 Split hand-foot malformation, isolated form, type 1 (SHFM1) AD 183600   2440  
 Split hand-foot Malformation, isolated form, type 2 (SHFM2) XL 313350   2440  
 Split hand-foot malformation, isolated form, type 3 (SHFM3) AD 246560   1307 FBXW4
 Split hand-foot malformation, isolated form, type 5 (SHFM5) AD 606708   2440  
Split-hand/foot malformation 1 with sensorineural hearing loss AR 220600   71271 DLX5
Split-hand/foot malformation 6 AR 225300   2440 WNT10B
 Al-Awadi Raas-Rothschild limb-pelvis hypoplasia-aplasia AR 276820   2879 WNT7A
 Fuhrmann syndrome AR 228930   2854 WNT7A
 RAPADILINO syndrome AR 266280 1204 3021 RECQL4
 Adams-Oliver syndrome AD/AR 100300   974 ARHGAP31, DOCK6, RBPJ, EOGT
 Femoral hypoplasia-unusual face syndrome (FHUFS) SP/AD? 134780   1988  
 Femur-fibula-ulna syndrome (FFU) SP? 228200   2019  
 Hanhart syndrome (hypoglossia-hypodactylia) AD 103300   989  
 Scapulo-iliac dysplasia (Kosenow) AD 169550   2839  

Please also refer to CHILD in group 20 and the mesomelic and acromesomelic dysplasias

  1. Polydactyly-Syndactyly-Triphalangism group

 

Pallister-Hall syndrome manifests with hypothalamic hamartoma, pituitary dysfunction, bifid epiglottis, laryngotracheal cleft, central polydactyly, and visceral malformations.

Meckel syndrome presents with variable combinations of renal cysts, developmental anomalies of the central nervous system (occipital encephalocele), hepatic ductal dysplasia and cysts, and polydactyly.

Group/name of disorder Inher. OMIM GR Orpha Gene
 Preaxial polydactyly type 1 (PPD1) AD 174400   93339 SHH
 Preaxial polydactyly type 1 (PPD1) AD 174400   93339 Other locus
 Preaxial polydactyly type 2 (PPD2)/triphalangeal thumb (TPT) AD 174500   2950 SHH
 Preaxial polydactyly type 3 (PPD3) AD 174600   93337 Other locus
 Preaxial polydactyly type 4 (PPD4) AD 174700   93338 GLI3
 Greig cephalopolysyndactyly syndrome AD 175700 1446 380 GLI3
 Pallister-Hall syndrome AD 146510 1465 672 GLI3
 Synpolydactyly (complex, fibulin1—associated) AD 608180   295197 FBLN1
 Synpolydactyly AD 186000   295195 HOXD13
 Townes-Brocks syndrome (Renal-Ear-Anal-Radial syndrome) AD 107480 1445 857 SALL1
 Lacrimo-auriculo-dento-digital syndrome (LADD) AD 149730   2363 FGFR2, FGFR3, FGF10
 Acrocallosal syndrome AR 200990   36 KIF7
 Acro-pectoral syndrome AD 605967   85203  
 Acro-pectoro-vertebral dysplasia (F-syndrome) AD 102510   957  
 Mirror-image polydactyly of hands and feet (Laurin-Sandrow syndrome) AD 135750   2378 SHH
 Mirror-image polydactyly of hands and feet (Laurin-Sandrow syndrome)         Other locus
 Cenani-Lenz syndactyly AR 212780   3258 LRP4
 Cenani-Lenz like syndactyly SP (AD?)       GREM1, FMN1
 Oligosyndactyly, radio-ulnar synostosis, hearing loss, and renal defects syndrome SP (AR?)       FMN1
 Syndactyly, Malik-Percin type AR 609432   157801 BHLHA9
 STAR syndrome (syndactyly of toes, telecanthus, ano-, and renal malformations) XL 300707   140952 FAM58A
 Syndactyly type 1 (III-IV) AD 185900   93402  
 Syndactyly type 3 (IV-V) AD 185900   93402 GJA1
 Syndactyly type 4 (I-V) Haas type AD 186200   93405 SHH
 Syndactyly type 5 (syndactyly with metacarpal and metatarsal fusion) AD 186300   93406 HOXD13
 Syndactyly with craniosynostosis (Philadelphia type) AD 601222   1527  
 Syndactyly with microcephaly and mental retardation (Filippi syndrome) AR 272440   3255 CKAP2L
Jawad syndrome AR 251255   313795 RBBP8
 Meckel syndrome type 1 AR 249000   564 MKS1
 Meckel syndrome type 2 AR 603194   564 TMEM216
 Meckel syndrome type 3 AR 607361   564 TMEM67
 Meckel syndrome type 4 AR 611134   564 CEP290
 Meckel syndrome type 5 AR 611561   564 RPGRIP1L
 Meckel syndrome type 6 AR 612284   564 CC2D2A

Note: the Smith-Lemli-Opitz syndrome can present with polydactyly and/or syndactyly. Please also refer to the SRPS group.

  1. Defects in joint formation and synostoses

Proximal symphalangism is characterized by fusion of the proximal interphalangeal joints, but can also involve the elbows, ankles and wrists leading to ankylosis. Conductive deafness secondary to fusion of the ossicles is also seen.

 

Group/name of disorder Inher. OMIM Orpha Gene
 Multiple synostoses syndrome type 1 AD 186500 3237 NOG
 Multiple synostoses syndrome type 2 AD 186500 3237 GDF5
 Multiple synostoses syndrome type 3 AD 612961 3237 FGF9
 Proximal symphalangism type 1 AD 185800 3250 NOG
 Proximal symphalangism type 2 AD 185800 3250 GDF5
 Radio-ulnar synostosis with amegakaryocytic thrombocytopenia AD 605432 71289 HOXA11

Please also refer to Spondylo-Carpal-Tarsal dysplasia; mesomelic dysplasia with acral synostoses; and others.

 

References

 

  1. M. L. Warman et al., Nosology and classification of genetic skeletal disorders: 2010 revision. American journal of medical genetics. Part A 155A, 943 (May, 2011).
  2. R. E. Stevenson, J. G. Hall, R. M. Goodman, Human malformations and related anomalies. (Oxford University Press, New York, 1993).
  3. J. r. W. Spranger, Bone dysplasias : an atlas of genetic disorders of skeletal development. (Oxford University Press, Oxford ; New York, ed. 3rd, 2012), pp. xxiii, 802 p.
  4. Y. Alanay, R. S. Lachman, A review of the principles of radiological assessment of skeletal dysplasias. Journal of clinical research in pediatric endocrinology 3, 163 (2011).
  5. S. R. Rose, M. G. Vogiatzi, K. C. Copeland, A general pediatric approach to evaluating a short child. Pediatrics in review / American Academy of Pediatrics 26, 410 (Nov, 2005).
  6. J. Spranger, A. Winterpacht, B. Zabel, The type II collagenopathies: a spectrum of chondrodysplasias. European journal of pediatrics 153, 56 (Feb, 1994).
  7. N. H. Elcioglu, C. M. Hall, Diagnostic dilemmas in the short rib-polydactyly syndrome group. American journal of medical genetics 111, 392 (Sep 1, 2002).
  8. S. Naik, I. K. Temple, Coat hanger appearances of the ribs: a useful diagnostic marker of paternal uniparental disomy of chromosome 14. Archives of disease in childhood 95, 909 (Nov, 2010).
  9. S. Unger, L. Bonafe, A. Superti-Furga, Multiple epiphyseal dysplasia: clinical and radiographic features, differential diagnosis and molecular basis. Best practice & research. Clinical rheumatology 22, 19 (Mar, 2008).
  10. Z. Stark, R. Savarirayan, Osteopetrosis. Orphanet journal of rare diseases 4, 5 (2009).
  11. R. Lachman et al., Radiologic and neuroradiologic findings in the mucopolysaccharidoses. Journal of pediatric rehabilitation medicine 3, 109 (2010).
  12. K. Chun, A. S. Teebi, C. Azimi, L. Steele, P. N. Ray, Screening of patients with craniosynostosis: molecular strategy. American journal of medical genetics. Part A 120A, 470 (Aug 1, 2003).
  13. G. C. Schwabe, S. Mundlos, Genetics of congenital hand anomalies. Handchirurgie, Mikrochirurgie, plastische Chirurgie : Organ der Deutschsprachigen Arbeitsgemeinschaft fur Handchirurgie : Organ der Deutschsprachigen Arbeitsgemeinschaft fur Mikrochirurgie der Peripheren Nerven und Gefasse 36, 85 (Apr-Jun, 2004).
  14. S. A. Temtamy, M. S. Aglan, Brachydactyly. Orphanet journal of rare diseases 3, 15 (2008).
  15. S. Mundlos, The brachydactylies: a molecular disease family. Clinical genetics 76, 123 (Aug, 2009).

 

 

Premenstrual Dysphoric Disorder (Formerly Premenstrual Syndrome)

 ABSTRACT

Premenstrual syndrome, the recurrent luteal phase deterioration in quality of life due to disruptive physical and psychiatric symptomatology, is a distinct clinical condition caused by an abnormal central nervous system response to the hormonal changes of the female reproductive cycle. Better definition and research based on strict inclusion/ exclusion criteria have allowed the development of successful treatments that are tailored to the severity of the lifestyle disruption and the specific individual constellation of symptoms. Charting and simple lifestyle changes may improve coping skills for many women. However, more severely affected individuals often require medical interventions to augment central serotonin/ norepinephrine levels or to suppress the hormonal changes of the menstrual cycle. For extended coverage of this and related topics, please see our FREE on-line web- text www.endotext.org.

INTRODUCTION

In the past fifty years premenstrual syndrome (PMS) has emerged as a well recognized phenomenon for which effective treatments are available. Unfortunately, because of the widespread public awareness of adverse premenstrual experiences, the term PMS has found usage in popular vernacular as a noun, adjective and verb (I’m PMS ing”). Over-the-counter remedies, often promoted by those who hope to profit by marketing a “sure cure” for a common condition, have exploited the fact that many women believe they suffer from PMS. Researchers have argued that there is a need to discriminate between the usual premenstrual experience of ovulatory women (wherein premenstrual molimina forewarn of impending menstruation or where more troublesome symptoms (PMS) are an annoyance) from Premenstrual Dysphoric Disorder (PMDD) wherein symptoms, particularly psychiatric, lead to major distress that is sufficient to interfere with day-to-day activities and disrupt interpersonal relationships. The challenge to the medical profession is to differentiate between these conditions and to offer appropriate and timely interventions.

 

Those with annoying premenstrual symptoms should be counseled about simple lifestyle changes that may attenuate these whereas those with marked psychiatric components such as irritability, anger, anxiety, or depression warrant early intervention with medications. Although the literature on PMS has focused almost entirely on women with adverse premenstrual experiences, there is evidence that 5-15% of women may experience positive changes in the premenstrum (1). Rarely do such women present challenges to the clinician. This chapter will review diagnosis, etiologic theories, and therapeutic approaches to adverse premenstrual experiences.

 

DEFINITIONS AND PREVALENCE:

Molimina, Premenstrual Syndrome [PMS], and Premenstrual Dysphoric Disorder [PMDD]

During the reproductive years, up to 80-90% of menstruating women will experience symptoms [breast pain, bloating, acne, constipation] that forewarn them of impending menstruation, so-called premenstrual molimina. Over 60% of women report swelling or bloating (2) although objective documentation of weight gain is lacking in most of these women (3). Cyclic breast symptoms affect 70% of women with 22% reporting moderate to extreme discomfort (4). Available data suggest that as many as 30%- 40% of these women are sufficiently bothered by molimina to seek relief.

 

The term PMS continues to be used; however, for the reasons mentioned above it may encompass a wide range of severity and therefore is not particularly useful in defining cohorts for research or in directing the most appropriate therapeutic interventions.

 

PMDD should be reserved for a more severe constellation of symptoms, mostly psychiatric, that lead to periodic interference with day-to-day activities and interpersonal relationships (5). Women with this degree of symptoms probably comprise 3-5% of women in their reproductive years (6, 7, 8).

 

Premenstrual Dysphoric Disorder now appears in the Diagnostic and Statistical Manual of Mental Health Disorders (fifth edition) of the American Psychiatric Association. After years of debate about whether this should be included as a distinct psychiatric condition (9,10), the importance of alerting psychiatrists to the critical involvement of the menstrual cycle in psychiatric disorders is now widely accepted (Table 1).

Table 1. Diagnostic Criteria for Premenstrual Dysphoric Disorder (PMDD)

Timing of symptoms
A)In the majority of menstrual cycles, at least 5 symptoms must be present in the final week before the onset of menses, start to improve within a few days after the onset of menses, and become minimal or absent in the week postmenses

Symptoms

B) One or more of the following symptoms must be present:1)    Marked affective lability (e.g., mood swings, feeling suddenly sad or tearful, or increased sensitivity to rejection)2)    Marked irritability or anger or increased interpersonal conflicts

3)    Markedly depressed mood, feelings of hopelessness, or self-deprecating thoughts

4)    Marked anxiety, tension, and/or feelings of being keyed up or on edge

 

C) One (or more) of the following symptoms must additionally be present to reach a total of 5 symptoms when combined with symptoms from criterion B above

1)     Decreased interest in usual activities

2)     Subjective difficulty in concentration

3)     Lethargy, easy fatigability, or marked lack of energy

4)     Marked change in appetite; overeating or specific food cravings

5)     Hypersomnia or insomnia

6)     A sense of being overwhelmed or out of control

7)     Physical symptoms such as breast tenderness or swelling; joint or muscle pain, a sensation of “bloating” or weight gain

 


Severity

D) The symptoms are associated with clinically significant distress or interference with work, school, usual social activities, or relationships with others.
E) Consider Other Psychiatric Disorders The disturbance is not merely an exacerbation of the symptoms of another disorder, such as major depressive disorder, panic disorder, persistent depressive disorder (dysthymia) or a personality disorder (although it may co-occur with any of these disorders).

Confirmation of the disorder

F) Criterion A should be confirmed by prospective daily ratings during at least 2 symptomatic cycles (although a provisional diagnosis may be made prior to this confirmation)Exclude other Medical Explanations

G) The symptoms are not attributable to the physiological effects of a substance (e.g., drug abuse, medication or other treatment) or another medical condition (e.g., hyperthyroidism).

 

(Adapted from: American Psychiatric Association: Diagnostic and Statistical manual of Mental Health Disorders, 5th edition. Washington D.C.2013 ) (11)

 

EPIDEMIOLOGY

It is likely that PMS has emerged as a twentieth century phenomenon in part due to the fact that women’s increasing control over reproduction has eliminated the cycle of repeated pregnancy and lactation that formerly characterized the lives of women from puberty to menopause (13). PMS-like behaviour has been reported both in humans and in non-human primates as long as they demonstrate menstrual cyclicity. In the non-human primate, zoologists have noted premenstrual changes in behaviour and appetite similar to those reported by women with PMS (14, 15).

 

PMS may affect woman at any stage of reproductive life. The common belief that PMS is a disorder of the older woman may have stemmed from the fact that mood swings in the teen are less likely to be considered an effect of menstrual cyclicity and more likely to be attributed to the “hormonal swings and heartbreaks” of adolescence. Severe PMS may start shortly after puberty and such cases tend to be recognized and brought to medical attention by a parent who recognizes the symptoms from her own experience. Little is known about the inheritance of PMS; however, there is support for a genetic predisposition. Surveys have found that as many as 70% of daughters of affected mothers were themselves PMS sufferers, whereas 63% of daughters of unaffected mothers were symptom free (16). PMS sufferers often relate that symptoms become progressively worse over time, and since women have increasing contact with health care providers for non-pregnancy related concerns in their later reproductive years, this may account for the preponderance of older women seeking help for PMS.

 

PMS disappears during suppression of the ovarian cycle (for example, during hypothalamic amenorrhea due to excessive physical, or nutritional stress, during lactational amenorrhea, during pregnancy, and after menopause – either natural or induced) (17). It is useful when evaluating a woman with suspected PMS to confirm that PMS symptoms did indeed disappear in these circumstances. Contrary to the popular belief, there is no convincing evidence that PMS begins after pregnancy or tubal ligation. This belief probably originated when PMS symptoms reappeared and seemed acutely worse after the hormonal “protection” of pre-existing pregnancy or lactation.

 

PMS disappears after natural, medically or surgically induced menopause although the reintroduction of exogenous hormone replacement therapy may be associated with the reappearance of symptoms (18, 19). Typically, the use of sequential progestin triggers PMS symptoms in susceptible women whereas continuous combined hormone replacement therapy is less likely to be associated adverse mood changes.

 

DIAGNOSIS

In 2008 an international multidisciplinary group of experts met at a face-to-face consensus meeting in Montreal, Canada, to review current definitions and diagnostic criteria for Premenstrual Disorders (PMD) (20). This group defined “Core Premenstrual Disorders (Core PMD) and Variant Premenstrual Disorders (Variant PMD)” as shown in Table 2 below.

Table 2 Classification of premenstrual disorders (PMD)
PMD category Characteristics
Core PMD Symptoms occur in ovulatory cycles
Symptoms are not specified—they may be somatic and/or psychological

 

The number of symptoms is not specified

 

Symptoms are absent after menstruation and before ovulation

 

They must recur in luteal phase

 

They must be prospectively rated (two cycles minimum)

 

Symptoms must cause significant impairment

 

Variants of PMD

 

Premenstrual exacerbation Symptoms of an underlying psychological or somatic disorder significantly worsen premenstrually
PMD due to non-ovulatory ovarian activity Symptoms arise from continued ovarian activity even though menstruation has been suppressed
Progestogen induced PMD Symptoms result (rarely) from ovarian activity other than those of ovulation
PMD with absent menstruation Symptoms result from exogenous progestogen administration
a Work, school, social activities, hobbies, interpersonal relationships, distress

 

Adapted from O'Brien PM. Backstrom T. Brown C. et al. Towards a consensus on diagnostic criteria, measurement and trial design of the premenstrual disorders: the ISPMD Montreal consensus. Arch Women's Mental Health 2011; 14(1):13-21

History

Physicians should make an effort to enquire about premenstrual symptoms as part of the menstrual and reproductive history of all women of reproductive age. For the woman with few symptoms, this provides education about molimina /PMS and may forestall fears that she is “losing her mind” should symptoms emerge in the later reproductive years. For the woman with significant symptoms, this will create the opportunity for counseling and reassurance and will set the stage for establishing the diagnosis and selecting appropriate therapy.

 

A typical woman with PMDD may relate that she is a productive employee and good mother for most of the month. However, starting sometime after ovulation (often 7-10 days prior to menstruation) she awakens in the morning with feelings of irritability, anger, anxiety, or sadness. At work, she may experience feelings of paranoia and wonder if co-workers are picking on her. Often she will report that she has difficulty concentrating on the task at hand. She may experience menopausal-like hot flashes and night sweats and often reports sleep disruption with vivid dreams. She states that premenstrually she overreacts to things that her children normally do around the house, and this makes her feel like a bad mother. She may feel down but be unable to understand why because she knows she has a good spouse, a good job, and healthy, happy, children. Minor things that her spouse says may be enough to trigger an argument, and nothing the spouse says can appease her. Although she would like to be held and comforted at such times, she reports that she cannot stand to be touched. In severe cases, she may try to isolate herself by locking the door to her room or unplugging her telephone. Occasionally depression, anger and aggression, or anxiety may be extreme, resulting in concerns for the welfare of the affected woman or her family members.

 

Caution is needed in immediately accepting such a typical sounding history as diagnostic of PMDD. Researchers have found that many other psychiatric conditions worsen premenstrually (so-called premenstrual exacerbation); hence, an individual with an underlying psychiatric disorder may recall and relate the symptoms that were most severe in the premenstrual week while ignoring the lower level of symptoms that exist throughout the month . Only by obtaining a prospective symptom record over a one- to two-month period can the clinician have confidence in the diagnosis. Any calendar used for this purpose must obtain information on four key areas: symptoms, severity, timing in relation to the menstrual cycle, and baseline level of symptoms in the follicular phase (Table 3). Information should be sought about stresses related to the woman's occupation and family life, as these may tend to exacerbate PMDD. Past medical and psychiatric diagnoses may be relevant in that a variety of medical and psychiatric disorders may show premenstrual exacerbation.

Table 3. Key elements of a prospective symptom record used for the diagnosis of PMDD.
  1. Daily listing of symptoms
  2. Ratings of symptom severity throughout the month
  3. Timing of symptoms in relation to menstruation
  4. Rating of baseline symptom severity during the follicular phase

Several of the medical interventions described below will work for both PMDD and other psychiatric conditions so that a pretreatment diagnosis is important in determining the most appropriate long term management of the condition.

 

Typically premenstrual symptoms appear after ovulation and worsen progressively leading up to menstruation. About 5-10% of PMS sufferers experience a brief burst of typical PMS symptoms coincident with the midcycle fall in estradiol that accompanies ovulation (21) (Figure 1). Premenstrual symptoms resolve at varying rates after onset of menstruation. In some women, there is almost immediate relief from psychiatric symptoms with the onset of bleeding while for others the return to normal is more gradual. The most severely affected women report that symptoms begin shortly after ovulation (two weeks before menstruation) and resolve at the end of menstruation. Such individuals typically report having only one “good week” per month (Figure 2). If this pattern is longstanding, then it becomes harder and harder for interpersonal relationships to rebound during the good week, with the result that the condition may start to take on the appearance of a chronic mood disorder. [Whenever charting leaves the diagnosis in doubt, a three-month trial of medical ovarian suppression (see below) will usually provide a definitive answer.]

Figure 1Figure 1

Figure 2Figure 2

One example of such a calendar record, the PRISM Calendar (Prospective Record of the Impact and Severity of Menstrual symptoms) (Figure 3) (9) allows rapid visual confirmation of the nature, timing, and severity of menstrual cycle-related symptomatology and at the same time provides information on life stressors and current therapies. Although symptoms are rated in severity on a scale from 1-3, the actual interpretation of the calendar requires no mathematical calculations. An arms length assessment of the month-long calendar usually allows a rapid distinction to be made between PMDD and other more chronic conditions (Figure 4). Other charting instruments, including the validated Daily Record of Severity of Problems (DRSP), the Premenstrual Symptoms Screening Tool (PSST), and the Calendar of Premenstrual Experiences (COPE), have been recently reviewed (22).

Figure 3

Figure 3

Figure 4Figure 4

Positive premenstrual changes associated with enhanced mood or performance are reported by up to 15% of women. Increased energy, excitement and well-being have been associated with increased activity, heightened sexuality and improved performance on certain types of tasks during the premenstrual phase. (1)

 

Physical Findings

There are no characteristic physical findings in women with PMS. When seen in the follicular phase of the cycle, PMS sufferers typically appear entirely normal. Premenstrually, a woman presenting with an acute episode of PMDD may appear anxious, tearful, or angry, depending on the nature of her symptom complex.

 

A thorough physical exam, including gynecological examination, is recommended in the assessment of all women being evaluated for PMDD. Organic causes of premenstrual symptoms must be ruled out. Marked fatigue may result from anemia, leukemia, hypothyroidism, or diuretic-induced potassium deficiency. Headaches may be due to intracranial lesions. Women attending clinics with premenstrual complaints have been found to have brain tumours, anemia, leukemia, thyroid dysfunction, gastrointestinal disorders, pelvic tumours including endometriosis, and other recurrent premenstrual phenomena such as arthritis, asthma, epilepsy, and pneumothorax (23).

 

Blood work

There is no endocrine test that helps in establishing the diagnosis in most circumstances (20). In a woman in whom the natural ovarian cycle has been disguised following hysterectomy, a serum progesterone determination at the time of symptoms may help to confirm the link between symptoms and the luteal phase of the cycle. At times a CBC and/or sensitive TSH may be indicated to rule out anemia, leukemia, or thyroid dysfunction as an explanation for symptoms.

 

ETIOLOGY

Although many theories of etiology have been proposed and disproved for this poorly understood condition, contemporary work suggests that PMDD is the result of an aberrant response of central neurotransmitters to normal changes in gonadal steroids during the menstrual cycle.

 

Other theories, while having some biological plausibility, have not or cannot be confirmed with available diagnostic techniques. No one theory has gained universal acceptance although consensus is developing that in some susceptible women normal swings in gonadal hormones appear to mediate changes in the activity of central neurotransmitters, such as serotonin, that in turn incite profound changes in mood and behaviour. Although it is likely that many of the physical symptoms (breast tenderness, bloating constipation) are the direct effect of gonadal steroids, it is intriguing that treatment of PMS with selective serotonin reuptake inhibitors will ameliorate the severity of not only psychological but also physical complaints.

 

Several lines of evidence from clinical medicine support this interrelationship between estrogen or lack of estrogen effect (perhaps mediated by progestin-induced depletion of estrogen receptors) and central serotonergic activity (24,25). Estrogen has been shown to alleviate clinical depression in hypoestrogenic women in double-blind clinical trials (26). The addition of sequential progestin therapy to estrogen replacement triggers characteristic PMS-like mood disturbance in some susceptible postmenopausal women (19). Anti-estrogens given for ovulation induction may, at times, provoke profound mood disruption. Women with premenstrual syndrome show a surprisingly high frequency of premenstrual and menstrual hot flashes (85% of PMS sufferers vs 15% of non- PMS controls) that are typical of those experienced by menopausal women (27, 28). Selective serotonin reuptake inhibitors (SSRIs) have been shown to relieve hot flashes in breast cancer survivors made menopausal by chemotherapy (29). In each of these circumstances a decrease in exposure to estrogen has been linked to mood disturbance, and in each case a decrease in serotonin activity (inferred from the response to SSRIs) appears to be the proximate cause [Figure 5].

Figure 5Figure 5

An emerging theory as to causation of PMDD involves a progesterone metabolite, allopregnanolone (ALLO), which acts centrally as a neuroactive steroid. As with progesterone, ALLO increases in the luteal phase and declines just prior to onset of menses. ALLO has a stimulating effect on the GABA-A receptor similar to alcohol and benzodiazepines with anxiolytic and sedative properties. One possibility is that women with PMDD have developed tolerance to the sedating GABA-a enhancing effects of ALLO (30). Preclinical and early clinical work have suggested that blockade of the production of ALLO with a 5-alpha reductase inhibitor can attenuate symptoms of PMDD (31).

 

 

THERAPY

Many women suffering from PMDD have suffered the fate of those with other poorly defined illnesses that lack a discrete diagnostic test. All too often their concerns have been dismissed as “a normal part of being a woman” and therapy has been denied. Typically affected women will suffer for long periods before seeking treatment, and most will have tried a variety of ineffective over-the–counter “PMS remedies”. Like other areas of confusion and uncertainty, the area of PMS is an attractive one for those promoting unorthodox treatments for personal gain. Many of the theories about causation of PMDD in the past 50 years appear to have emerged as a means to market specific therapeutic products. Much effort has been expended by conscientious investigators in an effort to rigorously evaluate the promotional claims of others. Randomized controlled trials have failed to confirm the efficacy of most of these purported treatments.

Lifestyle modification:

1) Communication strategies

When an individual is suffering to a degree that requires more than simple counselling and reassurance, measures aimed at lifestyle modification should first be explored. She should be encouraged to discuss the problem with those individuals who are central to her life, including spouse, other family members, and even a sympathetic co-worker. Often confrontations can be avoided if an understanding spouse or friend recognizes the cause for her upset and defers discussion of the controversial subject until another time. Strategies for stress reduction can be helpful. Communication skills and assertiveness may be improved with counselling. Group counselling in a program supervised by a clinical psychologist may be invaluable. While it is useful for PMS sufferers to learn to anticipate times in the month when vulnerability to emotional upset and confrontation may be greatest, the strategy of making important decisions "only on the good days" falls apart if premenstrual symptoms last for more than just a few days per month. For some women distressing premenstrual symptoms may last for a full three weeks, and advising them to restrict their important activities to the remaining days of the month is neither helpful nor warranted. Interventions aimed at reducing symptoms are more appropriate in this circumstance.

2) Diet

While there have been many books written which describe specific "PMS diets," few of the recommendations contained therein are founded on scientific fact. Several simple dietary measures may afford relief for women with PMS.

Most women with PMDD, despite feelings of bloating and tension, show no absolute increase in weight, no change in girth and no signs of peripheral edema (3, 20). Sudden shifts from low-sodium, low-carbohydrate intake to a diet high in these constituents can account for weight gain of as much as 5 kg in 24 hours in rare cases (32). Cravings for salty and sweet foods are commonly reported by women with PMDD, and these dietary alterations may account for unusual cases of premenstrual edema. For this reason reduction in the intake of salt and refined carbohydrates may help prevent edema and swelling in occasional women with this manifestation of PMS.

 

Although a link between methylxanthine intake and premenstrual breast pain has been suggested, available data are not convincing (33, 34). Nevertheless, a reduction in the intake of caffeine may prove useful in women where tension, anxiety, and insomnia predominate.

 

Several lines of evidence indicate that there is a tendency to increased alcohol intake premenstrually (35), and women should be cautioned that excessive use of alcohol is frequently an antecedent factor in marital discord.

 

Anecdotal evidence suggests that small, more frequent meals may occasionally alleviate mood swings. Based on recent evidence that cellular uptake of glucose may be impaired premenstrually, there is, at least, some theoretical basis for this dietary recommendation (36). Carbohydrates may exhibit mood altering effects through a number of mechanisms (37), but attempts to improve premenstrual symptoms through dietary supplements have met with limited success (38). Calcium supplementation has been shown to be marginally superior to placebo in a randomized placebo controlled trial (39, 40).

 

3) Exercise

Exercise is reported to reduce premenstrual molimina in women running in excess of 50 km/cycle (41). Lesser amounts of regular aerobic exercise may relieve symptoms, at least temporarily, in many women (42). As part of an overall program of lifestyle modification, exercise may reduce stress by providing a time away from the home and by providing a useful outlet for any anger or aggression. Some PMS sufferers report that exercise promotes relaxation and helps them sleep at night.

 

 

Medical interventions

The primary factor directing the selection of therapy should be the intensity and impact of premenstrual symptoms. Symptoms that are causing major disruption to quality of life rarely respond to lifestyle modification alone, and efforts to push this approach often do nothing more than delay effective therapy. Conversely, minor symptoms or symptoms that are short-lived each month seldom justify major medical interventions.

Attention should always initially be directed to symptoms for which simple, established treatments exist. For example, dysmenorrhea or menorrhagia may be satisfactorily relieved with prostaglandin synthetase inhibitors or oral contraceptives.

 

Mefenamic acid (500 mg tid) in the premenstrual and menstrual weeks has outperformed placebo for the treatment of PMS in some, but not all, clinical trials (43,44). It is likely that many of the end stage mediators of premenstrual symptomatology are prostaglandins; hence, this prostaglandin synthetase inhibitor may be working through a general inhibition of prostaglandin activity. Due to this, it is an ideal adjunct for any woman with coexisting dysmenorrhea and menorrhagia. In practice, however, its effectiveness for PMDD where psychological symptoms predominate is disappointing. Mefenamic acid is contraindicated in women with known sensitivity to aspirin or those at risk for peptic ulcers.

 

Until relatively recently trials comparing oral contraceptive therapy to placebo have not shown a beneficial effect on mood in most circumstances (45), although extended cycle combined hormonal contraceptives (46) and oral contraceptives containing the progestin drospirenone (47) have proven superior to placebo in randomized clinical trials. . When contraception is required in a woman with PMDD, especially in teens and if there is coexisting dysmenorrhea or menorrhagia, extended cycle hormonal contraceptives or those containing drospirenone can be tried initially.

 

Published data in regard to the efficacy of pyridoxine (Vitamin B6) have been contradictory (48); however, this medication in proper dosages (100 mg OD) is, at worst, a safe placebo that becomes one part of an overall management plan for the women with distressing molimina that should include lifestyle modification and changes in diet. Patients should be cautioned that these medications do not work for all women and that increasing the dose of pyridoxine in an effort to achieve complete relief of symptoms may lead to peripheral neuropathy. Pyridoxine should be discontinued if there is evidence of tingling or numbness of the extremities.

 

Neither progestin therapy (49, 50) nor oil of evening primrose (51) have been shown to be efficacious for PMDD in controlled clinical trials.

 

Premenstrual mastalgia which affects up to 70% of women in reproductive age may occur in isolation from other distressing premenstrual symptoms and, as such, should be considered a moliminal symptom. Low dose danazol (100 mg OD) for several cycles followed by maintenance doses in the luteal phase only (50 mg OD) (52) can bring about dramatic relief of mastalgia in most women; however, higher dosages (400 mg OD) may be required to relieve other symptoms of PMDD (53). Mastalgia may also respond to tamoxifen (10 mg daily) (54), but has not been shown to respond to diuretics, medroxyprogesterone acetate, or pyridoxine.

 

The routine use of diuretics in the treatment of PMS should be abandoned. Most women show only random weight fluctuations during the menstrual cycle despite the common sensation of bloating. In the absence of demonstrable weight gain it is likely that this symptom may result from constipation and/or bowel wall edema rather than from an overall fluid accumulation. In rare cases, ingestion of salt and refined carbohydrates has been shown to result in true fluid retention. In cases where a consistent increase in weight can be documented or where edema is demonstrable, limitation of intake of salt and refined carbohydrates should be tried first. If such dietary restrictions fail to relieve premenstrual fluid accumulation, use of a potassium-sparing diuretic, such as spironolactone, may be considered (55). Continued use of a diuretic activates the renin–angiotensin–aldosterone system resulting in rapid rebound fluid accumulation as soon as the diuretic is discontinued. Weight takes approximately two to three weeks to return to normal after discontinuation of a diuretic in some people. Unfortunately this leaves the affected women with the impression that she must use a diuretic to maintain normal fluid balance.

 

Some women report overriding symptoms of anxiety and tension or insomnia in the premenstrual week (56). New short-acting anxiolytics or hypnotics such as alprazolam (.25 mg po bid) or triazolam (.25 mg po qhs) may be prescribed sparingly for such individuals (57, 58). Buspirone has also proven useful for anxiety and may be particularly helpful in circumstances where SSRIs evoke sexual dysfunction (59).

 

Estrogen withdrawal has been implicated in menstrually-related migraines, and recent evidence indicates that estrogen supplementation commencing in the late luteal phase and continued through menstruation may alleviate headaches in some women (60, 61, 62). As discussed below, if headaches are severe and are unrelieved by short term estrogen supplementation, they can often be nicely controlled by intramuscular or oral sumatriptan therapy (63) or by medical ovarian suppression with GnRH agonists (64, 65) and continuous combined hormone replacement therapy.

 

Antidepressant Therapy

A range of newer antidepressant medications that augment central serotonin activity have been shown to alleviate severe premenstrual syndrome (66, 67). Since these agents will also relieve endogenous depression, a pretreatment diagnosis, achieved by prospective charting, is very important. Practically speaking, many women who attend a gynecology clinic to seek relief from premenstrual symptoms express reservations about taking an antidepressant, particularly if a short-term endpoint (3-6 months away) is not likely. Long term therapy may be required to control symptoms of PMDD from the late 30s until menopause.

 

For patients in whom psychiatric symptoms predominate antidepressant therapy may provide excellent results (Figure 6). Selective serotonin re-uptake inhibitors, such as fluoxetine, sertraline, paroxetine, fluvoxamine, and venlafaxine (a serotonin and norepinephrine re-uptake inhibitor) have all been successfully employed.

 

Figure 6

Figure 6

Symptom profiles may help in selecting the most appropriate agent (i.e., fluoxetine in patients where fatigue and depression predominate; sertraline if insomnia, irritability, and anxiety are paramount). SSRIs have been associated with loss of libido and anorgasmia, which are particularly distressing to this patient population, and appropriate pretreatment counseling is essential.

 

Tricyclic antidepressants (TCA) have not generally been effective with the exception of clomipramine, a TCA with strong serotoninergic activity. Intolerance to the side effects of TCAs is common.

 

Most women with PMDD would prefer to medicate themselves only during the symptomatic phase of the menstrual cycle. Recent studies have demonstrated that luteal phase therapy and even symptom-onset therapy may be effective for many women with PMS (68, 69). Practically speaking, it makes sense to start a trial of SSRI therapy with continuous use. After a woman has determined the optimal response that can be achieved with continuous therapy, it is reasonable for her to try luteal phase-only or symptom-onset therapy (70) to determine if the benefit is maintained.

 

Medical Ovarian Suppression

Suppression of cyclic ovarian function may afford dramatic relief for the woman with severe and long lasting symptoms (71, 72) (Figure 7). In each case, therapy should be directed toward suppression of cyclic ovarian activity while ensuring a constant low level of estrogen sufficient to prevent menopausal symptomatology and side effects.

Figure 7

Figure 7

Danazol (200 mg bid) will effect ovarian suppression in approximately 80% of women with prompt relief from symptoms (53). It also reduces breast pain and menstrual flow. However, danazol is an impeded androgen and at a dosage of 200 mg bid may have side effects that limit its use, such as hot flashes, muscle cramps, hirsutism or a worsening of the lipid profile. Because of this, the use of danazol has been largely supplanted by ovarian suppression with gonadotropin-releasing hormone agonists (GnRH Ag).

 

Gonadotropin releasing hormone agonists effect rapid medical ovarian suppression, thereby inducing a pseudo-menopause and affording relief from PMS (71, 72). This approach may effectively alleviate other less common menstrual cycle-related conditions such as asthma, epilepsy, migraine and irritable bowel syndrome (65). This approach is unsatisfactory in the long term, not only because of the troublesome menopausal symptoms it evokes, but also because if creates an increased risk for osteoporosis and ischemic heart disease. When combined with continuous combined hormone replacement therapy, GnRH Ag afford excellent relief from premenstrual symptomatology without the attendant risks and symptoms resulting from premature menopause. The major drawback to this therapeutic approach is the expense of medication and the need for the patient to take multiple medications on a long-term basis. For women approaching menopause, this therapy (a GnRH Ag and continuous combined hormone replacement therapy) can be maintained until menopause with satisfactory symptom control. Some women, despite complete relief of symptoms, cannot afford or choose not to take this combination of medications for prolonged intervals (as long as 10-15 years from diagnosis until menopause in some cases).

 

Though less well studied depo-medroxyprogesterone acetate (depo-MPA) (150 -300 mg IM q3m) may provide a cheaper way to attenuate symptoms of PMDD in women who require contraception. The major drawback to this approach is that a substantial percentage of women will get irregular bleeding and gradual weight gain. Patients should always be counseled about the potential for protracted anovulation following use of this medication.

 

Surgical Therapy

Medical approaches to PMS should be considered and explored prior to any consideration of surgery for PMDD. For the woman in whom there is unequivocal documentation that premenstrual symptoms are severe and disruptive to lifestyle and relationships, and in whom conservative medical therapies have failed (either due to lack of response, intolerable side effects, or prohibitive cost), the effect of medical ovarian suppression should be tested.

 

Where the family is complete and permanent contraception is desired, the pros and cons of oophorectomy for lasting relief from premenstrual symptomatology should be discussed with the patient (if she has failed other medical treatments and responded to a clinical trial of medical ovarian suppression). Clinical trials have clearly shown that oophorectomy with subsequent hormone replacement therapy is effective in the treatment of PMDD (73, 74, 75). Concomitant hysterectomy will avoid the need for progestin as part of the hormone replacement regimen and may avoid irregular bleeding and progestin-induced recrudescence of symptoms. An international group of specialists with clinical experience in management of PMDD has recently published a detailed consensus document which reviews the efficacy of existing therapies (76).

 

REFERENCES:

 

  1. Logue CM, Moos RH. Positive perimenstrual changes: toward a new perspective on the menstrual cycle. J Psychosom Res 1988;32(1): 31-40
  2. Lee KA, Rittenhouse CA Prevalence of perimenstrual symptoms in employed women. Women Health 1991; 17(3): 17-32
  3. Faratian B, Gaspar A, O'Brien PM, Johnson IR, Filshie GM, Prescott P. Premenstrual syndrome: weight, abdominal swelling, and perceived body image. Am J Obstet Gynecol 1984;150(2):200-4
  4. Ader DN, South-Paul J, Adera T, Deuster PA. Cyclical mastalgia: prevalence and associated health and behavioural factors. J Psychosom Obstet Gynaecol 2001; 22(2): 71-76
  5. Reid RL, Yen SS. Premenstrual syndrome. Am J Obstet Gynecol 1981; 139(1): 85-104.
  6. Wood NF, Most A, Dery GK. Prevalence of perimenstrual symptoms. Am J Public Health 1982; 72:1257- 1264
  7. Johnston SR, McChesney C, Bean JA. Epidemiology of premenstrual symptoms in a non clinical sample. I. Prevalence, natural history, and help seeking behaviour. J Reprod Med 33:340-346, 1988
  8. Rivera-Tovar AD, Frank E. Late luteal phase dysphoric disorder in young women. Am J Psychiatry 1990; 147:1634-1636
  9. Reid RL. Premenstrual syndrome. Curr Prob Obstet Gynecol and Fertil 1985; 8:(2): 1-57
  10. Epperson CN, Steiner M, Hartlage SA et al. Premenstrual dysphoric disorder: evidence for a new category for DSM-5. Am J Psychiatry 2012; 169(5):465-475
  11. Hartlage SA, Breaux CA, Yonkers KA. Addressing concerns about the inclusion of Premenstrual Dysphoric Disorder in DSM-5. J Clin Psychiatry 2014; 75L1): 70-76
  12. American Psychiatric Association: Diagnostic and Statistical Manual of Mental Health Disorders, 5th edition. Washington, DC, American Psychiatric Association; 2013
  13. Reid RL. Premenstrual syndrome. NEJM 1991; 324(17):1208-1210
  14. Sassenrath EN, Rowell TE, Hendrickx AG. Perimenstrual aggression in groups of female rhesus monkeys. J Reprod.Fertil 1973; 34:509-513
  15. Gilbert C, Gillman J. The changing pattern of food intake and appetite during the menstrual cycle of the baboon with a consideration of some of the controlling hormonal factors. S Afr J Med 1956; 21: 75-89
  16. Kantero RL, WidholmO. Correlations of menstrual traits between adolescent girls and their mothers. Acta Obstet Gynecol. Scand. 1977 Suppl 14:30-42
  17. Reid RL. Premenstrual syndrome: a time for introspection. Am J Obstet Gynecol 1986; 155(5): 921-926
  18. Kirkham C, Hahn PM, Van Vugt DA, Carmichael JA, Reid RL. A randomized, double-blind, placebo-controlled, cross-over trial to assess the side effects of medroxyprogesterone acetate in hormone replacement therapy. Obstetrics & Gynecology 1991; 78(1): 93-97.
  19. Bjorn I, Bixo M, Nojd KS, Nyberg S, Backstrom T. Negative mood changes during hormone replacement therapy: a comparison between two progestogens. Am J Obstet Gynecol 2000;183(6): 1419-26
  20. O'Brien PM. Backstrom T. Brown C. Dennerstein L. Endicott J. Epperson CN. Eriksson E. Freeman E. Halbreich U. Ismail KM. Panay N. Pearlstein T. Rapkin A. Reid R. Schmidt P. Steiner M. Studd J. Yonkers K. Towards a consensus on diagnostic criteria, measurement and trial design of the premenstrual disorders: the ISPMD Montreal consensus. Arch Women's Mental Health 2011; 14(1):13-21
  21. Reid RL. Endogenous opioid activity and the premenstrual syndrome. Lancet 1983; 2(8353):786
  22. Renske C. Bosman RC, Jung SE, Miloserdov K, Schoevers RA, Rot M. Daily symptom ratings for studying premenstrual dysphoric disorder: A review. J Affect Disord 2016;189:43–53
  23. Reid RL, Yen SS. The premenstrual syndrome. Clinical Obstetrics & Gynecology 1983; 26(3): 710-718.
  24. Rubinow DR, Schmidt PJ, Roca CA. Estrogen-serotonin interactions: implications for affective regulation. Biol Psychiatry 1998; 44(9); 839-850
  25. Halbreich U, Kahn LS. Role of estrogen in the aetiology and treatment of mood disorders. CNS Drugs 2001; 15(10): 797-817
  26. Soares CN, Almeida OP, Joffe H, Cohen LS. Efficacy of estradiol for the treatment of depressive disorders in perimenopausal women: a double-blind, randomized, placebo-controlled trial. Arch Gen Psychiatry 2001; 58(6): 529-34
  27. Hahn PM, Wong J, Reid RL. Menopausal-like hot flushes reported in women of reproductive age. Fertil Steril 1998; 70(5): 913-918.
  28. Casper RF, Graves GR, Reid RL. Objective measurement of hot flushes associated with the premenstrual syndrome. Fertil Steril 1987; 47(2): 341-344.
  29. Stearns V, Isaacs C, Rowland J, Crawford J, Ellis MJ, Kramer R, Lawrence W, Hanfelt JJ, Hayes DF. A pilot trial assessing the efficacy of paroxetine hydrochloride (Paxil) in controlling hot flashes in breast cancer survivors. Ann Oncol 2000;11(1):17-22
  30. Hantsoo L, Epperson CN. Premenstrual dysphoric disorder: epidemiology and treatment. Curr Psychiatry Rep 2015:17:87 (1-9)
  31. Martinez PE, Rubinow DR, Nieman LK, Koziol DE, Morrow AL, Schiller CE, Cintron D, Thompson KD, Khine KK, Schmidt PJ. 5α-Reductase inhibition prevents the luteal phase Increase in plasma allopregnanolone levels and mitigates symptoms in women with premenstrual dysphoric disorder. Neuropsychopharmacology (2016) 41, 1093–1102
  32. MacGregor GA, Markander ND, Roulston JE, Jones JC, de Wardener HE. Is “idiopathic” edema idiopathic? Lancet 1979; 1:397-400
  33. Minton JP, Foecking MK, Webster DJ, Matthews RH. Caffeine, cyclic nucleotides, and breast disease. Surgery 1979; 86:105-109
  34. Rossignol AM, Bonnlander H. Caffeine-containing beverages, total fluid consumption, and premenstrual syndrome. Am J Publ Health 1990; 80:1106-1110
  35. Mello NK, Mendelson JH, Lex BW. Alcohol use and premenstrual symptoms in social drinkers. Psychopharmacology 1990; 101(4): 448- 455
  36. Diamond M, Simonson CD, DeFronzo RA. Menstrual cyclicity has a profound effect on glucose homeostasis. Fertil Steril 1989; 52: 204-208.
  37. Young SN. Clinical nutrition: 3. The fuzzy boundary between nutrition and psychopharmacology. CMAJ 2002; 166 (2): 205-20938.
  38. Sayegh,R. Schiff,I. Wurtman,J. Spiers,P. McDermott,J. Wurtman,R. The effect of a carbohydrate-rich beverage on mood, appetite, and cognitive function in women with premenstrual syndrome. Obstet Gynecol 1995; 86(4):1-839
  39. Thys-Jacobs S, Starkey P, Bernstein D, Tian J. Calcium carbonate and the premenstrual syndrome: effects on premenstrual and menstrual symptoms. Premenstrual Syndrome Study Group. Am J Obstet Gynecol 1998 ; 179(2):444-52
  40. Yonkers KA. Pearlstein TB. Gotman N A pilot study to compare fluoxetine, calcium, and placebo in the treatment of premenstrual syndrome J Clinl Psychopharmacol. 2013; 33(5):614-20
  41. Prior JC, Vigna Y, Sciarretta D, Alojado N, Schulzer M. Conditioning exercise decreases premenstrual symptoms: a prospective, controlled 6-month trial. Fertil Steril 1987 Mar;47(3):402-8
  42. Steege JF, Blumenthal JA. The effects of aerobic exercise of premenstrual symptoms in middle aged women: A preliminary study. J Psychosom Res 1993; 37 (2):127-133.
  43. Mira M, McNeil D, Fraser IS, Vizzard J, Abraham S. Mefenamic acid in the treatment of premenstrual syndrome. Obstet Gynecol 1986;68:395-398
  44. Wood C, Jakubowicz D. The treatment of premenstrual symptoms with mefenamic acid. Br J Obstet Gynaecol 1980; 87(7):627-30
  45. Collins J, Crosignani PG, and the ESHRE Capri Working Group. Non Contraceptive Health Benefits of Combined Oral contraception. Human Reprod Update 2005; 11(5):513-525
  46. Coffee AL, Kuehl TJ, Willis S, Sulak PJ. Oral contraceptives and premenstrual symptoms: comparison of a 21/7 and extended regimen. Am J Obstet Gynecol 2006;195:1311-9.
  47. Yonkers KA et al. Efficacy of a new low dose oral contraceptive with drospirenone in PMDD. Obstet Gynecol 2005; 106(3): 492-501
  48. Wyatt KM, Dimmock PW, Jones PW, O’Brien PMS. Efficacy of vitamin B6 in treatment of premenstrual syndrome: systematic review. BMJ 1999: 318: 1375-1381
  49. Maddocks S, Hahn P, Moller F, Reid RL. A double-blind placebo-controlled trial of progesterone vaginal suppositories in the treatment of premenstrual syndrome. Am J Obstet Gynecol 1986; 154(3): 573-581.
  50. Wyatt K, Dimmock P, Jones P, Obhrai M, O’Brien PMS. Efficacy of progesterone and progestogens in management of premenstrual syndrome: systematic review. BMJ 2001; 323 (7316); 776-780
  51. Budeiri D, Li Wan Po A, Dornan JC. Is Evening Primrose Oil of value in the treatment of premenstrual syndrome? Controlled Clin Trials 1996; 17:60-68
  52. Gorins A, Perret F, Tourant B, Rogier C, Lipszyc J. A French double-blind crossover study (danazol versus placebo) in the treatment of severe fibrocystic breast disease. Eur J Gynaecol Oncol 1984;5(2):85-9
  53. Hahn PM, Van Vugt DA, Reid RL. A randomized placebo controlled crossover trial of danazol for the treatment of premenstrual syndrome. Psychoneuroendocrinology 1995; 20 (2):193-209.
  54. Messinis IE, Lolis D. Treatment of premenstrual mastalgia with Tamoxifen. Acta Obstet Gynecol scand 1988; 67-307-309
  55. O'Brien PM, Craven D, Selby C, Symonds EM Treatment of premenstrual syndrome by spironolactone. Br J Obstet Gynaecol 1979 ; 86(2): 142-7
  56. Mauri M, Reid RL, MacLean AW. Sleep in the premenstrual phase: a self-report study of PMS patients and normal controls. Acta Psychiat Scand 1988; 78(1): 82-86.
  57. Harrison WM, Endicott J, Nee J. Treatment of premenstrual dysphoria with alprazolam. A controlled study. Arch Gen Psychiatry 1990; 47(3): 270-5
  58. Berger CP, Presser B. Alprazolam in the treatment of two subsamples of patients with late luteal phase dysphoric disorder: A double blind placebo controlled crossover study. Obstet Gynecol 1994; 84: 379-385
  59. Landen M, Eriksson O, Sundblad C, Andersch B, Naessen T, Eriksson E. Compounds with affinity for serotonergic receptors in the treatment of premenstrual dysphoria: a comparison of buspirone, nefazodone and placebo. Psychopharmacology 2001; 155(3): 292-8
  60. MacGregor A. Migraine associated with menstruation. Funct Neurol 2000; 15 Suppl 3:143-153
  61. De Lignieres B, Vincens M, Mauvais-JarvisP et al. Prevention of menstrual migraine by percutaneous oestradiol. Br Med J 1986; 293:1540
  62. Magos AL, Zilkha KJ, Studd JW. Treatment of menstrual migraine by oestradiol implants. J Neurol Neurosurg Psychiattry 1983; 46: 1044-1046
  63. Salonen R, Saiers J. Sumatriptan is effective in the treatment of menstrual migraine; a review of prospective studies and retrospective analyses. Cephalgia 1999; 19:16-19
  64. Murray SC, Muse KN. Effective treatment of severe menstrual migraine headaches with gonadotropin-releasing hormone agonist and "add-back" therapy. Fertil Steril 1997; 67(2): 390-3
  65. Case AM, Reid RL. Effects of the menstrual cycle on medical disorders. Arch Intern Med 1998; 158(13):1405-1412.
  66. Steiner M, Korzekwa M, Lamont J, Stewart D, Carter D, Misri S, Reid RL, Steinberg S, Berger C, Grover D. Fluoxetine in the treatment of premenstrual dysphoria. NEJM 1995; 332(23): 1529-1534
  67. Marjoribanks J, Brown J, O Brien PMS, Wyatt K. Selective serotonin inhibitors for premenstrual syndrome. Update of Cochrane Database Syst Rev 6:CD001396 2013
  68. Steiner M, Korzekwa M, Lamont J, Wilkins A. Intermittent fluoxetine dosing in the treatment of women with premenstrual dysphoria. Psychopharmacology Bull 1997; 33(4): 771-774
  69. Steiner M, Li T. Luteal phase and symptom-onset dosing of SSRIs/SNRIs in the treatment of premenstrual dysphoric disorder: clinical evidence and rationale CNS Drugs 2013; 27: 583-589
  70. Yonkers KA, Kornstein SG, Gueorguieva R, Merry B, Steenburgh KV, Altemus M. Symptom-onset dosing of sertraline for the treatment of premenstrual dysphoric disorder. A randomized clinical trial. JAMA Psychiatry. 2015;72(10):1037-1044.
  71. Muse KN, Cetel NS, Futterman L, Yen SSC. The premenstrual syndrome: Effects of "medical ovariectomy". NEJM 1984; 311: 1345-1349.
  72. Mezrow G, Shoupe D, Spicer D, Lobo R, Leung B, Pike M. Depot leuprolide acetate with estrogen and progestin add back for long-term treatment of premenstrual syndrome. Fertil Steril 1994; 62(5): 932-937
  73. Casper RF, Hearn MT. The effect of hysterectomy and bilateral oophorectomy in women with severe premenstrual syndrome. Am J Obstet Gynecol 1990; 162: 105-109.
  74. Casson P, Hahn P, VanVugt DA, Reid RL. Lasting response to ovariectomy in severe intractable premenstrual syndrome. Am J Ob Gynecol 1990;162:99-102
  75. Reid RL. When should surgical treatment for Premenstrual Dysphoric Disorder be considered? Premenstrual disorders. Menopause International 2012; 18(2):77-81
  76. O’Brien PM, Backstrom T, Brown C, Dennerstein L, Endicott J, Epperson E, Freeman E, Halbreich U, Ismail KM, Panay N. Pearlstein T, Rapkin A, Reid RL, Schmidt O, Steiner M, Studd J, Yonkers K. ISPMD Consensus on the management of premenstrual disorders. Arch Women’s Mental Health 2013; 16(4):279-291

Endocrinology of the Male Reproductive System and Spermatogenesis

 ABSTRACT

The testes synthesize two important products: testosterone, needed for the development and maintenance of many physiological functions; and sperm, needed for male fertility. The synthesis of both products is regulated by endocrine hormones produced in the hypothalamus and pituitary, as well as locally within the testis. Testosterone is indispensable for sperm production, however both testosterone and Follicle Stimulating Hormone (FSH) are needed for optimal testicular development and maximal sperm production. Sperm are produced via the extraordinarily complex and dynamic process of spermatogenesis that requires co-operation between multiple testicular cell types. While it has long been known that testosterone and FSH regulate spermatogenesis, years of research has shed light on many of the intricate mechanisms by which spermatogonial stem cells develop into highly specialized, motile spermatozoa. Spermatogenesis involves the concerted interactions of endocrine hormones, but also many paracrine and growth factors, tightly co-ordinated gene and protein expression programs as well as epigenetic modifiers of the genome and different non-coding RNA species. This chapter provides a comprehensive overview of the fascinating process of spermatogenesis and of its regulation, and emphasises the endocrine regulation of testicular somatic cells and germ cells. The chapter also provides a summary of the clinically significant aspects of the endocrine regulation of spermatogenesis. For complete coverage of all related areas of Endocrinology, please see our online FREE web-book, www.endotext.org.

 

CLINICAL SUMMARY

The testes synthesize two essential products: testosterone, needed for the development and maintenance of many physiological functions including normal testis function; and sperm, needed for male fertility. The synthesis of both products is regulated by endocrine hormones produced in the hypothalamus and pituitary, as well as locally within the testis.

 

The secretion of hypothalamic gonadotropin-releasing hormone (GnRH) stimulates production of luteinizing hormone (LH) and follicle stimulating hormone (FSH) by the pituitary. LH is transported in the blood stream to the testes, where it stimulates Leydig cells to produce testosterone: this can act as an androgen (via interaction with androgen receptors) but can also be aromatized to produce estrogens. The testes, in turn, feedback on the hypothalamus and the pituitary via testosterone and inhibin secretion, in a negative feedback loop to limit GnRH and gonodotropin production. Both androgens and FSH act on receptors within the supporting somatic cells, the Sertoli cells, to stimulate various functions needed for optimal sperm production. Spermatogenesis is the process by which immature male germ cells divide, undergo meiosis and differentiate into highly specialized haploid spermatozoa. Optimal spermatogenesis requires the action of both testosterone (via androgen receptors) and FSH.

 

Spermatogenesis takes place within the seminiferous tubules of the testis. These tubules form long convoluted loops that pass into the mediastinum of the testis and join an anastomosing network of tubules called the rete testis. Spermatozoa exit the testes via the rete and enter the efferent ductules prior to their passage through, and final maturation in, the epididymis. The seminiferous tubules are comprised of the seminiferous epithelium: the somatic Sertoli cells and the developing male germ cells at various stages of development. Surrounding the seminiferous epithelium is a layer of basement membrane and layers of modified myofibroblastic cells termed peritubular myoid cells. Between the tubules is the interstitial space that contains blood and lymphatic vessels, immune cells including macrophages and lymphocytes, and the steroidogenic Leydig cells.

 

Male germ cell development relies absolutely on the structural and nutritional support of the somatic Sertoli cells. Sertoli cells are large columnar cells, with their base residing on basement membrane on the outside of the seminiferous tubules, and their apical processes surrounding germ cells as they develop into spermatozoa. Androgens (and estrogens) and FSH act on receptors within Sertoli cells: germ cells lack both androgen and FSH receptors, therefore these hormones act directly on Sertoli cells to support spermatogenesis. Sertoli cells regulate the internal environment of the seminiferous tubule by secreting paracrine factors and expressing cell surface receptors needed for germ cell development. Sertoli cells form intercellular tight junctions at their base: these occluding junctions prevent the diffusion of substances from the interstitium into the tubules and create a specialized milieu required for germ cell development. These junctions are a major component of the so-called ‘blood-testis-barrier’, wherein the passage of substances from the circulation is prevented from entering the inner part of the seminiferous tubules. The most immature germ cells, including germline stem cells, reside near the basement membrane of the seminiferous tubules and thus have free access to factors from the interstitium, however germ cells undergoing meiosis and haploid cell differentiation develop “above” the blood-testis-barrier and thus are entirely reliant on the Sertoli cell microenvironment. The seminiferous tubules are also an immune-privileged environment. Meiotic and post-meiotic germ cells develop after the establishment of immune tolerance, and could thus be recognized as “foreign” by the immune system, therefore the seminiferous tubules, via a number of different mechanisms including the blood-testis-barrier, actively exclude immune cells and factors from entering the seminiferous tubules and being exposed to meiotic and haploid germ cells.

 

The number of Sertoli cells determines the ultimate spermatogenic output of the testes. In humans, Sertoli cells proliferate during the fetal and early neonatal period and again prior to puberty. At puberty, Sertoli cells cease proliferation and attain a mature, terminally differentiated phenotype that is able to support spermatogenesis. Disturbances to Sertoli cell proliferation during these times can result in smaller testes with lower sperm production. Conversely, disturbances to the cessation of proliferation can result in larger testes with more Sertoli cells and a greater sperm output. It seems likely that the failure of many men with congenital hypogonadotropic hypogonadism (HH) to achieve normal testicular size and sperm output, when treated by gonadotropic stimulation, may result from deficient Sertoli cell proliferation during fetal and prepubertal life. The action of both androgens and FSH on Sertoli cells is necessary for the ability of Sertoli cells to support full spermatogenesis. In addition, the expression of many genes and paracrine factors within Sertoli cells is necessary for spermatogenesis.

 

Spermatogenesis relies on the ability of Leydig cells to produce testosterone under the influence of LH. Fetal Leydig cells appear following gonadal sex differentiation (gestational weeks 7-8 in humans) and, under the stimulation of placental human chorionic gonadotropin (hCG), results in the production of testosterone during gestation. In humans, fetal cells decrease in number towards term and are lost from the interstitium at about twelve months of age. The adult population of Leydig cells in the human arises from the division and differentiation of mesenchymal precursor cells under the influence of LH at puberty. Factors secreted by Sertoli cells and peritubular myoid cells are also necessary for Leydig cell development and steroidogenesis. Optimal Leydig cell steroidogenesis also relies on a normal complement of macrophages within the testicular interstitium as well as on the presence of androgen receptors in peritubular myoid cells, presumably because these cells secrete factors necessary for Leydig cell development and function.

 

The process of spermatogenesis begins in the fetal testis, when the Sertoli cell population is specified in the embryonic testis under the influence of male sex determining factors, such as SRY and SOX9. Newly-specified Sertoli cells enclose and form seminiferous cord structures and direct primordial germ cells to commit to the male pathway of gene expression. Fetal Sertoli cells proliferate and drive seminiferous cord elongation; this process is also dependent on factors secreted by Leydig cells. In the neonatal testis, primordial germ cells undergo further maturation and migrate to the basement of the seminiferous tubules where they provide a pool of precursor germ cells for postnatal spermatogenesis.

 

Spermatogonia are the most immature germ cell type. This heterogeneous population includes spermatogonial stem cells, which self-renew throughout life to provide a pool of stem cells available for spermatogenesis, as well as proliferating cells that differentiate and become committed to entry into meiosis. Spermatogonial development is hormonally independent and as such they are present even in the absence of GnRH. Spermatogonia eventually differentiate into spermatocytes that proceed through the process of meiosis that begins with DNA synthesis resulting in a tetraploid gamete. During the long meiotic prophase, which lasts ~2 weeks, homologous chromosomes pair and meiotic recombination occurs; this involves the induction and repair of DNA double-strand breaks allowing the exchange of genetic information between paired chromosomes, thereby creating genetic diversity between gametes. At the end of prophase, the meiotic cells proceed through two rapid and successive reductive divisions to yield haploid spermatids. The completion of meiosis depends absolutely on androgen action in Sertoli cells; in the absence of androgen, no haploid spermatids will be produced.

 

Newly formed haploid round spermatids differentiate, with no further division, into the highly specialized spermatozoan during the process of spermiogenesis. This involves many complex processes, including development of the acrosome (an organelle on the surface of the sperm head that contains enzymes required to penetrate the zona pellucida of the oocyte and thus facilitate fertilization), the flagella (the motile microtubule-based structure required for sperm motility) and the remodelling of the spermatid’s DNA into a tightly coiled structure within a small, streamlined nucleus that will not hinder motility. This remodelling of the DNA involves the cessation of gene transcription up to 2 weeks prior to the final maturation of the sperm; therefore spermiogenesis involves the translational delay of many mRNA species which must then be translated at precise times throughout their final development. Spermatogenesis ends with the process of spermiation. This involves removal of the spermatid’s large cytoplasm, revealing the streamlined mature spermatozoa, and the final disengagement of sperm from the Sertoli cells into the tubule lumen, prior to their passage to the epididymis. Both the survival of spermatids during spermiogenesis and their release at the end of spermiation is dependent on optimal levels of androgen and FSH.

 

Spermatogenesis is a long process, taking up to 64 days in the human, and its inherent complexity demands precise timing and spatial organization. Within the seminiferous tubules, Sertoli cells and surrounding germ cells in various phases of development are highly organized into a series of cell associations, known as stages. These stages result from the fact that a particular spermatogonial cell type, when it appears in the epithelium, is always associated with a specific stage of meiosis and spermatid development. The stages follow one another along the length of the seminiferous tubule, and the completion of a series of stages is termed a “cycle”. This cycle along the length of tubule is obvious in rodents, however in humans several cycles are intertwined in a helical pattern; thus a human seminiferous tubule viewed in cross section will contain up to three stages. The completion of one cycle results in the release of mature spermatozoa into the tubule lumen; the cycles are repeated along the tubules, resulting in constant “pulses” of sperm production. These pulses of sperm release allow the testes to continually produce millions of sperm, with the average normospermic man able to produce approximately 1000 sperm per heartbeat.

 

The precise timing and co-ordination of spermatogenesis is achieved by many factors. Emerging evidence suggests that retinoic acid, metabolized within the testis from circulating retinol (a product of vitamin A) is a major driver of spermatogenesis. A precise pulse of retinoic acid action is delivered to a particular stage of the spermatogenic cycle; this pulse is achieved by the constrained expression of enzymes involved in retinoic acid synthesis, degradation and storage, as well as the local expression of retinoic acid receptors. This pulse of retinoic acid acts directly on spermatogonia to stimulate their entry into the pathway committed to meiosis. It also acts directly on Sertoli cells to regulate its cyclic functions. Sertoli cells contain an internal “clock” that allows them to express genes and proteins at precise times. This clock appears to be set by retinoic acid, however the timing of the clock can be influenced by the germ cells themselves.

 

The timing of spermatogenesis also relies on an extraordinarily complex program of gene transcription and protein translation. Alternative splicing of mRNA is highly prevalent in the testis, and generates many germ cell-specific transcripts that are important for the ordered procession of germ cell development. Noncoding RNAs, including microRNAs, small interfering RNAs, piRNAs and long noncoding RNAs, are highly expressed in the testis, particularly by the germ cells. Indeed, studies on male germ cells have revealed much of what is known about the biology and function of non-coding RNAs. These non-coding RNAs have many and varied roles and are particularly required for the transcriptional program executed during meiosis and spermiogenesis.

 

The male germ cell transmits both genetic and epigenetic information to the offspring. Epigenetic modifications of the genome are heritable; epigenetic processes such as DNA methylation and histone modifications regulate chromatin structure and modulate gene transcription and silencing. The male germ cell undergoes major epigenetic programming in the fetal testis, during the genome wide de-methylation and re-methylation to establish the germline-specific epigenetic pattern that is eventually transmitted to the offspring. The sperm epigenome is then further remodelled during postnatal spermatogenesis by various mechanisms. It is now known that a man’s sperm epigenome can be altered by environmental factors (including diet and lifestyle as well as exposure to environmental factors) throughout his lifetime and this altered sperm epigenome can influence both his fertility and the health of his future children.

 

It is clear from the above summary that spermatogenesis relies on many intrinsic and extrinsic factors. However spermatogenesis is absolutely dependent on androgen-secretion by the Leydig cells; androgens stimulate and maintain germ cell development throughout life. Testicular testosterone levels are very high, by virtue of its local production, however they are considerably higher than those required for the initiation and maintenance of spermatogenesis. Androgen action on receptors within Leydig cells, peritubular myoid cells and Sertoli cells is essential for normal steroidogenesis and spermatogenesis. While testosterone is essential for spermatogenesis, it is also important to note that exogenous testosterone administration resulting in even slightly supraphysiological serum levels suppresses gonadotropin secretion via negative feedback effects on the hypothalamus and pituitary, leading to the cessation of sperm production.

 

In contrast to androgens, spermatogenesis can proceed in the absence of FSH; however, testes are smaller and sperm output is reduced. This is due to FSH’s role in the peri-pubertal proliferation and differentiation of Sertoli cells and in the maintenance of germ cell survival. While FSH is thus not essential for spermatogenesis, it is generally considered that optimal spermatogenesis requires the combined actions of both androgen and FSH, with both hormones having independent, co-operative and synergistic effects to promote maximal sperm output.

 

These factors are an important consideration in the stimulation of spermatogenesis in the setting of HH. As androgens are essential for the initiation of sperm production, the induction of spermatogenesis in HH acquired after puberty is achieved by the administration of hCG (as an LH substitute). Prolonged therapy is required to produce sperm in the ejaculate, given that human spermatogenesis takes more than 2 months to produce sperm from spermatogonia. Treatment with hCG alone may be sufficient for the induction of spermatogenesis in men with larger testes due to potential residual FSH action, however, for many men, and particularly for those with congenital HH, the co-administration of FSH is needed for maximal stimulation of sperm output. In men with congenital HH, FSH is needed to induce Sertoli cell maturation, whereas men with acquired HH and smaller testes benefit from the co-administration of FSH due to the synergistic actions of FSH and androgens on spermatogenesis.

 

In summary, the testes, under the influence of gonadotropins, produce testosterone and sperm. These processes require the co-ordinated actions of multiple cell types and the secretion of paracrine factors. Spermatogenesis is a long and complex process that relies on multiple somatic cells as well as on the co-ordinated expression of genes, proteins and non-coding RNAs. Inherent vulnerabilities exist in spermatogenesis meaning that lifestyle and environmental factors can potentially influence a man’s sperm epigenome, his fertility and the health of his future children.

 

 

GENERAL ANATOMY OF THE MALE REPRODUCTIVE SYSTEM

The Testis

The testis lies within the scrotum and is covered on all surfaces, except its posterior border, by a serous membrane called the tunica vaginalis. This structure forms a closed cavity representing the remnants of the processus vaginalis into which the testis descends during fetal development (Figure 1). Along its posterior border, the testis is loosely linked to the epididymis which at its lower pole gives rise to the vas deferens (1).

Figure 1. The relationships of the tunica vaginalis to the testis and epididymis is illustrated from the lateral view and two cross sections at the level of the head and mid-body of the epididymis. The large arrows indicate the sinus of the epididymis posteriorly. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

Figure 1. The relationships of the tunica vaginalis to the testis and epididymis is illustrated from the lateral view and two cross sections at the level of the head and mid-body of the epididymis. The large arrows indicate the sinus of the epididymis posteriorly. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

The testis is covered by a thick fibrous connective tissue capsule called the tunica albuginea. From this structure, thin imperfect septa run in a posterior direction to join a fibrous thickening of the posterior part of the tunica albuginea called the mediastinum of the testis. The testis is thus incompletely divided into a series of lobules.

Within these lobules, the seminiferous tubules form loops, the terminal ends of which extend as straight tubular extensions, called tubuli recti, which pass into the mediastinum of the testis and join an anastomosing network of tubules called the rete testis. From the rete testis, in the human, a series of six to twelve fine efferent ducts join to form the duct of the epididymis. This duct, approximately 5-6m long in the human, is extensively coiled and forms the structure of the epididymis that can be divided into the head, body and tail of the epididymis (1). At its distal pole, the tail of the epididymis gives rise to the vas deferens (Figure 2).

Figure 2. The arrangement of the efferent ducts and the subdivisions of the epididymis and vas are shown. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

Figure 2. The arrangement of the efferent ducts and the subdivisions of the epididymis and vas are shown. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

The arterial supply to the testis arises at the level of the second lumbar vertebra from the aorta on the right and the renal artery on the left and these vessels descend retroperitoneally to descend through the inguinal canal forming part of the spermatic cord. The testicular artery enters the testis on its posterior surface, sending a network of branches that run deep to the tunica albuginea before entering the substance of the testis (2). The venous drainage passes posteriorly and emerges at the upper pole of the testis as a plexus of veins termed the pampiniform plexus (Figure 3). As these veins ascend they surround the testicular artery, forming the basis of a countercurrent heat exchange system which assists in the maintenance of a temperature differential between the scrotally placed testis and the intra-abdominal temperature (3).

Figure 3. The arrangement of the vasculature of the testis in the region of the distal spermatic cord and testis is shown. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

Figure 3. The arrangement of the vasculature of the testis in the region of the distal spermatic cord and testis is shown. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

The Distal Reproductive Tract

The vas deferens ascends from the testis on its posterior surface as a component of the spermatic cord passing through the inguinal canal and descends on the posterolateral wall of the pelvis to reach the posterior aspect of the bladder where its distal end is dilated forming the ampulla of the vas (Figure 4). At this site it is joined by the duct of the seminal vesicle, on each side, to form an ejaculatory duct that passes through the substance of the prostate to enter the prostatic urethra. The seminal vesicles and the prostate, the latter of which opens by a series of small ducts into the prostatic urethra, contribute approximately 90-95% of the volume of the ejaculate. During the process of ejaculation, these contents, together with sperm transported through the vas, are discharged through the prostatic and penile urethra. Retrograde ejaculation is prevented by contraction of the internal sphincter of the bladder during ejaculation. Failure of this sphincter to contract results in retrograde ejaculation and a low semen volume.

Figure 4. The diagram depicts the relationship between the vas deferens, the seminal vesicles, the posterior aspect of the bladder and the prostate gland. The cytological features of the epithelium of the seminal vesicles is shown: this tissue is androgen dependent. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

Figure 4. The diagram depicts the relationship between the vas deferens, the seminal vesicles, the posterior aspect of the bladder and the prostate gland. The cytological features of the epithelium of the seminal vesicles is shown: this tissue is androgen dependent. Reproduced with permission from de Kretser et.al.1982 in 'Disturbances in Male Fertility' Eds K Bandhauer and J Frick, Springer - Verlag Berlin.

AN OVERVIEW OF SPERMATOGENESIS

Spermatogenesis is the process by which precursor germ cells termed spermatogonia undergo a complex series of divisions to give rise to spermatozoa (4-5). This process takes place within the seminiferous epithelium (Figure 5), a complex structure composed of germ cells and radially-oriented supporting somatic cells called Sertoli cells. The latter cells extend from the basement membrane of the seminiferous tubules to reach the lumen. The cytoplasmic profiles of the Sertoli cells are extremely complex as this cell extends a series of processes that surround the adjacent germ cells in an arboreal pattern (5-7).

Figure 5. The top panel illustrates the typical structure of the human seminiferous epithelium containing the germ cells and Sertoli cells. The position of Sertoli cell nuclei within the epithelium is indicated, as is the tubule lumen. The tubules are surrounded by thin plate-like contractile cells called peritubular myoid cells. The Leydig cells and blood vessels lie within the interstitium. The bottom panel illustrates the nuclear morphology of the major cell types found within the human seminiferous epithelium, showing the progress of spermatogenesis from immature spermatogonia through meiosis and spermiogenesis to produce mature elongated spermatids. Abbreviations: Ad: A dark spermatogonia, Ap: A pale spermatogonia, B: type B spermatogonia, Pl: preleptotene spermatocyte, L-Z: leptotene to zygotene spermatocyte, PS: pachytene spermatocyte, M: meiotic division, rST: round spermatid, elST: elongating spermatid, eST: elongated spermatid. All germ cell micrographs were taken at the same magnification to indicate relative size. Micrograph of seminiferous epithelium was provided by Dr Sarah Meachem.

Figure 5. The top panel illustrates the typical structure of the human seminiferous epithelium containing the germ cells and Sertoli cells. The position of Sertoli cell nuclei within the epithelium is indicated, as is the tubule lumen. The tubules are surrounded by thin plate-like contractile cells called peritubular myoid cells. The Leydig cells and blood vessels lie within the interstitium. The bottom panel illustrates the nuclear morphology of the major cell types found within the human seminiferous epithelium, showing the progress of spermatogenesis from immature spermatogonia through meiosis and spermiogenesis to produce mature elongated spermatids. Abbreviations: Ad: A dark spermatogonia, Ap: A pale spermatogonia, B: type B spermatogonia, Pl: preleptotene spermatocyte, L-Z: leptotene to zygotene spermatocyte, PS: pachytene spermatocyte, M: meiotic division, rST: round spermatid, elST: elongating spermatid, eST: elongated spermatid. All germ cell micrographs were taken at the same magnification to indicate relative size. Micrograph of seminiferous epithelium was provided by Dr Sarah Meachem.

Spermatogenesis can be divided into three major phases (i) proliferation and differentiation of spermatogonia, (ii) meiosis, and (iii) spermiogenesis which represents a complex metamorphosis of round haploid germ cells into the highly specialized structure of the spermatozoon (Figure 5). It is important to note that, as germ cells divide and differentiate through these phases, they do not separate completely after mitosis but remain joined by intercellular bridges (8). These intercellular bridges persist throughout all stages of spermatogenesis and are thought to facilitate biochemical interactions allowing synchrony of germ cell maturation.

Spermatogonial Renewal and Differentiation

Spermatogonia are precursor male germ cells that reside near the basement membrane of the seminiferous epithelium. Spermatogonial stem cells (SSC) divide to renew the stem cell population and to provide spermatogonia that are committed to the spermatogenic differentiation pathway. Adult mouse and human SSC are pluripotent, and have the ability to differentiate into derivatives of all three germ layers (9-10).

In general, two main types of spermatogonia, known as Type A and B, can identified in mammalian testes on the basis of nuclear morphology (5). Type A spermatogonia exhibit fine pale-staining nuclear chromatin and are considered to include the SSC pool, the undifferentiated spermatogonia (Aundiff) pool, and spermatogonia which have become committed to differentiation (Adiff). The Aundiff pool is comprised of the SSC, single A spermatogonia (As), and interconnected cysts of either 2 (known as A paired, or Apr) or more (aligned or Aal) undifferentiated spermatogonia that remain connected by intercellular bridges. Once per cycle (see section below), the Aundiff cells transform into Adiff cells, which are then designated A1, A2, etc. Adiff spermatogonia ultimately divide to produce type B spermatogonia. Type B spermatogonia show coarse chromatin collections close to the nuclear membrane (11) and represent the more differentiated spermatogonia that are committed to entry into meiosis (12).

Recent studies have focused on dissecting the molecular properties of the various A spermatogonial subtypes in an effort to identify the SSC population of the testis. Studies have also investigated their clonal behavior as they divide and differentiate. The pioneering technique of spermatogonial transplantation (13-16) is used to determine the regenerative capacity of a cell population and to define subtypes with SSC potential.

The current, widely-accepted model of Type A spermatogonial division and differentiation includes the concept of As representing the least differentiated spermatogonial population. Within this population, some As cells express the ID4 protein and have both regenerative and self-renewal properties, suggesting these are the true stem cells of the adult testis (17-18). As can divide completely to renew their population, or divide incompletely to produce Apr cells, which represents an initial step towards differentiation. The Apr cells subsequently divide to produce Aal cells which then divide to produce chains (or cysts) of more differentiated spermatogonia, termed Aal4-16. As the A spermatogonia subtypes progress through these steps, there are changes in their molecular signature and the expression of cell surface markers, likely reflecting their differentiation state and functional capabilities, see (19).

Recent in vivo imaging studies of fluorescently-tagged A spermatogonial subtypes challenge some aspects of the current model (20-21). These studies suggest that there may be more fluidity in the transition between undifferentiated A spermatogonial subtypes (i.e. As, Apr, Aal), and in their ability to attain SSC characteristics (20-21). In vivo imaging and pulse labeling studies suggest that fragmentation of spermatogonial cysts (e.g fragmentation of Apr or Aal clones) to produce As is a commonly observed phenomenon, and biophysical modeling studies suggest that fragmentation of Apr and Aal clones may be an important source of As that can then exhibit SSC behavior (20). Thus there may be a less linear relationship between As→Apr→Aal, and more flexibility as they fragment and transition back and forth between subtypes. Clone fragmentation appears likely to be an important aspect of steady state spermatogonial kinetics, as well as during the repopulation of the testis following an insult to spermatogenesis, such as via radiation or chemotherapeutic agents (20).

In humans and other primates, the Type A spermatogonia can only be classified into two subtypes; A dark (Ad) and A pale (Ap) spermatogonia (12). Some investigators have proposed that the Ad spermatogonia are similar to As in the rodent, and thus represent the SSC or reserve spermatogonial population (22-24) whereas others have suggested that the Ap spermatogonia are the true stem cell of the testis (25). More recent studies suggest that Ap spermatogonia also show characteristics of As spermatogonia in rodents, reviewed in (26), however it remains unclear how primate Type A spermatogonial subtypes relate to those in rodents. In primates, both Ap and Ad spermatogonia express GFRα (27), a marker of Aundiff in rodents, reviewed in (19). Like rodent Aundiff spermatogonia, there are heterogeneous subpopulations within GFRα1+ human Ap spermatogonia (28). Differentiation of A spermatogonia in monkeys is associated with the cytoplasmic to nuclear translocation of the transcription factor SHLH1 (27). Further studies on markers of rodent spermatogonial subtypes, including SSC, and their analysis in primate and human testes will inform our understanding of human spermatogonial biology (26).

 

Meiosis

Meiosis is the process by which gametes undergo reductive division to provide a haploid spermatid, and in which genetic diversity of the gamete is assured via the exchange of genetic material. During meiosis I, DNA synthesis is initiated, resulting in a tetraploid gamete. The exchange of genetic information is achieved during meiotic recombination, which involves the induction of DNA double-strand breaks (DSBs) during pairing of homologous chromosomes and the subsequent repair of DSBs using homologous chromosomes as templates. Once exchange of genetic material is complete, the cells proceed through two successive reductive divisions to yield haploid spermatids. This process is governed by genetically programmed checkpoint systems.

Meiosis commences when Type B spermatogonia lose their contact with the basement membrane and form preleptotene primary spermatocytes. The preleptotene primary spermatocytes commence DNA synthesis and the condensation of individual chromosomes begins, resulting in the appearance of thin filaments in the nucleus which identify the leptotene stage (29). At this stage, each chromosome consists of a pair of chromatids (Figure 6). As the cells move into the zygotene stage, there is further thickening of these chromatids and the pairing of homologous chromosomes. The further enlargement of the nucleus and condensation of the pairs of homologous chromosomes, termed bivalents, provides the nuclear characteristics of the pachytene stage primary spermatocyte. During this stage, there is exchange of genetic material between homologous chromosomes derived from maternal and paternal sources, thus ensuring genetic diversity of the gametes. The sites of exchange of genetic material are marked by the appearance of chiasmata and these become visible when the homologous chromosomes separate slightly during diplotene. The exchange of genetic material involves DNA strand breakage and subsequent repair (30).

Figure 6. The diagrammatic representation of the events occurring between homologous chromosomes during the prophase of the first meiotic division shows the period of DNA synthesis, the formation of the synaptonemal complex and the processes involved in recombination. Reproduced with permission from de Kretser and Kerr (1994) in "The Physiology of Reproduction" Ed E Knobil & J D Neill, Lippincott, Williams & Wilkins.

Figure 6. The diagrammatic representation of the events occurring between homologous chromosomes during the prophase of the first meiotic division shows the period of DNA synthesis, the formation of the synaptonemal complex and the processes involved in recombination. Reproduced with permission from de Kretser and Kerr (1994) in "The Physiology of Reproduction" Ed E Knobil & J D Neill, Lippincott, Williams & Wilkins.

The diplotene stage is recognized by partial separation of the homologous pairs of chromosomes that still remain joined at their chiasmata and each is still composed of a pair of chromatids. With dissolution of the nuclear membrane, the chromosomes align on a spindle and each member of the homologous pair moves to opposite poles of the spindle during anaphase. The resultant daughter cells are called secondary spermatocytes and contain the haploid number of chromosomes but, since each chromosome is composed of a pair of chromatids, the DNA content is still diploid. After a short interphase, which in the human represents approximately six hours, the secondary spermatocytes commence a second meiotic division during which the chromatids of each chromosome move to opposite poles of the spindle forming daughter cells that are known as round spermatids (12, 31). Meiotic maturation in the human takes about 24 days to proceed from the preleptotene stage to the formation of round spermatids.

It is well known that advancing maternal age is associated with increased meiotic errors leading to reduced gamete quality, however whether this phenomenon occurs in males has been the subject of debate. A recent study in mice showed that advanced age was associated with increased defects in chromosome pairing, however no increase in anueploidy was observed at Metaphase II, suggesting that such errors were corrected during metaphase checkpoints in males (32). Therefore advanced age, at least in mice, has more of an impact on gamete aneuploidy in females compared to males.

Spermiogenesis and Spermiation

The transformation of a round spermatid into a spermatozoon represents a complex sequence of events that constitute the process of spermiogenesis. No cell division occurs, but a conventional round cell becomes converted into a spermatozoon with the capacity for motility. The basic steps in this process (Figure 7) are consistent between all species and consist of (a) the formation of the acrosome (b) nuclear changes (c) the development of the flagellum or sperm tail (d) the reorganisation of the cytoplasm and cell organelles and (e) the process of release from the Sertoli cell termed spermiation (5, 33-37).

Figure 7. The changes during spermiogenesis involving the transformation of a round spermatid to a mature spermatozoon are shown. Redrawn with permission from de Kretser and Kerr (1994) in "The Physiology of Reproduction" Ed E Knobil & J D Neill, Lippincott, Williams & Wilkins.

Figure 7. The changes during spermiogenesis involving the transformation of a round spermatid to a mature spermatozoon are shown. Redrawn with permission from de Kretser and Kerr (1994) in "The Physiology of Reproduction" Ed E Knobil & J D Neill, Lippincott, Williams & Wilkins.

The formation of the acrosome commences by the coalescence of a series of granules from the Golgi complex. These migrate to come into contact with the nuclear membrane where they form a cap-like structure which becomes applied over approximately 30-50% of the nuclear surface (33). Acrosome biogenesis begins early in round spermatid development, and progressively extends as a “cap” over the nucleus as round spermatids differentiate further.

Once the acrosome is fully extended, round spermatids begin what is known as the elongation phase of spermiogenesis. As spermatid elongation commences, the nucleus polarizes to one side of the cell (Figure 7) and comes into close apposition with the cell membrane in a region where it is covered by the acrosomal cap. Soon after this polarization, the spermatid’s chromatin starts to visibly condense, forming progressively larger and more electron dense granules together with a change in the shape of the condensed nucleus. This change in nuclear shape varies significantly between species. The condensation of chromatin is achieved by the replacement of lysine-rich histones with transitional proteins which in turn are subsequently replaced by arginine-rich protamines (38-39). The spermatid chromatin subsequently becomes highly stabilized and resistant to digestion by the enzyme DNAse. Associated with these changes is a marked decrease in nuclear volume and, importantly, the cessation of gene transcription (40). Therefore, the subsequent spermatid elongation phase proceeds in the absence of active gene transcription (see (36)).

At the commencement of spermatid elongation, a complex, microtubule-based structure known as the manchette is formed. The microtubule network emanates from a perinuclear ring at the base of the acrosome and extends outwards into the cytoplasm. The manchette is closely opposed to the nuclear membrane, and is thought to participate in nuclear head shaping, perhaps by exerting a force on the nucleus as it progressively moves distally towards the posterior portion of the nucleus (41-43).

The formation of the tail commences early in spermiogenesis in the round spermatid phase, when a filamentous structure emerges from one of the pair of centrioles which lie close to the Golgi complex. Associated with the changing nuclear-cytoplasmic relationships, the developing flagellum and the pair of centrioles become lodged in a fossa in the nucleus at the opposite pole to the acrosome. The central core of the flagella’s axial filament, called the axoneme, consists of nine doublet microtubules surrounding two single central microtubules, which represents a common pattern found in cilia. This basic structure is modified at the region of its articulation with the nucleus through the formation of a complex structure known as the connecting piece (44).

Metamorphosis of the flagella proceeds during the elongation phase, as it acquires its characteristic neck region, mid-, principal- and end-pieces (37). The development of the flagella is thought to involve a mechanism known as Intra-Manchette Transport (IMT), which is proposed to be similar to the Intra-Flagellar Transport (IFT) systems used in other ciliated cells. IMT involves proteins being “shuttled” from the spermatid nucleus down to the developing flagellum via molecular motors travelling along “tracks” of microtubules and filamentous actin (42-43, 45).

The middle and principal pieces contain the mitochondrial and fibrous sheath components, respectively, and include the outer dense fibers. The biochemical characteristics of these components of the sperm tail are emerging (46-51), reviewed in (37). While these components provide some structural stability to the tail, evidence suggests that they may serve as a molecular scaffold to position key enzymes critical to successful sperm motility. For instance, CatSper 1, an ion channel plasma membrane-associated protein present in the principal-piece, has been shown to regulate calcium ion fluxes critical for the process of hyperactivation of sperm motility associated with capacitation (52). Studies demonstrate that CatSper, or a directly associated protein, is a non-genomic progesterone receptor that mediates the effects of progesterone on sperm hyperactivation and acrosome reaction (53-54). Further studies have shown that plasma membrane calcium-ATPase 1 is also located in the principal-piece and has been shown to be critical for the process of hyperactivation of sperm motility (55). While these are plasma membrane-located complexes, TPX1 (also called CRISP2), a protein localized to the outer dense fibers of the tail and acrosome (56) has been shown to regulate ryanodine receptor calcium signalling (57).

The formation of the mitochondrial sheath occurs at the time of the final reorganization of the cytoplasm and organelles of the spermatid (5, 33, 58). The mitochondria that had remained around the periphery of the spermatid aggregate around the proximal part of the flagellum to form a complex helical structure (Figure 8).

The mature elongated spermatids undergo a further complex remodeling during spermiation, the process by which the mature spermatids are remodeled and then released from the Sertoli cells prior to their passage to the epididymis, see (35) for review. This remodeling includes the removal of specialized adhesion junctions that have ensured tight adhesion of the spermatid to the Sertoli cell during its elongation process, further remodeling of the spermatid head and acrosome and removal of the extensive cytoplasm to produce the streamlined spermatozoon. The cytoplasm of the spermatid migrates to a caudal position around the tail and is markedly reduced in volume. Some observations suggest that prolongations of Sertoli cell cytoplasm send finger-like projections which invaginate the cell membrane of the spermatid cytoplasm and literally 'pull' the residual cytoplasm off the spermatid (33). The remnants of the spermatid cytoplasm form what is termed the residual body. The residual bodies contain mitochondria, lipid and ribosomal particles, and are phagocytosed and moved to the base of the Sertoli cell where they are broken down by lysosomal mechanisms. The final release of sperm at the end of spermiation is an instantaneous event, and likely involves phosphorylation-dependent signaling cascades within the Sertoli cell resulting in changes in the adhesive nature of cell adhesion molecules (35), culminating in the Sertoli cell “letting go” of the mature spermatid (59). The morphological features of spermiation are relatively conserved between species, particularly among mammals (60). Spermiation is highly susceptible to perturbation by pharmacological modulators and by agents that suppress gonadotropins, reviewed in (35), and failure of spermiation can be recognized by the presence of mature elongated spermatid nuclei being phagocytosed by the Sertoli cells (61).

Figure 8. A cross-section through the developing mid-piece of the sperm tail shows the aggregation of mitochondria (arrows) surrounding the outer dense fibres (labelled 1-9) which in turn surround the axoneme composed of 9 doublet microtubules surrounding two central microtubules. Reproduced with permission from "Visual atlas of human sperm structure and function for assisted reproductive technology" Ed A.H. Sathanathan 1996.

Figure 8. A cross-section through the developing mid-piece of the sperm tail shows the aggregation of mitochondria (arrows) surrounding the outer dense fibres (labelled 1-9) which in turn surround the axoneme composed of 9 doublet microtubules surrounding two central microtubules. Reproduced with permission from "Visual atlas of human sperm structure and function for assisted reproductive technology" Ed A.H. Sathanathan 1996.

The Cycle of the Seminiferous Epithelium

Within the seminiferous epithelium, the cell types that constitute the process of spermatogenesis are highly organized to form a series of cell associations or stages. These cell associations, or stages of spermatogenesis, result from the fact that a particular spermatogonial cell type, when it appears in the epithelium, is always associated with a specific stage of meiosis and spermatid development. The stages follow one another along the length of the seminiferous tubule, and the completion of a series of stages is termed a “cycle” (see Figure 9). This cycle along the length of tubule is obvious in rodents, however in humans the situation is more complex (see below). The completion of one cycle results in the release of mature spermatozoa into the tubule lumen; the cycles are repeated along the length of the tubules (Figure 9), resulting in constant “pulses” of sperm production along the tubules. Thus the cyclic nature of spermatogenesis enables continual sperm production within the testis. These pulses of sperm release along the length of the seminiferous tubules allow the testes to continually produce millions of sperm, with the average normospermic man able to produce approximately 1000 sperm per heartbeat.

The cycle of the seminiferous epithelium was defined by LeBlond and Clermont (62), as the series of changes in a given area of the seminiferous tubule between two appearances of the same developmental stage or cell association. They defined 14 stages in the rat cycle based on the 19 phases of spermiogenesis (Figure 9) as identified by the periodic acid Schiff (PAS) stain. In effect, if it was possible to observe the same region of the seminiferous epithelium by phase contrast microscopy over time, the appearance would progress through the 14 stages before stage I reappeared. They also demonstrated that the duration of any one stage was proportional to the frequency with which it was observed in the testis. As type A spermatogonia in any one area of the epithelium progress through meiosis and spermiogenesis to become spermatozoa, the specific area of the tubule would pass through the 14 stages four times. In each progression, the progeny of the spermatogonia progressively move toward the lumen of the tubule.

Figure 9. The top panel shows a diagrammatic representation of the stages of the seminiferous cycle in the rat and shows the types of germ cell associations which form the stages. The stage is denoted by roman numerals. These stages follow one another in a cyclic manner along the length of the seminiferous tubule, as illustrated in the diagram in the middle panel. Examples of the histology of the seminiferous epithelium at two different stages are given in the bottom panel.

Figure 9. The top panel shows a diagrammatic representation of the stages of the seminiferous cycle in the rat and shows the types of germ cell associations which form the stages. The stage is denoted by roman numerals. These stages follow one another in a cyclic manner along the length of the seminiferous tubule, as illustrated in the diagram in the middle panel. Examples of the histology of the seminiferous epithelium at two different stages are given in the bottom panel.

Studies in many mammalian species demonstrated that the cycle of spermatogenesis could be identified for each species but showed that the duration of the cycle varied for each species (12). In many species, especially the rat, the same stage of spermatogenesis extends over several millimetres of the adjacent tubule and it is possible, by observation under transillumination, to dissect lengths of seminiferous tubules at the same phase of spermatogenesis (63). Such observations amply confirmed the earlier studies of Perey and colleagues (64), that the stages of spermatogenesis were sequentially arranged along the length of the tubule (Figure 9). As the cycle progress, this arrangement resulted in a "wave of spermatogenesis" along the tubule. Regaud (65) interpreted his observations correctly by the statement "the wave is in space what the cycle is in time".

For many years, investigators believed that such a cycle did not occur in the human testis but the careful studies of Clermont (66) showed that human spermatogenesis could be subdivided into 6 stages. However unlike the rat, each stage often only occupied one quadrant of any given tubule cross section giving the disorganized appearance. By careful studies using tritiated thymidine injections into the testis, Clermont and Heller (31) demonstrated that the duration of the cycle in the human took 16 days and the progression from spermatogonia to sperm took 70 days or four and a half cycles of the seminiferous cycle. Other studies showed that the cycle length was specific for each species (eg rat 49 days) and the progression of each cell type in spermatogenesis involved a defined duration (12). It is likely that the relatively poor definition of stages in human seminiferous tubules, compared to the rat, is due to a greater number of spermatogonia entering each phase of the cycle in the rat, their cell progeny therefore occupying a greater length of the tubule.

Transcriptional profiling studies described the changing patterns of gene expression across the rat spermatogenic cycle, and demonstrated that Sertoli cells and germ cells showed highly co-ordinated stage-dependent changes in gene expression (67). The mechanisms underlying these temporal constraints on spermatogenesis have been the subject of speculation as to whether these were intrinsic or were imposed by the Sertoli cells. The latter proposition is supported by the demonstration that when rat germ cells were transplanted into the mouse testis, spermatogenesis proceeded at the normal rate for the rat, indicating that the kinetics of the spermatogenic cycle are determined by intrinsic mechanisms within germ cells (68). In contrast however, Sertoli cells demonstrate cyclic expression of certain proteins in the embryonic and pre-pubertal period, even in the absence of germ cells (69). Recent studies demonstrate that retinoic acid “sets the clock” within post-pubertal Sertoli cells, however differentiating germ cells are required to “fine tune” the clock (70) (see below for further information). Taken together, these observations demonstrate that the Sertoli cell contains a “clock” that modulates cyclic gene and protein expression, and that the precise timing of this clock is modulated by germ cells.

THE ROLE OF SERTOLI CELLS IN SPERMATOGENESIS

Sertoli cells have an intimate physical relationship with the germ cells (Figure 10) during the process of spermatogenesis (5, 7, 71). The cytoplasmic extensions that pass between the germ cell populations surrounding the Sertoli cell provides structural support through a microfilament and microtubular network present in the cytoplasm of the Sertoli cell (72). This architecture is not static but changes in the tubule depending on the stage of the spermatogenic process.

Sertoli cells regulate the internal environment of the seminiferous tubule. This regulation is facilitated by specialized inter-Sertoli cell occluding-type junctions which are formed at the sites where processes of Sertoli cell cytoplasm from adjacent cells meet (73). These junctions contribute to the blood-testis barrier that regulates the entry of a variety of substances into the seminiferous tubule (74). These occluding junctions towards the base of Sertoli cells prevent the diffusion of substances from the interstitium into the inner part of the seminiferous tubule (see Figure 11). Because of the location of the junctions, spermatogonia have free access to substances from the interstitium (including the vasculature), however the germ cells “above” this junction, including meiotic and post-meiotic germ cells, have their access to factors from the interstitium restricted by the blood-testis-barrier. This effectively divides the seminiferous epithelium into a basal compartment containing spermatogonia, and an adluminal compartment containing meiotic and post-meiotic germ cells. As preleptotene spermatocytes migrate from the basement membrane of the tubule into the adluminal compartment, these tight junctions open up to allow this cellular migration to take place (Figure 11) and reform beneath the preleptotene spermatocytes which have now left the basement membrane to form leptotene spermatocytes. The formation and dissolution of these junctional specializations are under the control of numerous physiological regulators including endocrine (75-76) and paracrine (77) factors, see for (78) recent review.

The Sertoli cell junctions and the blood-testis barrier are required for fertility (79). These junctions allow the environment of meiotic and post-meiotic germ cells to be precisely controlled by the Sertoli cell, enabling the precisely timed delivery of factors uniquely required for germ cell development. For example, the Sertoli cell provides substrates for germ cell glycolysis (80-82); lactate rather than glucose is the preferred substrate for glycolysis in primary spermatocytes and Sertoli cells generate lactate from glucose.

The blood-testis barrier has long been thought to contribute to the immune-privileged environment within the seminiferous epithelium. Meiotic and post-meiotic germ cells develop after the establishment of immune tolerance, and could thus be recognized as “foreign” by the immune system, therefore this barrier protects the developing germ cells from immune cell attack (83). However some studies show that seminiferous tubules continue to exclude immune cells when Sertoli cell junctions are absent (79) or even when Sertoli cells are ablated (84), raising questions as to the precise role of these junctions in immune privilege. It seems likely that many factors, including the production of anti-inflammatory cytokines, regulate the immune privileged environment of the testis.

In the adult rat testis, activin A protein peaks at the time of blood-testis-barrier remodeling and migration of leptotene spermatocytes into the adluminal compartment, suggesting that activin A could regulate blood-testis barrier function (for review see (85)). More recently it has been shown that elevated activin A action in vivo and in vitro suppresses the Sertoli cell tight junctions that form a major component of the blood-testis barrier (86), suggesting that activin A could facilitate blood-testis-barrier remodeling.

Recent studies have revealed that the blood-testis-barrier shows differential permeability and can exclude different sized molecules depending on its functional status (87). Tracer studies showed that the barrier can exclude all molecules between 0.6-150kDa in size when it is “fully sealed”, however in some situations and stages it can exclude large (150kDa+ molecules) but remain permeable to smaller molecules. These studies reveal that the barrier is more selective in its function than previously thought, and highlight the complexity of this structure and its important role in spermatogenesis.

Sertoli cells are indispensible for germ cell development, as they provide physical, metabolic and nutritional support at precisely timed intervals as dictated by the spermatogenic process. Transgenic mouse models have revealed many Sertoli cell genes that are required for all aspects of spermatogenesis, reviewed in (88). For example, the Etv5 transcription factor within Sertoli cells is essential for the maintenance of the stem cell niche (89), reviewed in (90). Sertoli cells respond to the changing needs of the developing germ cells as evidenced by the remarkable stage-specificity in the expression patterns of many Sertoli cell genes (67).

The differentiation status of Sertoli cells is related to their capacity to support spermatogenesis. For example, perinatal hypothyroidism extends the duration of Sertoli cell proliferation but also delays their maturation; this is also associated with a delay in the onset of spermatogenesis (91-92). It was widely believed that once Sertoli cells ceased pre-pubertal proliferation, they attained a so-called “terminally differentiated” phenotype. However it is now clear that Sertoli cells can de-differentiate in certain conditions of impaired spermatogenesis, reviewed in (93). For example, a loss of claudin 11 (a protein involved in Sertoli cell occluding junctions) causes Sertoli cells to remain proliferative during development and to lose their epithelial phenotype (94). De-differentiated Sertoli cells in cell cycle are not observed in normospermic men, but are present in men after 12 weeks of gonadotropin suppression (95). Intriguingly, adult Sertoli cells can even trans-differentiate into granulosa cells in the absence of the Sertoli cell transcription factor Dmrt1; this activates Foxl2-mediated female somatic cell programming (96). Therefore the maintenance of an adult Sertoli cell phenotype is essential for normal spermatogenesis.

While it has long been known that a healthy Sertoli cell is required for germ cell development, it is now clear that Sertoli cells support the development and function of other testicular cells. Recent studies using a mouse model of acute and specific ablation of Sertoli cells have revealed they are essential for the maintenance of peritubular myoid cell fate and function, and are required for Leydig cell development and normal steroidogenesis (84, 97). Therefore Sertoli cells are required for both sperm and androgen production within the testis.

Figure 10. The general architecture of the Sertoli cell is shown. Note the thin cytoplasmic processes that extend between the germ cells. The Sertoli cell is in contact with a variety of germ cells and adjacent Sertoli cells when three dimensional perspectives are considered.

Figure 10. The general architecture of the Sertoli cell is shown. Note the thin cytoplasmic processes that extend between the germ cells. The Sertoli cell is in contact with a variety of germ cells and adjacent Sertoli cells when three dimensional perspectives are considered.

Figure 11. The position of the blood testis barrier in the seminiferous epithelium, which is formed by tight, occluding and adhesion junctions between adjacent Sertoli cells. This barrier restricts the diffusion of substances from the interstitum and blood vessels, and thus allows the Sertoli cell to determine the microenvironment above the junctions. This barrier effectively divides the seminiferous epithelium into two compartments, the basal compartment with free access to substances from outside the tubule, and the adluminal compartment, the environment of which is controlled by the Sertoli cell. Meiosis and the differentiation of spermatids occurs in the adluminal compartment. The inter-Sertoli cell junctions transiently remodel to allow germ cells to move from the basal to the adluminal compartments, whilst protecting the functionality of the barrier. Diagram provided by Jenna Haverfield.

Figure 11. The position of the blood testis barrier in the seminiferous epithelium, which is formed by tight, occluding and adhesion junctions between adjacent Sertoli cells. This barrier restricts the diffusion of substances from the interstitum and blood vessels, and thus allows the Sertoli cell to determine the microenvironment above the junctions. This barrier effectively divides the seminiferous epithelium into two compartments, the basal compartment with free access to substances from outside the tubule, and the adluminal compartment, the environment of which is controlled by the Sertoli cell. Meiosis and the differentiation of spermatids occurs in the adluminal compartment. The inter-Sertoli cell junctions transiently remodel to allow germ cells to move from the basal to the adluminal compartments, whilst protecting the functionality of the barrier. Diagram provided by Jenna Haverfield.

The number of Sertoli cells determines the ultimate spermatogenic potential of the testis. In rodents, Sertoli cells proliferate in fetal and early postnatal life and even into adulthood, reviewed in (93), whereas in humans there are two waves of proliferation; during the fetal and early neonatal period when the population increases 5 fold, and again prior to puberty when the population increases more than two fold (98), reviewed in (93, 99). Studies in mice show that apoptosis of Sertoli cells during fetal life results in abnormal cord development, smaller testes and reduced seminiferous tubule size (100), suggesting the proliferation of Sertoli cells during the fetal period is an important driver of seminiferous tubule formation. That Sertoli cell number determines the total sperm output of the testis, reviewed in (93, 101), is emphasized by studies showing that perinatal induction of hypothyroidism extends the duration of Sertoli cell proliferation, which in turn leads to increased Sertoli cell numbers and increased sperm output of the adult testis (91, 102). Other Sertoli cell mitogens such as FSH and activin (103-104), together with thyroxine, can also exert significant changes in the number of Sertoli cells in the testis, depending on the temporal pattern of their secretion. The latter must occur before the cessation of Sertoli cell proliferation. In the rat, this occurs at about 20 days whereas in the human, Sertoli cells cease to divide during the pubertal process (98). It is possible that the failure of many men with hypogonadotropic hypogonadism to achieve normal testicular size and normal sperm counts, when treated by gonadotropic stimulation, may result from abnormal Sertoli cell proliferation during fetal and prepubertal life resulting in a decreased Sertoli cell complement (105).

LEYDIG CELLS AND STEROIDOGENESIS

The Leydig cells lie within the intertubular regions of the testis and are found adjacent to blood vessels and the seminiferous tubules (5, 106). They are the cell type responsible for testosterone production which is essential for the maintenance of spermatogenesis. There are very significant organizational differences in the intertubular tissue betweens species reflecting the number of Leydig cells and differing architecture involving blood vessels and lymphatic sinusoids (107). Additionally, fibroblasts, macrophages, lymphocytes and small numbers of mast cells are found in the intertubular regions of the testis (108-109), reviewed in (110-111).

In most species there are two populations of Leydig cells, fetal and adult (112-113), that differ in terms of morphology, androgen synthesis, and regulation by paracrine and autocrine factors, reviewed in (110, 114-115). The fetal population appears following gonadal sex differentiation (gestational weeks 7-8 in humans) and, under the stimulation of hCG, results in the production of testosterone during gestation (116). In the human, these cells decrease in number towards term and degenerate and are lost from the intertubular region at about twelve months of age (117), although recent lineage-tracing experiments have indicated that fetal Leydig cells persist in the postnatal rodent testis (118). The adult population of Leydig cells in the human results from LH stimulation commencing at the time of puberty. This generation arises by division and differentiation of mesenchymal precursors under the influence of LH (119). Evidence in humans also supports a third neonatal Leydig cell population that peaks at 2-4 months after birth although their function is poorly understood (120), for review see (110). Whether or not the various Leydig cell populations share a common stem cell precursor also remains unclear (111).

Much of the data investigating gene regulatory systems that control fetal and adult Leydig cell differentiation is derived from rodent models, and differences may exist in the human. For example, placental hCG action via the LH/hCG receptor is required for human fetal Leydig cell development but not for mouse fetal Leydig cells (121). However, both species have in common the two main factors that influence fetal Leydig cell differentiation; Desert hedgehog (Dhh) and Platelet-derived growth factor A (Pdgfa). Interestingly both of these factors are Sertoli cell-derived and act in a paracrine fashion via their respective receptors, Patched1 (Ptch1) and platelet-derived growth factor receptor A (Pdgfra), on fetal Leydig cells to stimulate differentiation and steroidogenesis ((122-124), also see (110) and references therein). Dhh and Pdgfra also play an important role in adult Leydig cell development (124-126). Targeted deletion of Sertoli cell Dhh in mice causes major reductions in fetal Leydig cell number and androgen synthesis and results in undescended testes and feminized external genitalia (124, 127). A similar phenotype, termed complete pure gonadal dysgenesis, is observed in 46,XY patients with mutations in the DHH gene (128). A number of other important regulatory genes are also recognized to influence fetal and adult Leydig cell differentiation [e.g. Wt1, (129), Nrg1 (130), Inhba (125), for review see (110).

Leydig cells have the capacity to synthesize cholesterol from acetate or to take up this substrate for steroidogenesis from lipoproteins (106, 131). Typical of any steroid secreting cell, the Leydig cell contains abundant smooth endoplasmic reticulum and mitochondria which have tubular cristae that are unique to steroidogenic cells. The enzymes required for steroidogenesis are located in the mitochondria and in endoplasmic reticulum requiring intracellular transport of substrates between these organelles to achieve successful androgen production.

Leydig cells also produce the peptide hormone, insulin-like factor 3 (INSL3), which is structurally related to the insulin, IGF1 and IGF2 family (132-133), for review see (134). Targeted disruption of the Insl3 gene in mice causes bilateral cryptorchidism due to failure of gubernaculum development during embryogenesis (133). In the adult testis, INSL3 acts via its receptor, RXFP2 (formerly known as LGR8) found both on meiotic and post-meiotic germ cells, and on Leydig cells themselves (135-136). In gubernacular tissue, RXFP2 expression is up-regulated by androgen and abolished by an androgen receptor antagonist, suggesting a link between INSL3 and androgen signaling pathways (137). INSL3 has an anti-apoptotic function in the germ cell compartment (136), and could form part of an autocrine feedback loop in Leydig cells (135) which respond in vitro by increasing cyclic AMP and testosterone (138). In the human testis, INSL3 is a constitutive biomarker of both Leydig cell differentiation status and cell number, otherwise known as Leydig cell ‘functional capacity’ (134). This functionality has been useful to follow pubertal onset and increasing testicular volume (139) or to evaluate treatment for hypogonadism (134, 140), but does not have predictive value for sperm retrieval in patients with Klinefelter’s syndrome (141).

Control of Testosterone Production

Testosterone is the major androgen secreted by the Leydig cells found in the inter-tubular spaces of the testis. These cells arise from mesenchymal precursors and studies in the rat have identified that these precursors express the platelet-derived growth factor-α but not 3β hydroxysteroid dehydrogenase (142). Further, they suggest that many of these precursors are situated in close proximity to the surface of the seminiferous tubules. A normal male produces approximately 7 mg testosterone daily but also produces lesser amounts of weaker androgens such as androstenedione and dihydroepiandrosterone. In addition to testosterone, through the actions of the enzyme 5α reductase, the more potent androgen dihydrotestosterone is produced by the testis in smaller amounts. The testis also contributes approximately 25% of the total daily production of 17β-estradiol through the local action of the enzyme aromatase which converts androgenic substrates to this estrogen (143) (also see Endotext, Endocrinology of Male Reproduction, Chapter 17, Estrogens and Male Reproduction (144)). The remainder of the circulating estradiol is produced by the adrenal and peripheral tissues through the actions of aromatase. The biosynthesis and regulation of testosterone production is covered extensively elsewhere in Endotext (Endotext, Endocrinology of Male Reproduction, Chapter 2, Androgen Physiology, Pharmacology and Abuse (145)).

It is important to recognise that LH enhances the transcription of genes that encode a range of enzymes in the steroidogenic pathway (for reviews see (111, 114)) and that continued LH stimulation results in Leydig cell hypertrophy and hyperplasia (119, 146-147). In the normal male, the episodic nature of LH stimulation is likely to avoid prolonged periods of Leydig cell refractoriness to LH stimulation (148). It is recognized that the testosterone secretory capacity of the human testis declines in ageing men (for review see (149)) and this has been shown to result from a reduction in the efficacy of the ageing testis to respond to intravenous pulses of LH (150). These researchers showed that the estimated down-regulation of the Leydig cell achieved by exogenous LH pulses was augmented in these healthy older men making them refractory to further pulses for a longer period. (138).

It is well accepted that the level of production of androgens and estrogens by the testis can regulate bone mass, with decreased production causing osteoporosis. More recently, the production of osteocalcin by bone has been shown to influence testicular function (151), reviewed in (152). Using co-cultures of osteoblasts with testicular tissue, osteocalcin acted via G-protein coupled receptors (Gprc6a) to stimulate testosterone production (153).

 

Control of Leydig Cell Function by Other Testicular Cell Types

As alluded to earlier, Leydig cell development and function is critically dependent on other testicular cell types including Sertoli-, germ-, macrophages and peritubular myoid (see below) cells. In particular, a significant body of evidence has accumulated from studies in rodents to suggest that the seminiferous tubules influence Leydig cell number, maturation and testosterone production (154-155) (156). This data emerges from various experimental approaches where changes in Leydig cell function have been demonstrated, including knockout or over-expression of the androgen receptor or other signaling genes in Sertoli cells (157-158), temporary disruption of spermatogenesis via antagonist or toxicant treatment (159) or heat-treatment (160-161), or acute ablation of Sertoli or germ cell types to study global changes in Leydig cell function ((84, 97) for review see (162)). Collectively, these data show that Sertoli cells support adult Leydig cell development and survival by recruiting and maintaining their progenitors, and by regulating steroidogenic function (158, 162). These conclusions are supported by observations from unilateral testicular damage, such as that induced by cryptorchidism or efferent duct ligation, wherein the Leydig cells from the testis with spermatogenic damage show an increased capacity for testosterone biosynthesis and a decrease in LH receptor number (163-164). In contrast, germ cells appear to have little direct impact on Leydig cell gene expression in adulthood (156, 159), although post-meiotic germ cells have major impacts on Sertoli cell gene expression (162).

While similar mechanisms are difficult to identify in the human, it is recognized that elevated LH and low to low-normal testosterone concentrations, indicative of compromised Leydig cell function, are found in 15-20% of men with severe seminiferous tubule failure. Further support for the concept that the state of spermatogenesis can affect the function of the Leydig cells in men has emerged from the studies of Andersson et al (165), who showed that lower testosterone and higher estradiol concentrations were present, and accompanied by higher LH levels in infertile men. They concluded that this may reflect an extension of testicular dysgenesis to affect steroidogenesis or alternatively may result from inter-compartment interactions in the testis. There is also increasing support for the concept that environmental factors such as the phthalates are able to influence Leydig cell function (166). In utero exposure of rats to di(n-butyl)phthalate during the masculinization programming window in fetal life has been shown to cause focal testicular dysgenesis as expressed by Leydig cell aggregation and malformed seminiferous tubules (166). These features were linked to impaired intra-testicular testosterone levels and a decreased ano-genital distance, an emerging marker of deficient androgen action in utero.

Compelling evidence exists to demonstrate that other interstitial cells can also impact Leydig cell function. In particular, when resident testicular macrophages are absent, Leydig cells fail to develop normally, whereas activated macrophages suppress Leydig cell steroidogenesis (for reviews see (85, 162, 167)). Androgen action via the peritubular myoid cell androgen receptor is also essential for the normal differentiation and function of adult Leydig cells (discussed below) (168). The nature of the factors and molecular mechanisms involved in intercellular communication between Leydig cells and the various other testicular cell types remains unknown.

ROLE OF PERITUBULAR MYOID CELLS

External to the basement membrane of the seminiferous tubule, are several layers of modified myofibroblastic cells termed peritubular myoid cells (PMCs) (169-170). PMCs are contractile and are responsible for the irregular contractions of the seminiferous tubules which propel seminiferous tubule fluid and released spermatozoa through the tubular network to the rete testis (171). PMC contractility is stimulated by various factors, reviewed in (171) including endothelin, prostaglandin F2 alpha and angiotensin (172-174). These contractions are associated with dramatic changes in PMC shape and their cytoskeletal actin networks (175). PMCs and Sertoli cells both contribute to the composition of the basement membrane that surrounds the seminiferous tubules, reviewed in (171). PMCs also produce various growth factors such as activin A and platelet derived growth factors (176-177), that may influence the function of other testicular cells, reviewed in (171).

PMCs have long been known to influence Sertoli cell function and protein expression, reviewed in (178) and the presence of Sertoli cells is required for normal PMC development and function (84, 97). PMCs influence Sertoli cell number, function and ability to support germ cell development, as revealed by studies in mice lacking androgen receptor expression in PMCs (179). This model also revealed that PMCs influence Leydig cell development and steroidogenesis (168). Further studies in transgenic mice reveal that an R-spondin receptor, LGR4, is selectively expressed in PMCs, participates in Wnt/β-catenin signaling and is necessary for germ cell development during meiosis (180). PMCs, under the influence of androgen, secrete the growth factor glial cell line-derived neurotrophic factor (GDNF), which is necessary for the maintenance of the spermatogonial stem cell niche (181-182). Therefore it is clear that PMCs modulate spermatogenesis via the regulation of Leydig, Sertoli and germ cell development and function.

 

THE REGULATION OF SPERMATOGENESIS

 

Many studies in the past 30 years have focused on the endocrine regulation of spermatogenesis. It is clear that the gonadotropins LH and FSH are required for the initiation and maintenance of quantitatively normal spermatogenesis. LH targets the Leydig cells to stimulate androgen biosynthesis, and the resulting androgens (testosterone and its androgen metabolites) act on receptors within the seminiferous epithelium to stimulate and support spermatogenesis. FSH targets receptors in the Sertoli cells directly to support spermatogenesis. However the roles of other endocrine factors, such as vitamin A and its metabolite retinoic acid, are emerging. While both androgens and FSH are required for optimal spermatogenesis (see below), spermatogenesis relies on the local production of growth factors, signaling molecules and other intrinsic mechanisms.

 

The following sections consider key aspects of the regulation of Sertoli cells and germ cell development and function, with the roles of the “traditional” endocrine regulators, androgen and FSH, briefly discussed at the end of each section. The role of estrogens in spermatogenesis is considered elsewhere in Endotext (Endocrinology of Male Reproduction Section, Chapter 17, Estrogens and Male Reproduction (144)).

 

Regulation of Sertoli cell Development and Function

The complexity of the Sertoli cell’s structure and function is reflected in the complexity of its regulation. A detailed review on the many factors regulating Sertoli cell function is out of the scope of this chapter, and only a few important functions will be discussed here. The reader is referred to the excellent book on Sertoli cell Biology (183) for comprehensive information.

The Sertoli cell population is specified in the embryonic testis, under the influence of male sex determining factors, such as Sry and Sox9, reviewed in (184-185). Newly-specified embryonic Sertoli cells enclose and form seminiferous cord structures around primordial germ cells. Expression of the retinoic acid degrading enzyme Cyp26b1 and other factors by early Sertoli cells (E12.5 in the mouse) controls the specification of primordial germ cells to commit to the male pathway of gene expression and meiosis (186). Sertoli cells proliferate and drive seminiferous cord elongation late in embryonic development; this process is dependent on activin A signaling from Leydig cells to Sertoli cells, reviewed in (185).

Sertoli cells proliferate in late fetal life and before puberty. Prior to puberty, the exit of Sertoli cells from an immature, proliferative phase to a non-proliferative, maturation phase represents an important cell fate decision that results in the establishment of the adult Sertoli cell population. Experimental modifications that interfere with these periods of Sertoli cell proliferation and maturation can impact on the ultimate size and spermatogenic output of the adult testis; extended periods of Sertoli cell proliferation increase testis size (e.g. (187)), whereas premature cessation of proliferation and entry into the maturation phase results in smaller testes (e.g. (188)). Several factors act as mitogens for immature Sertoli cell proliferation, including FSH (189), thyroid hormone (187), and transcription factors, such as Dmrt1 (190) and Rhox genes (191), and various other genes are essential for the proliferation to maturation switch, reviewed in (88).

As the Sertoli cells attain an adult phenotype capable of supporting sperm production, their nucleus moves to the base of the cell, they attain the specialized cytoskeletal features characteristic of these cells (192) and they form the so-called ‘blood testis barrier’ tight junctions necessary for the entry of germ cells into meiosis (78). As Sertoli cells develop during puberty and the first wave of spermatogenesis, they show an extraordinary degree of plasticity in terms of their gene expression program, which reflect functional changes, and their response to the appearance of different germ cell types, as they mature (193). In adulthood, Sertoli cells increase or decrease the expression of genes depending on the stage of the spermatogenic cycle (67). This cyclic expression of genes allows the Sertoli cell to respond to the changing needs of germ cells as they proceed through spermatogenesis.

It has been known for many years that an absence of vitamin A disrupts cyclic function of Sertoli cells and spermatogenesis. It is now clear that the metabolism of Vitamin A to the active metabolite retinoic acid (RA) is essential for the cyclic activity of Sertoli cells, reviewed in (194). Retinoic acid signaling is mediated through nuclear RA receptors (RARs) that bind to DNA and either activate or suppress target genes. Mice lacking RARα expression in Sertoli cells show disruption of the spermatogenic cell cycle, whereas the administration of exogenous RA to testes without advanced germ cells causes all Sertoli cells to “reset” to stage VII of the spermatogenic cycle (70). These studies indicate that RA is a master driver of Sertoli cell cyclic gene expression.

Multiple lines of evidence suggest there is a very specific pulse of RA synthesis at the mid-spermatogenic stages VII and VIII ((70, 195), reviewed in (194)) which have been confirmed by studies measuring RA in synchronized testes (196). This pulse may be achieved by a combination of events including an increase in RA synthesis enzymes (ALDH enzymes), a decrease in enzymes that store or degrade RA, and an increase in the RA uptake protein Stra6 in Sertoli cells. Advanced germ cells such as pachytene spermatocytes could possibly synthesise RA and may contribute to this mid-cycle peak (see (194). Recent studies suggest that ALDH enzymes are unlikely to play a major role in the mid-cycle RA pulse (197) but stage-specific expression of enzymes involved in the rate limiting conversion of retinol to retinaldehyde, or enzymes involved in retinol availability, could play a role (196-197). Termination of the RA pulse in late stage VIII could be facilitated by a sharp increase in the expression of the RA degradation enzyme Cyp26a1 (70), however other studies did not support this concept (196).

This pulse of RA in the mid-spermatogenic stages is thus likely to be a driver of Sertoli cell function. It not only appears to be necessary for the entry of spermatogonia into meiosis (see below) but it also likely regulates other important Sertoli cell functions occurring in these stages, notably sperm release (see (198) and references therein) and the formation and maintenance of the blood-testis-barrier (e.g. (197, 199-200), reviewed in (194)). Therefore the mid-cycle peak of RA likely drives these stage-specific Sertoli cell functions and cycle-dependent gene expression, highlighting its role as a driver of Sertoli cell cyclic function. The precise mechanisms governing the pulsatile nature of the RA production and response pathways in the seminiferous epithelium, however, remain to be elucidated.

FSH and Androgen Regulation of Sertoli Cells

Sertoli cells, unlike germ cells, express receptors for androgens and FSH, and thus “transduce” the effects of these hormones to the developing germ cells. Spermatogenesis does not proceed in the absence of androgens, whereas spermatogenesis can proceed but is quantitatively reduced in the absence of FSH (reviewed in (156, 201-203)). It is well known that both of these hormones are needed for quantitatively normal spermatogenesis. Both androgens and FSH have independent effects on Sertoli cells, but also act co-operatively and synergistically to initiate and maintain normal spermatogenesis and, by inference, optimal Sertoli cell functions.

 

FSH acts a mitogen for pubertal Sertoli cell proliferation and in the absence of FSH or its receptor, testes are smaller, Sertoli cell populations are reduced, as is sperm output (201, 204). Interestingly, FSH requires the insulin/IGF signaling pathways to mediate its effects on pubertal Sertoli cell proliferation (205). Thus FSH supports postnatal Sertoli cell proliferation to establish a quantitatively normal population and, since Sertoli cell number determines sperm output (see The Role of Sertoli Cells in Spermatogenesis), is required for the production of normal numbers of sperm. Another event occurring during the establishment of spermatogenesis is a wave of germ cell apoptosis that is important for establishing future spermatogenesis, perhaps by achieving a balance in the Sertoli cell:germ cell ratio (206). Since reductions in FSH at this time cause even greater apoptosis, it is possible that FSH acts on Sertoli cells to limit this apoptotic wave and establish normal spermatogenesis, reviewed in (201). FSH appears to support various Sertoli cell functions and their ability to support normal numbers of germ cells, as evidenced by reduced Sertoli cell-germ cell ratios in mice lacking FSHβ (207) and abnormal Sertoli cell morphology in mice lacking FSH receptor (208). FSH can maintain germ cell development in gonadotropin-deficient men for 6 weeks (209), and has permissive effects on spermatogenesis in non-human primates and men, see (210-211). Therefore FSH is not essential for spermatogenesis, but is required for normal Sertoli cell number and function.

 

Androgens, including testosterone and DHT, act on androgen receptors (AR) in the testis to support normal spermatogenic function. Androgens can act on the AR and produce the so-called classical signaling pathway, whereby ligand-bound AR translocates to the nucleus, binds to Androgen Response Elements (AREs) in the promoter region of androgen-dependent genes, and modulates transcription. This pathway produces a response hours after androgen stimulation. However androgens can have much more rapid effects via non-classical pathways, involving AR-mediated intracellular calcium influx or activation of SRC and the ERK phosphorylation pathway, reviewed in (212). Both classical and non-classical pathways are active in Sertoli cells (212) and both are necessary for spermatogenesis (213).

 

In the absence of AR in Sertoli cells, no sperm are produced and spermatogenesis is arrested at the end of meiosis (214-215), highlighting the fact that androgen action on Sertoli cells is needed for the completion of meiosis and spermiogenesis. Androgens regulate Sertoli cell number during pubertal development (reviewed in (201)) and are a driver of Sertoli cell maturation; this latter requirement was demonstrated in transgenic mice with premature activation of AR expression in postnatal Sertoli cells, causing Sertoli cells to prematurely exit the proliferative phase and enter the maturation phase, leading to a reduction in Sertoli cell numbers (188). Thus the precise timing of AR expression in Sertoli cells is important for normal testis development and optimal sperm output. Androgens are necessary for the normal formation of tight junctions between Sertoli cells which contribute to the blood-testis-barrier, reviewed in (201), and they drive the expression and translation of many genes expressed in the Sertoli cells themselves, and indirectly modulate the expression of genes in germ cells (e.g. (216)). Interestingly, Sertoli cell morphology, function and androgen-dependent gene expression is impaired when AR is ablated from peritubular myoid cells (179), indicating that androgen action on these cells also supports Sertoli cell function and spermatogenesis.

As reviewed extensively elsewhere, androgens and FSH have co-operative and synergistic effects on spermatogenesis (156, 201, 203-204) and, since Sertoli cells are the only testicular cells to express both FSH and androgen receptors, some synergistic actions likely occur within the Sertoli cells themselves. Their ability to support germ cells is impaired when Sertoli cells lack expression of either FSH receptors or AR, however the effect is exacerbated when both receptors are depleted (217). Similar synergistic actions of FSH and androgen in Sertoli cells are apparent when measuring the ability of Sertoli cells to release mature sperm at spermiation (218). FSH and androgen signaling pathways can converge in Sertoli cells, for example in activating the MAP kinase pathway and elevating intracellular Ca2+ levels, reviewed in (219) and both hormones co-operate to modulate the Sertoli cell expression of particular miRNAs (220).

 

 

Regulation of Spermatogonial Proliferation and Development

Spermatogonia and SSC reside within a specialized microenvironment within the testis known as the “niche”, where the balance between SSC renewal and differentiation is regulated. This niche is comprised of cells, extracellular matrix and soluble factors that regulate the functions of cells within the niche. Within this niche, the expansion of spermatogonial clones and their commitment to differentiation are the foundation for the continual production of spermatozoa during adulthood.

Maintenance of the niche and the balance between SSC renewal and differentiation in the testis is regulated by a number of factors, see (221-223) for reviews. The Sertoli cell directly dictates the number and function of spermatogonial niches (224). Sertoli cells secrete Glial-cell line Derived Neurotrophic Factor (GDNF) which acts on receptors on undifferentiated spermatogonia to control differentiation and self-renewal of SSC (225-229) via the regulation of several transcription factors (221). Sertoli cells also regulate the stem cell niche via the expression of the Etv5 gene and by mediating FGF9 responses, reviewed in (90) as well as by the production of other factors such as activin A, reviewed in (223).

Other somatic cells within the testis are important for SSC self-renewal and differentiation. An example is colony stimulating factor (Csf1), expressed by the surrounding peritubular myoid cells and Leydig cells, that has been demonstrated to be important for SSC self-renewal (230). Intriguingly, macrophages have recently been shown to be important for maintenance of the spermatogonial niche; distinct macrophage populations aggregate on the surface of the seminiferous tubule over regions containing undifferentiated spermatogonia, and their depletion disrupts spermatogonial differentiation (231). The mechanism by which local resident macrophages may promote spermatogonial differentiation is not yet known, but it may involve their expression of Csf1 and enzymes involved in retinoic acid synthesis (231).

In the neonatal period, the migration and proliferation of the primordial germ cells and the subsequent pre-spermatogonia (gonocytes) represents a crucial step in the establishment of spermatogenesis (232-233). In turn, the constant commitment of type A spermatogonia to differentiation and entry into meiosis is a key aspect driving the spermatogenic cell cycle (70) and thus in providing the “pulses” of sperm production along the seminiferous tubule. A fundamental requirement for both gonocyte differentiation and spermatogonial commitment to meiosis is the action of stem cell factor (SCF) produced by the Sertoli cells and its receptor, c-KIT, located on spermatogonia (232). Action of Sertoli cell-derived SCF on c-KIT induces the PI3 kinase signaling pathway in spermatogonia which is required for their entry into meiosis (234). The acquirement of c-KIT protein on the surface of spermatogonia is a key marker of differentiation and is essential for spermatogonial development and entry into meiosis (232-233).

Vitamin A and the retinoic acid signaling pathway are emerging as critical regulators of spermatogonial differentiation. As described above (see Regulation of Sertoli cell Development and Function) a “pulse” of retinoic acid is generated at the mid-stages of spermatogenesis via a tightly controlled series of events, including the regulation of retinoic acid synthesis and degradation enzymes (70, 196, 235-236). Retinoic acid is required for the differentiation of neonatal gonocytes and for the differentiation of spermatogonia in the post-pubertal testis, and thus is an essential factor required to drive entry of spermatogonia into meiosis, reviewed in (196, 222, 237). Ectopic expression of retinoic acid receptor gamma drives undifferentiated spermatogonia to differentiate (238), highlighting a direct action of retinoic acid on spermatogonia. Retinoic acid may drive spermatogonial differentiation by stimulating the PI3K-AKT-mTOR signalling pathway to induce the translation of c-KIT protein (239) as well as other proteins involved in spermatogonial differentiation (240).

 

FSH and Androgen Regulation of Spermatogonia

The above section demonstrates that local factors within the testis support the spermatogonial stem cell nice, the expansion of cohorts of both undifferentiated and differentiated spermatogonia and entry into meiosis. What impacts do the major endocrine regulators of spermatogenesis, FSH and androgen, have on spermatogonial differentiation and proliferation?

Combined suppression of androgen and FSH results in a relatively small decrease in spermatogonial populations in rodents but causes a major block in spermatogonial development in primates and humans, reviewed in (241). It is clear that androgen and FSH have supportive effects on spermatogonia, but there is species-specific variability in the sensitivity of these cells to each of these hormones, reviewed in (204, 241-242).

Spermatogonia lack receptors for both FSH and androgen and therefore actions of these hormones must be indirect, via Sertoli cells and/or other testicular somatic cells. Studies in rodents suggest that spermatogonial development is not particularly susceptible to a loss of androgens and that spermatogonia can enter meiosis in the absence of androgen action on Sertoli cells (e.g. (215, 243)). Ablation of AR within peritubular myoid cells results in reduced numbers of spermatogonia (179) however it is not clear if this is a peritubular myoid cell-mediated effect, or whether the high concentrations of testicular testosterone produced in this model have inhibitory effects on spermatogonia, as noted in other studies (244). Conversely, spermatogonia are very sensitive to FSH in rodents and monkeys, e.g. (243, 245-246), therefore it is possible that the major reductions in spermatogonial populations in response to androgen and FSH suppression in monkeys and humans is primarily a consequence of FSH, rather than androgen, depletion. The mechanism by which FSH supports spermatogonia is likely to be via stimulating the Sertoli cell to provide a supportive environment for maintenance of the SSC niche, as well as on spermatogonial proliferation and differentiation. Studies in rodents have shown that FSH regulates the levels of GDNF and FGF2 in Sertoli cells, which in turn are essential for spermatogonial development, reviewed in (247). A recent study in transgenic mice suggests that maintenance of the SSC niche is normal in mice lacking FSH and therefore it may not play a major role in stimulating GDNF-dependent effects on SSC (248). Interestingly, this study also revealed that SSC renewal is enhanced during LH (and probably testosterone) suppression, and this effect is mediated by the transcription factor Wnt5a in Sertoli cells (248); perhaps this mechanism could preserve the SSC pool in situations where endocrine factors are temporarily compromised.

Regulation of Meiosis

 

Meiosis technically begins with the differentiation of type B spermatogonia into preleptotene spermatocytes which begin DNA synthesis. However, spermatogonia become committed to further differentiation and entry into meiosis during the A to A1 transition; this commitment to meiosis is an irreversible step leading to the production of preleptotene spermatocytes (237). There is abundant evidence that entry into meiosis in both sexes, and the production of spermatocytes in males in particular, requires the RA pathway, reviewed in (237). In the absence of the RA-inducible gene Stra8, preleptotene spermatocytes are formed and replicate their DNA, but their subsequent entry into the meiotic prophase is prevented (249-250). RA also induces Rec8, a meiosis-specific component of the cohesion complex, in a Stra8-independent manner, suggesting that RA acts through multiple pathways to initiate meiosis (251). However initiation of meiosis is not solely dependent on RA, as it also requires a RA-independent protein, MEIOC, that stabilizes mRNA transcripts from multiple meiosis-associated genes (252).

Many studies, including those in transgenic mouse models, have identified proteins necessary for the completion of male meiosis, reviewed in (253). Targeted gene disruption approaches have also identified sexually dimorphic meiosis-associated proteins, suggesting different levels of checkpoint control between males and females, particularly in terms of chromosome recombination and homologous pairing, see (254). Failure of normal meiotic recombination events is related to an increased incidence of gamete aneuploidy, which has a higher incidence in infertile men compared to case controls, reviewed in (255). Many proteins have been shown to be essential for male meiotic recombination events, including those involved in synaptonemal complexes and DNA repair mechanisms, reviewed in (253, 255) . For example, genetic ablation of the DNA repair protein PMS2 results in very few synaptonemal complexes forming and improper homologous chromosome pairing (256). Meiosis is not arrested however, and some abnormal sperm are produced (256). The induction of spermatocyte apoptosis and arrest at the spermatocyte phase is commonly observed in other transgenic models in which the expression of other meiotic recombination proteins is perturbed , reviewed in (253).

Many proteins are required for male meiotic division, see (253). For example, the testis-specific heat shock protein, HSP 70-2, is essential for male meiosis. It is required for desynapsis of the synaptonemal complexes and for the activation of CDC2 to form the active CDC2/cyclin B1 complex to enable progression into the first meiotic division (34, 257). The ability of HSP 70-2 to activate CDC2 is regulated by the interaction with a testis-specific linker histone chaperone, tNASP (258). Recent studies have revealed that a neuropeptide, nociceptin, in Sertoli cells acts on its receptor in spermatocytes to stimulate the phosphorylation of Rec8, a key regulatory component of the cohesin complex that mediates chromosome dynamics during meiosis, including synaptonemal complex formation and chromosome recombination (259). Nociceptin-mediated Rec8 phosphorylation stimulates chromosome dynamics and meiotic prophase progression, reviewed in (260). These latter studies highlight the fact that the progression and completion of meiosis relies on cues from the Sertoli cell.

The transcriptional regulator A-MYB (encoded by the Mybl gene) is likely a important regulator of male meiosis (261). A point mutation in Mybl1 in mice causes spermatocyte arrest, aberrant chromosome synapsis, defects in DSB repair and abnormal cell cycle progression. Chromatin immunoprecipitation experiments revealed that A-MYB directly targets various genes involved in different aspects of meiosis, suggesting that A-MYB is a “master” transcriptional regulator of male meiosis (261).

FSH and Androgen Regulation of Meiosis

It is well known that the completion of meiosis requires androgen. Meiosis arrests at the pachytene/diplotene stage in mice lacking AR in Sertoli cells, and no haploid spermatids are produced (214-215). However, spermatocyte numbers are even further reduced when AR is ablated from peritubular myoid cells (179), suggesting that androgenic support of meiosis is mediated via both Sertoli cells and peritubular myoid cells. Meiosis was disrupted in pubertal rats when the non-classical AR pathway was blocked, suggesting that meiosis requires rapid actions of androgen on testicular somatic cells (213). Interestingly, while the completion of meiosis is absolutely dependent on androgen, it requires comparatively lower levels of androgen than the later process of spermiogenesis (203, 262-263).

Mice lacking FSH show a modest but significant reduction in the progression of meiosis (207), perhaps via effects on spermatocyte survival. It is well known the both FSH, as well as androgen, can support meiotic cell survival, particularly in the hormone-sensitive stages VII and VIII. Preleptotene and pachytene spermatocytes in stages VII and VIII are particularly vulnerable to FSH and/or androgen suppression, and apoptosis of these cells is a feature of gonadotropin suppression, reviewed in (203). The replacement of either FSH or androgen prevents spermatocyte loss/apoptosis in rodents (264) and humans (209), highlighting the fact that both of these hormones can support meiotic germ cell survival.

Regulation of Spermiogenesis and Spermiation

 

As discussed earlier in this chapter, the steps in the formation of a sperm from its precursor, the haploid round spermatid, represent a fascinating process in cell biology. The development of the sperm tail, the remarkable nuclear changes involving the condensation and complexing of DNA, the cessation of transcription and delay in protein translation, and the changes in the relative positions of the nucleus, cell organelles and the cytoplasm, all pose innumerable questions as to how these events are controlled. Many genes and proteins have been implicated in the control of these cellular processes during spermiogenesis, as demonstrated by transgenic mouse models, reviewed in (36-37).

The intrinsic and tightly-regulated control of gene transcription and translation is especially important for the complex cellular differentiation occurring during spermiogenesis. Haploid spermatids, as well as meiotic spermatocytes, express many unique genes that are not expressed in somatic cells (265). Alternative splicing is highly prevalent in the testis, and generates many germ cell-specific transcripts likely important for carrying out the ordered procession of germ cell development (266). One example of the importance for alternative splicing in spermiogenesis is the CREM gene, whereby the use of alternative splicing mechanisms regulates the expression of either repressor or activator forms of the CREM transcription factor (267).

Alternative polyadenylation is another mechanism that is particularly utilized within the testis to increase the diversity of the transcriptional program. mRNA polyadenylation involves cleavage of the pre-mRNA at its 3’ end, followed by the addition of multiple adenosine residues, creating what is known as the polyA tail. Polyadenylation can modulate the mRNA transcript’s stability, localization, splicing and translation (268). The position at which the polyA sequences are inserted can vary on a cell and tissue-specific basis, leading to a phenomenon known as alternative polyadenylation, reviewed in (269). Many mRNAs in the testis are subjected to alternative polyadenylation, and can lead to the production of germ cell-specific isoforms (269). RNASeq analyses have revealed widespread alternative polyadenylation (including within introns and exons) and 3’UTR shortening during germ cell development, with the shortest 3’UTRs observed in round spermatids (270). Not all round spermatid genes displayed shortened 3’UTRs, however those that did had functions associated with sperm maturation and protein ubiquitination. The authors propose that alternative polyadenylation is a major feature of germ cell development, and that 3’UTR shortening may be important for the storage and translation of spermatid-specific mRNAs during spermiogenesis (270).

Spermiogenesis uses other unique mechanisms to modulate transcription (also see Gene Transcription and Translation During Spermatogenesis: Roles of Noncoding RNAs and Epigenetic Modifiers). The transcription factor CREM is a master regulator of the transcription of many genes involved in haploid spermatid development reviewed in (271). The activation of CREM target genes is influenced by CREM binding to a spermatid-specific co-activator protein known as ACT. The localization of ACT in the nucleus of spermatids is controlled by a kinesin, whereby the kinesin effectively exports ACT out of the nucleus at certain stages, thus inhibiting CREM-dependent gene transcription (272). These studies reveal sophisticated and unique mechanisms governing the control of gene transcription during spermiogenesis. Other round spermatid transcription factors that could be “master transcriptional regulators” and influence the expression of a large number of genes involved in spermiogenesis include TRF2 (273) and RFX2 (274); the latter appears to target a cohort of genes involved in the development of the flagella (274).

As spermatids lose their ability to perform active gene transcription during the remodeling of their chromatin into the compact sperm nucleus, the post-transcriptional and translational control of gene expression becomes particularly important. All mRNA transcripts expressed in meiotic and post-meiotic germ cells are subjected to some degree of translational repression and there are many examples whereby genes transcribed earlier on in germ cell development are translationally repressed until the proteins are required during spermatid elongation, reviewed in (275). mRNAs are stored in free messenger ribonucleoproteins (mRNPs) for 3 or more days in round spermatids, followed by translational activation in elongating or elongated spermatids. The mechanisms governing translational repression are not well understood, but an emerging candidate is the YBX2 protein. This protein binds to sequences near the 3’ end of the 3’UTR in well known translationally repressed genes, such as Prm1, and likely interacts with various proteins and cis-elements to promote the assembly of a repressive complex that inhibits translation (276-277). YBX2 can therefore selectively inhibit the translation of certain genes, however it is also likely to participate in global mRNA translational repression in round spermatids (276-277).

The proper development of the sperm flagella is essential for sperm motility and hence fertility. Many proteins are now known to be required for flagella development and motility, reviewed in (37). Even structurally normal sperm can fail to move as shown by the genetic inactivation of the gene encoding a sperm calcium ion channel (278). Mutations in a number of genes required for assembly of the axoneme, such as dyenin, are associated with a syndrome known as Primary Ciliary Dyskinesia (PCD). PCD is associated with a range of pathologies, including male infertility, and is caused by a failure of proper development and function of cilia in various organs, including the sperm flagellum (279). The identification of the molecular mechanisms governing flagellar development and motility is important for the development of new therapies for male infertility.

Both spermiogenesis and spermiation appear to be regulated by the retinoic acid signaling pathway, reviewed in (194). Sertoli cell-derived RA acting on RARα/RXRβ heterodimers in Sertoli cells is essential for spermiation, reviewed in (35, 194). Deletion of the gene encoding RARα, Rara, from Sertoli cells causes abnormalities in both spermiogenesis and spermiation, reviewed in (194). Interestingly, the expression of Rara in spermatids rescues the spermiogenesis and spermiation defects seen in Rara null mice, suggesting that germline expression of Rara is also important for spermiogenesis and spermiation (280).

 

The regulation of spermiation is very complex, as reviewed extensively elsewhere (35, 60) The complexity of its regulation is due to the fact that spermiation is actually a multifaceted process involving a co-ordinated series of cellular processes, signaling cascades, endocytic pathways and adhesion complexes. Abnormalities in different aspects of spermiation are seen in many experimental settings, including the administration of pharmacological agents, toxicants and environmental stressors, the suppression of hormones and the introduction of genetic mutations (35, 59, 61). It seems likely that the Sertoli cell directs spermiation; the mature spermatid at this time is transcriptionally inactive and thus likely plays a fairly passive role in the process (35, 59). However, there are examples of mutations in genes expressed in spermatids that impair the ability of the spermatid’s cytoplasm to by removed during spermiation, leading to a failure of spermatid release, reviewed in (35). Therefore the regulation of spermiation is governed by the Sertoli cell, but defects within spermatids can influence their ability to be released.

FSH and Androgen Regulation of Spermiogenesis and Spermiation

Both spermiogenesis and spermiation are well known targets of androgen action in the testis. While the complete ablation of androgen action in Sertoli cells causes an arrest at the end of meiosis (214-215), androgen insufficiency causes a failure of round spermatids to attach to Sertoli cells and enter the elongation phase of spermiogenesis, and the failure of mature spermatids to be released at the end of spermiation, e.g. (218, 281-282), see (202-203) for review. Spermiation failure is an early feature of androgen suppression during adult spermatogenesis, however continued suppression eventually causes the death and/or detachment of round spermatids from Sertoli cells so that they are unable to elongate into mature spermatids (218).

Spermiation failure is observed when gonadotropins are suppressed in rodents, monkeys and men (241). It is induced rapidly after gonadotropin suppression and is the first morphological disturbance to spermatogenesis (35). In men undergoing gonadotropin suppression for the purpose of male contraception, spermiation failure can occur early in some men, leading to a rapid decline in sperm counts (283). Whether or not spermiation failure is achieved could determine whether male hormonal contraceptive-mediated gonadotropin suppression induces azoospermia (zero sperm in the ejaculate) or oligospermia (low but detectable levels of sperm in the ejaculate), reviewed in (35).

It seems likely that androgens and FSH co-operate to regulate spermiation. Acute suppression of FSH alone causes spermiation failure in rats (218), whereas the administration of FSH to men undergoing gonadotropin suppression can support spermiation (209). Suppression of either FSH or testosterone alone causes significant spermiation failure in rats, but the suppression of both has a synergistc effect, indicating that both hormones co-operate to promote spermiation (218). Thus the action of both testosterone and FSH on Sertoli cells support the normal release of sperm at the end of spermatogenesis.

Regulation of Gene Transcription and Translation During Spermatogenesis: Roles of Noncoding RNAs and DNA methylation

The long process of spermatogenesis, taking up to 64 days in men (284), involves an incredibly complex program whereby the transcription and translation of thousands of genes is precisely constrained as the germ cell proceeds through proliferation, meiosis and spermiogenesis. The male germ cell transmits both genetic and epigenetic information to the offspring, and as such the modulation of the germ cell genome has a major impact on subsequent generations. Epigenetic modifications of the genome are heritable; epigenetic processes such as DNA methylation and histone modifications regulate chromatin structure and modulate gene transcription and silencing.

The transcriptome of the male germ cell during meiosis and spermiogenesis is the most complex transcriptome of all tissues in the body; substantially more of the germ cell genome is transcribed and subjected to more complex alternative splicing compared to other tissues (285). The regulation of this transcriptome is central to successful spermatogenesis and for male fertility. The precise constraints on gene and protein expression in germ cells, and on the sperm genome as a whole, are achieved via a number of different processes including RNA binding proteins, epigenetic modifiers, such as DNA methylation and transposable elements, and multiple types of noncoding RNAs (ncRNAs). The following section provides a brief overview of the ncRNA and epigenetic processes that contribute to each stage of male germ cell development. The reader is encouraged to seek more detailed reviews on specific mechanisms, e.g. (286-292), and references therein.

ncRNAs do not encode proteins but regulate gene transcription and translation. They are arbitrarily classified into small ncRNAs of less than 200 nucleotides (nt) and long ncRNAs (lncRNAs, >200nt). Small ncRNAs are further sub-classified based on their size, function, mode of action and whether they interact with PIWI proteins (expressed only in the germline) or AGO proteins (widespread expression). Three major classes of small ncRNAs have been shown to play essential roles in spermatogenesis: 1) MicroRNAs (miRNAs) interact with AGO family proteins and generally act at the post-transcriptional level to regulate mRNA stability and/or translation; 2) Endogenous small interfering RNAs (endo-siRNAs) are derived from double stranded RNAs, interact with AGO proteins and can silence both gene and transposon transcripts; 3) PIWI-associated RNAs (piRNAs) are derived from single-stranded piRNA precursors and interact with PIWI proteins (a sub family of the AGO protein family). piRNAs are predominantly, but not exclusively, found in the male germline and regulate transposable element activity as well post-transcriptional gene expression and are required for normal spermatogenesis (see below). miRNA and endo-siRNA generation involves the RNA processing enzyme Dicer, whereas piRNA generation is Dicer-independent (reviewed in (291)).

Although these ncRNAs have various roles including regulating the epigenome (see below), they are probably best known for their role in RNA silencing; prevention of an mRNA transcript being translated into a protein. This is accomplished by the RNA silencing-induced complex (RISC), the core of which consists of an AGO/PIWI protein and the ncRNA that acts to “guide” the RISC to its target mRNA. Silencing of the target mRNA is then achieved by cleavage (by the “slicer” activity of various proteins), or by recruiting other proteins that affect translation, transcript stability or chromatin structure, reviewed in (286-287, 291).

The Embryonic Testis

The control of epigenetic modifications of the genome, and the participation of ncRNAs, is very important in the fetal testis. The genomes of primordial germ cells undergo widespread demethylation as they colonise the embryonic gonad; this erasure of epigenetic information allows the subsequent establishment of a germline-specific epigenetic pattern that is eventually transmitted to the offspring (reviewed in (292)). After sex determination in the embryonic gonads, primordial germ cells become committed to the male pathway of differentiation and cease proliferation, entering a period of mitotic quiescence. During differentiation and the subsequent mitotic quiescence, remarkable modifications are made to the male germ cell genome. After the erasure of vast areas of DNA methylation earlier in development, fetal male germ cells undergo the re-establishment of DNA methylation marks by de novo DNA methyltransferases; this process is essential for gametogenesis and creates an epigenome that is required for successful embryonic development of the offspring (reviewed in (292)). During this time there are also extensive histone modifications of the genome that impact on chromatin structure and, ultimately, on embryonic development of the offspring (reviewed in (292)).

A striking feature of fetal male germ cells is the regulation of transposons, or transposable elements, which is central to the successful development and function of the male gamete. Transposable elements are DNA sequences that are “mobile”; they can literally move from one area of the genome to another. Retrotransposons make up the majority of transposable elements in the genome and are replicated by a “copy and paste” mechanism whereby the transcription of the transposon’s DNA sequence is “copied” into RNA and, via reverse transcription, into DNA, and then inserted (pasted) into another area of the genome (293). Transposons can thus create heritable alterations of the genome. At least 48% of the human genome is comprised of transposable elements and these elements are a major driver of generating genetic diversity during evolution (see (294) for recent review). Transposons can modulate gene expression by a variety of mechanisms, such as by modulating regulatory elements within promoter regions or generating noncoding functional elements that will impact on gene transcription and translation (294). However, transposons rarely produce beneficial effects and instead could have potentially deleterious consequences, thus evolution has produced sophisticated mechanisms to control their activity.

Potentially harmful transposon activity in the genome is repressed by the methylation of transposon DNA sequences. However the genome-wide de-methylation that occurs during fetal male germ cell re-programming could leave the germ cell genome vulnerable to increased transposition. For example, deletion of the de novo DNA methyltransferase Dnmt3l or of the Morc1 gene in male germ cells disrupts the methylation of retrotransposon sequences and leads to an activation of retrotransposon transcription and the eventual failure of germ cell development (295-296). Thus the processes governing DNA methylation of transposons during male germ cell development is essential for safeguarding the genome against unwanted transposable element activity.

Another important mechanism that has evolved within germ cells for the control of transposable elements involves piRNAs. The fetal testis expresses a unique set of piRNAs, termed fetal piRNAs ((297), reviewed in (291)). piRNAs expressed in pro-spermatogonia in the fetal testis and in spermatogonia in the postnatal testis are further classified as “pre-pachytene piRNAs”, to distinguish them from piRNAs involved in the postnatal development of spermatocytes. Fetal pre-pachytene piRNAs associate with the PIWI proteins MIWI and MILI2, and approximately half of all fetal piRNAs arise from sequences within transposable elements (reviewed in (288)). piRNAs and the MIWI and MILI2 proteins are essential for transposable element suppression in the fetal testis (298-299). piRNAs, in association with PIWI proteins, appear to silence transposon activity in the genome at a) the posttranscriptional level, by targeting and cleavage of transposable element transcripts, and b) at the epigenetic level, via the recruitment of DNA methylation machinery to re-establish repressive methylation marks on the promoters of transposable elements (reviewed in (286, 288, 291)). Thus piRNAs and their associated proteins defend the genome against inappropriate transposable element activity during fetal male germ cell development (287).

 

Although mechanisms to control transposon activity in the male germline have evolved, it is apparent that fetal male germ cells are still inherently vulnerable to transposable elements during genome de-methylation and re-methylation. Intriguingly, it has been proposed that the transposon-mediated generation of genetic diversity within individual male germ cells could be critical for the evolution of complex species such as mammals (300). Evolution is driven by a basic algorithm of “generate variation and test”: the generation of individuals with genetic and phenotypic variation, and the subsequent natural selection of those variants that offer the best opportunity to survive and reproduce. Transposons introduce genetic diversity, particularly into the promoter regions of the germ cell genome, and could thus be an important driver of the generation of genetic variation. The consequences of such genetic diversity derived by transposable elements are then tested by the subsequent survival and reproduction of the offspring. However, this “testing” of the genetic variation could also occur during the spermatogenic process itself, as individual germ cells proceed through spermatogenesis and fertilization. Such tests could include whether the gamete: is eliminated via apoptosis during spermatogenesis; is released by the Sertoli cell at the end of spermiation; survives and traverses the female reproductive tract; achieves fertilization; contributes to a viable zygote. Therefore transposon-mediated shuffling of the germ cell genome and the subsequent selection of sperm could be an important driver of mammalian evolution (300).

The Postnatal Testis

The male germline expresses high levels of ncRNAs that are involved not only in the generation of sperm, but also in shaping the sperm epigenome and in the ability of the sperm genome to have influence future generations (e.g. (286-288, 291, 301-302)). In germ cells, a specialized organelle known as the nuage, or germ granule, exists. The germ granule contains various ncRNAs and other related molecules (290, 303) and changes its structure and composition as germ cells develop through the fetal and postnatal periods. The germ granule exists in a form known as intermitochondrial cement (IMC) in fetal germ cells, postnatal spermatogonia and spermatocytes and as an intriguing germ cell-specific structure called the chromatoid body in spermatocytes and spermatids (290, 304). In round spermatids, the chromatoid body is highly mobile, moving rapidly around the nucleus, frequently making contact with nuclear pores, and even moving across intercellular bridges into adjacent spermatids (290). The chromatoid body is thought to function as an organizing center for RNA and ncRNA, performing important roles in the post-transcriptional processing of germ cell gene products (290).

While the modulation of DNA methylation of the epigenome of germ cells is a major feature of embryonic testis development (see above), it is worth noting that epigenetic modifications of the DNA in developing postnatal germ cells is also important for successful spermatogenesis, see (305). An example of this is the epigenetic “switch” involved in spermatogonial differentiation (306). Spermatogonia exhibit major epigenetic differences in DNA methylation patterns as they develop from Aal to A1 spermatogonia, and the DNA methylation machinery is involved in the shift from an undifferentiated, KIT- state towards a differentiating KIT+ state (306). Changes in DNA methylation of the germ cell genome throughout meiosis and spermiogenesis are associated with the ability of germ cells to transcribe RNA (285). Epigenetic modifications in the mature sperm are particularly important for the development of the offspring (see next section).

Analysis of the testis transcriptome has revealed that spermatocytes and spermatids transcribe more of their genome than any other tissue examined (285). While round spermatids are very abundant and are a major contributor to the testicular transcriptome, pachytene spermatocytes transcribe very high levels of RNA, ~6 times more than round spermatids, and therefore also contribute to the diversity of the testis transcriptome. Spermatocytes and spermatids transcribe substantially more genic and intergenic regions of DNA than other tissues, including many lncRNAs and pseudogenes, and exhibit a much more complex pattern of alternative splicing. The widespread transcription of the genome in these cells is associated with decreased DNA methylation and an open and transcriptional active chromatin state (285); presumably this open chromatin state is a consequence of the dramatic remodeling of the chromosomes and chromatin that occurs during meiosis and spermiogenesis. This “promiscuous” germ cell transcription is conserved across amniote species and could have important evolutionary consequences (285). While it is likely that much of this transcription is “leaky’” and non-functional, it could also be associated with the emergence of new genes and the generation of genetic diversity during mammalian evolution (285).

miRNAs are highly conserved and bind to complementary sequences in target mRNAs, preventing their efficient translation into proteins via a number of mechanisms including transcript cleavage and destabilization. A single miRNA can target many mRNA transcripts, and a single mRNA transcript can be the target of multiple miRNAs; in this way miRNAs are estimated to regulate ~60% of the protein coding genes in the genome (reviewed in (286)). miRNAs are generated from short hairpin loop RNA sequences that are subjected to a series of processing steps in the nucleus and then in the cytoplasmic RISC, reviewed in (286, 291, 307-308). miRNAs commonly arise from sequences within the introns of protein coding genes, reviewed in (286), and miRNA genes are significantly enriched within the X chromosome compared to autosomes, see (291). The enzymes RNA processing enzymes DROSHA and DICER are essential for miRNA biogenesis and are both required male fertility, see (286, 291, 307); spermiogenesis is disrupted when Drosha and Dicer are ablated from postnatal germ cells (309). Many miRNAs are preferentially expressed in the testis and in particular germ cells, including in spermatids and spermatozoa (310), reviewed in (286, 291). A number of germ cell miRNAs have now been shown to play defined roles during spermatogenesis, reviewed in (286, 291). Androgens and FSH can regulate particular miRNA species in Sertoli cells which in turn modulate the expression of particular proteins (220). Various studies suggest a correlation between altered miRNA profiles and particular disorders of human spermatogenesis, suggesting that miRNA-regulated pathways have important consequences for human male fertility, reviewed in (286).

The role of endo-siRNAs in spermatogenesis is less clear, but these small RNAs have the potential to influence the spermatogenic transcriptional program. As is the case for miRNAs, endo-siRNAs require processing by DICER and interactions with AGO proteins to exert their RNA interference activity, however unlike miRNAs, endo-siRNAs do not require processing by the DROSHA enzyme reviewed in (286, 291). In C. elegans, the male germline expresses specific endo-siRNAS that are important for spermatogenesis (311) and mutants with defective endo-siRNA expression exhibit male sterility (312). Mouse spermatogenic cells express 75 endo-siRNAs that have the potential to target hundreds of transcripts (313). Interestingly, the fact that these endo-siRNAs map to thousands of sequences within DNA (313) has lead to the hypothesis that these small RNAs could have an impact on the sperm epigenome (291, 313).

piRNAs are essential for adult spermatogenesis. This class of ncRNA consists of sequences ~25-30nt in length, slightly longer than miRNAs and siRNAs (288). piRNAs are predominantly expressed in the germline, however piRNA-like species (pilRNAs) have now been described in various somatic cells, including Sertoli cells (314). Millions of distinct piRNA sequences are thought to exist in mammals, although these sequences are poorly conserved between species (288). Their biogenesis is distinct from, and less well characterized than, the biogenesis of miRNAs, and piRNAs are 2’O-methylated on their 3’ end to prevent their degradation (286-288). piRNAs specifically interact with the PIWI sub-family of the AGO proteins, which includes PIWIL1, PIWIL2, and PIWIL4 (also known as MIWI, MILI, and MIWI2, respectively); PIWIL2 and 4 interact with piRNAs in gonocytes whereas PIWIL1 and 2 interact with piRNAs in meiotic and post-meiotic germ cells. Different sub-species of piRNAs exist in the postnatal testis: the so-called “pre-pachytene piRNAs” are expressed in fetal gonocytes and spermatogonia in the postnatal testis; whereas “pachytene piRNAs” are expressed in spermatocytes and spermatids of the postnatal testis. While pre-pachytene piRNAs are often derived from transposon sequences (see above), pachytene piRNAs are mostly derived from intergenic regions known as piRNA clusters, reviewed in (288). Pachytene piRNAs constitute approximately 95% of piRNAs and are very highly expressed in meiotic and post-meiotic germ cells, reviewed in (286-288, 291). A fundamental role for piRNAs in adult spermatogenesis has been revealed in many studies, reviewed in (286-288, 291); transgenic mice with targeted disruption of piRNA interacting proteins are usually infertile, with germ cell DNA damage and an arrest of spermatogenesis during meiosis or spermiogenesis being commonly observed, see (288).

While a role for piRNAs in the regulation of the epigenome in fetal gonocytes has been well described (see above), the specific roles of piRNAs in adult spermatogenesis are less clear, possibly because piRNAs could have widespread functions in the postnatal testis. Elevation of transposon sequences is seen in adult germ cells from mice with various genetic defects in piRNA associated proteins (e.g (315-317)), indicating that piRNAs may also repress transposable elements during adult spermatogenesis. Consistent with this proposition, the most abundant piRNAs in human sperm target LINE1 retrotransposon sequences (318). Various studies suggest that piRNAs are essential for the execution of the complex meiotic and post-meiotic transcriptional program (e.g. (319-321)). Pachytene spermatocytes from transgenic mice lacking a functional Henmt1 methylation gene have abnormally methylated piRNAs, which influences their stability and results in their degradation (315). Spermatocytes from these mice had a more “open” and transcriptionally permissive chromatin state compared to wildtype, suggesting a role for piRNAs in maintaining normal chromatin structure in germ cells. This abnormal chromatin state was associated with premature germ cell gene transcription, suggesting that piRNAs might regulate postnatal germ cell gene transcription via epigenetic mechanisms (315). Another way that piRNAs may influence the germ cell transcriptional program is by negatively regulating the expression of mRNAs from particular protein coding genes as well as lncRNA transcripts (320). Intriguingly this latter study also revealed that piRNAs can arise from non-coding pseudogenes and target the mRNAs arising from that pseudogene’s cognate functional protein-coding gene (320). Consistent with this finding, human sperm contain piRNAs that arise from pseudogenes and are predicted to target the expression of protein-coding genes (318). Thus the mechanisms by which piRNAs and their associated proteins regulate the spermatogenic program are likely many and varied, and the role of the piRNA pathway in spermatogenesis is the subject of ongoing studies.

In comparison to short ncRNAs, lncRNAs are less well studied and are a relatively recent addition to the field of male fertility research. lncRNAs (generally >200nt) can regulate gene expression by a number of mechanisms. For example, lncRNAs can act as repressors or enhancers of epigenetic modifiers of the genome, and can influence gene expression by regulating DNA methylation and histone modifications, reviewed in (322-323). RNASeq experiments show that lncRNAs are more abundant in testis than other tissues; this enrichment of lncRNAs in the testis is due to an over abundance of lncRNA transcripts particularly in spermatocytes and spermatids (285). lncRNAs are significantly more testis-specific than mRNAs (324), suggesting a particular requirement of lncRNAs for the spermatogenic process. Consistent with this, mouse germ cells at each developmental stage express specific lncRNAs (325). Experiments performed in Drosophila ablated 105 of the 128 testis-specific lncRNAs; a third of these mutants showed reduced or ablated male fertility and defects in spermiogenesis, suggesting that lncRNAs are particularly important for spermatid development (326). lncRNAs have also been implicated in regulating other aspects of germ cell development, reviewed in (322-323). A recent study identified the elegant mechanisms by which lncRNAs can influence spermatogenesis by studying a lncRNA essential for the maintenance of the spermatogonial stem cell niche (327). Transcription of this lncRNA (lncRNA03386) was stimulated in SSCs by the growth factor GDNF. This lncRNA is an antisense transcript of the Gfra1 gene, the receptor for GDNF, and it interacts directly with the Gfra1 gene, stimulating its transcription. Therefore GDNF stimulates the expression of a lncRNA which in turn enhances expression of its own receptor and facilitates its ability to stimulate SSCs (327). lncRNAs will likely emerge as important regulators of spermatogenesis and male fertility.

 

Sperm Epigenetic Modifications and Transgenerational Inheritance

 

Many epidemiological studies have shown that parental exposure to various lifestyle and environmental factors can increase the risk of chronic, non-genetic diseases in offspring, suggesting that epigenetic factors are transmitted from parents to their children. It is now clear that epigenetic modifications of the germ cell genome can be inherited and impact on multiple generations of offspring, i.e. have transgenerational effects. As detailed above, the genome of male gametes is remodeled during embryogenesis and postnatal spermatogenesis, resulting in the genome of mature sperm being extensively modified by DNA methylation and the retention of specific histone modifications, reviewed in (328). Alterations to the male germ cell epigenome can thus arise during the male’s embryonic development or during postnatal spermatogenesis (329).

 

There are now many examples of alterations in the sperm epigenome impacting on subsequent generations, reviewed in (330-331). The first evidence of epigenetic transgenerational inheritance via the male germ line came from studies in mice exposed to the endocrine disruptor vinclozolin, which is an agricultural fungicide with antiandrogenic activity, reviewed in (332-333). Female mice exposed to vinclozolin produced male offspring with spermatogenic and fertility defects and altered sperm DNA methylation; changes in the expression of DNA methylation enzymes and the sperm epigenome arose during the males’ embryonic exposure to vinclozolin (334) and these alterations were transmitted via the male germ line through subsequent generations of male offspring (335). Paternal obesity can also alter the sperm epigenome and have transgenerational impacts on offspring. Obesity induced by a high fat diet in male rats results in their female offspring exhibiting increased adiposity, insulin sensitivity, impaired glucose metabolism and pancreatic β cell dysfunction (336). Male mice fed a high fat diet also produce offspring with increased adiposity and insulin resistance, and this phenotype is associated with altered testicular mRNA, DNA methylation and sperm miRNA signatures in the fathers (337). While maternal deficiencies in folate are well known to cause abnormalities in offspring, it has been shown that male mice consuming a diet low in folate have altered sperm epigenetic profiles and produce offspring with various birth defects (338), providing further evidence of paternal diet being able to influence the sperm epigenome and the health of future generations. It is also important to note that alterations in the epigenome of sperm in men is associated with sperm quality and can influence their fertility, reviewed in (339).

 

Therefore it is clear that a man’s sperm epigenome can be altered by environmental (including diet and lifestyle) factors throughout his lifetime (329), and this altered sperm epigenome can influence both his fertility and the health of his future children. The mechanisms of male-specific transgenerational inheritance could involve multiple factors, such as the sperm epigenome, seminal fluid signaling and microbiome transfer (340). Transgenerational effects can be mediated by sperm via the alteration of its epigenome by DNA methylation machinery and the regulation of histone modifications, but also by RNAs and proteins within the sperm that can diffuse into the oocyte at fertilization (341). Some authors have speculated that alterations in sperm DNA methylation and histone marks may have less of an impact on subsequent generations, whereas sperm-borne RNAs could be of greater importance (328, 341). In C.elegans, male germline epigenetic inheritance involves Argonaute proteins and the generation of small ncRNAs that target female-specific germline mRNAs (342). Paternal miRNAs and endo-siRNAs in mouse sperm can regulate the transcriptome of fertilized eggs and early embryos (343) and traumatic stress in early life in male mice can impact on the health of subsequent generations via sperm-borne small RNAs (344). Recent studies have shed new light on an intriguing mechanism by which diet-induced changes in the sperm epigenome can impact on the offspring. A low protein diet in male mice affected the complement of small tRNAs fragments in sperm, and these tRNA fragments regulated genes that are highly expressed during early embryo development (345). Surprisingly, the sperm acquired this tRNA fragment complement during their post-testicular maturation as they traversed the epididymis, via the release of small vesicles called epididymosomes from epididymal cells (345). Therefore multiple pathways exist that can modulate the paternal sperm epigenome to impact on the offspring.

 

THE HYPOTHALAMIC-PITUITARY-TESTIS AXIS

The successful initiation of testicular function is dependent on the hypothalamic secretion of GnRH which in turn stimulates FSH and LH to act on the testis. These actions initiate spermatogenesis and testosterone production.It is well recognised that the testis in turn, through the secretion of hormones produced in the Sertoli and Leydig cells, exerts a negative feedback control on the production of gonadotropins.

The presence of such a negative feedback control by the testis on pituitary FSH and LH secretion is best demonstrated by the rapid rise of FSH and LH after castration. The mechanisms by which the secretion of FSH and LH increases in response to castration involves a rise in the hypothalamic secretion of GnRH and also involves direct actions at the pituitary level which allow an increase in pulse amplitude. Further, the fact that LH and FSH are co-secreted by the majority of gonadotroph cells in the anterior pituitary raises a number of unresolved questions as to how GnRH and the inhibitory signals act on the pituitary to result in the differential regulation of FSH and LH secretion.

The secretion of the gonadotropins FSH and LH are regulated by the episodic secretion of gonadotropin releasing hormone (GnRH) produced in the hypothalamus (also see Endotext, Endocrinology of Male Reproduction section, Chapter 5,Hypogonadotropic Hypgonadism (HH) and Gonadotropin Therapy (346)). There is now a substantial body of evidence that indicates that the kisspeptins, a family of neuropeptides localized to the arcuate nucleus of the brain are upstream regulators of GnRH secretion (for reviews see (347) (348). For instance, arcuate kisspeptin-neurokinin B-dynorphin expressing hypothalamic neurons are critically involved in the increase in gonadotropin secretion that occurs after gonadectomy (349). The regulation is further complicated by the isolation and characterization of gonadotropin-inhibitory hormone (GnIH), which acts both upstream of GnRH and also may operate at the levels of the gonads as an autocrine/paracrine regulator of steroidogenesis (350-351).

The pituitary secretion of FSH and LH by the gonadotrophs is also controlled by the feedback inhibition that occurs via the steroids, testosterone and estradiol (for an extensive review on the role of estradiol in the hypothalamic-pituitary-testis axis, see Endotext, Endocrinology of Male Reproduction section, Chapter 17, Estrogens and Male Reproduction (144)). The secretion of FSH and LH is also regulated by protein inhibitors, inhibin, secreted by the gonads, and follistatin, produced locally within the pituitary by the follicular-stellate cells (352), reviewed in (353). Follistatin exerts its inhibition of FSH secretion by its capacity to bind and block the actions of the activins A and B, the latter locally produced by the pituitary gland (354).

Control of LH Secretion

There is a substantial body of evidence to indicate that the steroid hormones testosterone, estradiol and dihydrotestosterone inhibit LH secretion (355). The demonstration that non-aromatisable androgens could inhibit LH secretion established that testosterone can exert its action directly without metabolism to estradiol or dihydrotestosterone (356-357). From the studies by Santen and Bardin (358), it is evident that testosterone acts at the hypothalamic level by decreasing GnRH pulse frequency without a change in pulse amplitude. The action of estradiol appears to be predominantly at the pituitary where it decreases LH pulse amplitude without changing pulse frequency (359). Further support for the action of testosterone at the hypothalamus emerged from the observation of a decrease in GnRH pulse frequency in portal blood (360). In addition, these studies demonstrated that treatment with estradiol lowered LH levels by decreasing LH pulse amplitude without altering GnRH secretory patterns in portal blood. These conclusions have been challenged by observations that a selective aromatase inhibitor, anastrozole, caused an increase in LH pulse amplitude and pulse frequency (359). These changes were seen in the presence of increased testosterone concentrations and were accompanied by an increase in LH and FSH. The investigators concluded that estradiol exerted a negative feedback by acting at the hypothalamus to decrease GnRH pulse frequency and at the pituitary to decrease the responsiveness to GnRH, both actions lowering LH secretion. Also see Endotext, Endocrinology of Male Reproduction section, Chapter 17, Estrogens and Male Reproduction (144).

Control of FSH Secretion

In addition to their feedback regulation of LH, testosterone and estradiol are also capable of suppressing FSH in the male (361). For many years, it was proposed that the action of the steroid hormones could account for the entire negative feedback exerted on FSH levels by the testis despite the existence of a hypothesis that a specific non-steroidal FSH feedback regulator named inhibin existed (362).

Over the past thirty years, a substantial body of evidence has accumulated to confirm the existence of a glycoprotein hormone termed inhibin that exerts a specific negative feedback inhibition on FSH secretion at the pituitary level (363). Two forms of inhibin have been isolated, namely inhibin A and inhibin B (364-367). These proteins represent disulphide-linked dimers of an α and β subunit. The alpha subunit is common both to inhibin A and B but the β subunits, though closely related, are different (αβA = inhibin A: aβB = inhibin B). Both inhibin A and inhibin B have the capacity to specifically inhibit FSH secretion by pituitary cells in culture. However, the circulating form in males is inhibin B. In contrast, dimers of the β subunit, termed activins (activin A = βAβA: activin B = βBβB; activin AB = βAβB) all have the capacity to stimulate FSH secretion by pituitary cells in culture (368-369). Finally, a structurally unrelated protein termed follistatin, has the capacity to suppress FSH secretion specifically by pituitary cells in culture (370-372). This action has been demonstrated to be due to the capacity of follistatin to bind and neutralize the actions of activin thereby suppressing FSH secretion (373).

In men and males from other species, testosterone, when administered in an amount similar or greater to its production rate, can suppress FSH as well as LH (355). However, in most instances there was a parallel and often greater suppression of LH secretion in contrast to the actions of inhibin (361). Further, there appears to be a difference in the response of FSH to testosterone in primates, where the actions are totally inhibitory in contrast to rats, where following an initial suppression of FSH by testosterone, higher doses caused a return of FSH levels to baseline (374-375).

Clear evidence for a physiological role of testosterone in the control of FSH can be shown in experiments in which the Leydig cells were destroyed by the cytotoxin ethane dimethane sulphonate (EDS). This treatment results in a rapid decline in testosterone levels and a concomitant increase in FSH concentrations to levels which were only 50% of those found in castrates (376). Since the inhibin levels in these experiments did not change, the maintenance of FSH levels at 50% of those seen in castrate animals was likely to be due to the continuing feedback control by inhibin (377). Further support for the dual role of testosterone and inhibin in the control of FSH emerged from the use of EDS in cryptorchid rats where baseline FSH levels were increased in association with decreased inhibin concentration. The removal of testosterone feedback in these animals with low basal inhibin levels resulted in an increase in FSH to the castrate range (378). The observation of an increase in FSH levels in men treated with a selective aromatase inhibitor raised the possibility that estradiol exerts a negative feedback action on FSH especially since the treated men experienced a concomitant significant increase in testosterone (359).

It is now well accepted that in the male, inhibin is produced by the Sertoli cell and is secreted both basally across the basement membrane of the seminiferous tubule and also into the lumen (379-380). Several studies have now demonstrated that the predominant form of inhibin secreted by the testis is inhibin B since the predominant mRNA was βB (381-382). The levels of inhibin B in males, measured by a specific ELISA, are inversely related to the levels of FSH (383-384). However, FSH predominantly stimulates inhibin α subunit production and does not alter the β subunit message (379, 385). This action results in the testis predominantly secreting inhibin rather than activin. Further support for this concept emerges from the studies of men undergoing chemotherapy where declining inhibin B levels are associated with a rise in FSH. However, with assays that detect α subunit products, there was a clear increase in these substances under the stimulation of elevated FSH levels (386). There is also evidence that a subunit of inhibin can be produced by Leydig cells (387) and increased LH levels result in the release of α subunit products into the circulation (388-389). There is still controversy as to whether the Leydig cells can produce bioactive inhibin (387).

In men, testosterone-induced gonadotropin suppression reduced circulating inhibin B and α subunit (measured as the pro-alpha C form of the α subunit) levels by only 25% and 50%, respectively, indicating that their secretion is not fully gonadotropin-dependent (390). In that model, exogenous FSH and LH both restored pro-alpha C levels supporting the view that Sertoli and Leydig cell are the origins of alpha subunit peptides, respectively, but only FSH restored inhibin B presumably reflecting Sertoli cell βB synthesis.

While there is evidence that the Sertoli cells, Leydig cells and peritubular myoid cells can produce activin, castration does not result in a decrease in circulating activin A levels (176, 391-393). Unfortunately, due to the lack of a suitable assay to measure activin B, there is no data available concerning the behaviour of this substance after castration. While activin acts on the pituitary, it also exerts local actions within the testis such as the stimulation of spermatogonial mitosis (394), Sertoli cell mitosis during testis development (103-104, 395-397) and possibly acts directly on germ cells (398).

Follistatin is also produced in the Sertoli cells, spermatogonia, primary spermatocytes and round spermatids in the testis (399-400). However, castration does not result in a net decrease in follistatin levels in the circulation suggesting that the testis does not contribute significantly to circulating levels of follistatin (401). In fact, in these studies follistatin levels rose but the rise was also found in the sham operated rams indicating that the follistatin response was part of the acute phase response to surgery, further supported by the demonstration that IL1β could also cause such an increase (402).

The fact that activin and follistatin remain unchanged after castration yet inhibin B in the circulation becomes undetectable strongly suggests that the gonadal feedback signal on FSH secretion is inhibin B. This is supported by studies in arcuate nucleus-lesioned monkeys maintained on a constant GnRH pulse regime, where testosterone could prevent the post-castration rise in LH but not FSH (403) (for review see (347)). The infusion or injection of recombinant human inhibin A in several species caused a rapid and specific fall in FSH secretion (404-406) and inhibin A administration to castrate rams suppressed FSH levels in the absence of testosterone (407).

Activin and follistatin can exert a paracrine role directly in the pituitary gland. The α and β subunit mRNAs are present in gonadotropes within the pituitary gland (408). The studies of Corrigan et al (409) strongly suggest that these substances exert a local action on FSH secretion since the inhibition of the action of activin B in pituitary cells in vivo suppressed endogenous FSH secretion. Follistatin mRNAs are also present in a number of different pituitary cell types including the folliculo-stellate cells (408, 410). This local production of follistatin also has the capacity to regulate the actions of activin (411). Additionally, the studies of Bilizekian et al have demonstrated that GnRH and the sex steroids estradiol and testosterone can modulate the local production of α, βA, βB and follistatin mRNAs within the pituitary (412-413). Clearly these interactions are complex and no clear answer can be given as to the relative roles of paracrine and endocrine actions of these glycoprotein hormones.

Some correlative evidence supporting the action of inhibin on FSH secretion is the decrease in inhibin production by Sertoli cells in parallel with the rise in FSH in a number of models of spermatogenic damage (414-415). The levels of circulating inhibin B appear to be inversely related to the levels of FSH following testicular damage in a number of studies (383-384, 416). Further, even in studies of large numbers of normal men, there is an inverse relationship between serum inhibin B levels and FSH (416). It is therefore likely that the actions of inhibin are predominantly exerted through secretion from the testis and transport via the peripheral circulation whereas the actions of activins and follistatin on FSH secretion occur through paracrine actions at the level of the pituitary gland. Further evidence supporting the stimulation of FSH by activin secretion emerges from the decline in FSH levels in mice with targeted disruption of the activin type II receptor gene (417).

 

SUMMARY OF THE ENDOCRINE REGULATION OF SPERM PRODUCTION: CLINICAL CONSIDERATIONS

Androgens and Spermatogenesis

The primary stimulus for the initiation of spermatogenesis is the LH-induced rise in testosterone at puberty. The absolute requirement of androgen for the initiation of spermatogenesis is demonstrated by the ability of the non-aromatisable androgen DHT to initiate complete spermatogenesis in hpg mice (418), and by the observation that spermatogenesis proceeds only to meiosis in mice lacking Sertoli cell AR expression (214-215). While androgens together with FSH are required for quantitatively normal spermatogenesis (see below), it is clear that androgens can initiate and support some degree of sperm production. Once spermatogenesis has been initiated during puberty, androgen alone can restore or maintain adult sperm production after experimentally-induced gonadotropin suppression, as has been demonstrated in many rodent, primate and human studies (reviewed in (156, 203, 241, 263, 419)).

By virtue of its local production in the testis, testicular concentrations of testosterone are 50 fold higher than that is serum, and are above those required for the initiation and maintenance of spermatogenesis. Adult spermatogenesis can be maintained by testicular testosterone levels at least 4 fold lower than normal as demonstrated in rodent models (420), reviewed in (419). When testicular testosterone levels are low, such as in the pre-pubertal testis and during gonadotropin suppression, the 5α-reduction of testosterone to the more potent androgen DHT appears necessary to amplify the androgenic signal and exert its stimulatory effects on spermatogenesis, as highlighted by studies in rodents (reviewed in (203)). However in the normal adult testis when testosterone levels are very high, it is likely that testosterone acts directly on the AR to maintain androgen-dependent functions (421).

The initiation of spermatogenesis during puberty requires a higher concentration of androgen than is required to maintain adult spermatogenesis once it is initiated, as exemplified by studies in hpg mice (422). Also, the restoration of adult spermatogenesis following gonadotropin suppression occurs over a very narrow dose range, wherein small changes in testicular androgen levels can produce large changes in sperm production, reviewed in (419). It is also worth noting that even very low levels of androgen are likely to produce a stimulatory effect on spermatogenesis. This can be illustrated by the demonstration of low levels of sperm production in older mice lacking LH receptor expression (423). Therefore, when considering the androgenic stimulation of adult spermatogenesis, “a little goes a long way”, and continued androgen action on AR can occur in the absence of gonadotropin stimulation, reviewed in (419).

Within the testis, AR is expressed in Sertoli cells, peritubular myoid cells, Leydig cells and vascular endothelial cells ((424-426), whereas germ cells lack AR and rely solely on somatic AR expression (427-428). Therefore androgens act on AR within the testicular somatic cells to support spermatogenesis. Studies in mice show that androgen action on AR in each of the testicular somatic cell types is important for testis function. AR expression in Sertoli cells is essential, as no sperm are produced in mice with targeted deletion of Sertoli cell AR expression (214-215) or in mice where the DNA binding domain of Sertoli cell AR has been deleted (429). However AR expression in peritubular myoid cells is also important for normal spermatogenesis (179) and for development and function of Leydig cells (168). The autocrine action of androgen on AR in Leydig cells is required for normal steroidogenesis and hence optimal testosterone production (428), and AR in endothelial cells of the testicular arterioles is involved in maintaining normal fluid dynamics and vasomotion in the testis (426). In summary, androgens act on AR in various testicular somatic cells, but not germ cells, to support normal testicular function and sperm production.

As summarized above, various phases of germ cell development are known to rely on androgen action. In the absence of androgen signaling in Sertoli cells, spermatocytes cannot complete meiotic division, and no haploid round spermatids are produced e.g. (214-215, 217, 429). The progression of haploid spermatids through spermiogenesis also relies on androgens, and in the absence of androgen, round spermatid development is halted during mid-spermiogenesis due round spermatid apoptosis and an inability of newly elongating spermatids to adhere to Sertoli cells (281, 430-431). The final release of spermatids during the process of spermiation is also sensitive to androgen and/or gonadotropin inhibition, reviewed in (35). Many functions of Sertoli cells are androgen-dependent, such as the maintenance of tight junction function at the blood testis barrier (432-434) and the production of androgen-responsive miRNAs (220), and are necessary to support germ cell development.

The mechanisms by which Sertoli cells support each androgen-dependent phase of germ cell development however, such as the signal required for the completion of meiosis (reviewed in (263)), are as yet unknown. Interestingly, the different androgen-dependent processes within germ cell development have different sensitivities to, or requirements for, androgens, reviewed in (419). For example, the completion of meiosis requires more androgen action than the completion of spermiogenesis (418). Individual variations in the sensitivities of different spermatogenic processes to androgens may explain why a correlation between sperm output and testicular testosterone levels has been so difficult to establish in gonadotropin-suppressed monkeys and men (390, 435-437).

FSH and Spermatogenesis

For many years, the relative roles of androgen vs FSH in initiating, restoring and maintaining spermatogenesis were unclear. This was in part due to the synergistic actions of these two hormones (see below), but also due to difficulties associated with investigating FSH action in a setting of complete androgen ablation. Transgenic mouse models have provided important information regarding specific roles for FSH in spermatogenesis, reviewed in (156, 263, 438). FSH receptors are found only on Sertoli cells and are expressed in a stage-dependent manner (439-440).

One of the most important functions of FSH is to establish a quantitatively normal adult Sertoli cell population. FSH acts as a mitogen for postnatal Sertoli cell proliferation and is required for establishing normal Sertoli cell numbers in mice, reviewed in (156, 204). Since Sertoli cell number determines spermatogenic output in adulthood (101), this function of FSH is important for optimal sperm production. Observations in transgenic mice also show that FSH is needed for normal Sertoli cell morphology and for their ability to support the maximal number of germ cells, e.g. (207-208, 217, 441).

FSH also plays an important role in the regulation of spermatogonia, as revealed in studies in hpg mice (217, 243) and primates (245-246). Numbers of type B spermatogonia correlate more closely with circulating FSH than testicular testosterone levels in gonadotropin-suppressed monkeys and humans (283, 442), indicating that these cells may be particularly supported by FSH. Transgenic human FSH expressed in hpg mice can also exert stimulatory effects on spermatocyte numbers, indicating a permissive effect on meiosis, (243) however FSH alone cannot support the completion of spermiogenesis. The acute suppression of FSH alone can also cause spermiation failure, presumably via effects on the Sertoli cell’s ability to release mature spermatids (218).

 

Optimal Spermatogenesis Requires Synergistic Actions of Androgens and FSH

The data reviewed above indicate that androgens and FSH have distinct roles in spermatogenesis but that these hormones also act co-operatively and synergistically to promote maximal spermatogenic output (156, 203-204, 219).

Androgens and FSH co-operate by supporting different aspects of germ cell development, for example FSH stimulation of spermatogonial populations and androgen stimulation of spermiogenesis. FSH establishes a quantitatively normal Sertoli cell population, whereas androgen initiates and maintains sperm production, thus both hormones co-operate via independent functions to enable maximal spermatogenic output.

Both androgens and FSH facilitate normal Sertoli cell morphology and function, which are likely essential for the ability of Sertoli cells to support the maximum number of germ cells. Both hormones also promote germ cell survival, particularly of spermatocytes and round spermatids in the mid-spermatogenic stages in rodents (264), reviewed in (204). The fact that both hormones can prevent germ cell apoptosis explains why either hormone can maintain germ cell development, at least in the short term, following gonadotropin suppression in humans (209).

There are many examples of synergy between testosterone and FSH, reviewed in (156, 203-204). It has been demonstrated in many experimental settings that testosterone and FSH can support spermatogenesis at a lower dose when the other is present, reviewed in (203). Testosterone and FSH likely act synergistically in the control of signaling pathways and gene expression in Sertoli cells, which in turn are important for germ cell development (156, 219). An example of such synergism is the demonstration that, after acute suppression of either androgen or FSH in rats, approximately 10% of mature spermatids failed to be released at spermiation, whereas suppression of both hormones resulted in 50% of spermatids failing to spermiate (218). Both testosterone and FSH modulate the expression of many miRNA species in Sertoli cells, which likely mediate a large spectrum of proteomic changes important for Sertoli and germ cell function (220).

It should be noted that there are species differences in the response of spermatogenesis to combined androgen and FSH suppression, reviewed in (241, 443). In rodents, suppression of gonadotropins causes a decline in spermatogonial populations but spermatogenesis is primarily arrested at the spermatocyte stage (444). In monkeys and humans however, spermatogenesis is primarily arrested at spermatogonial development, however meiosis and spermiogenesis can be maintained until they undergo a gradual attrition due to the lack of spermatogonia entering meiosis (241, 435, 442).

The requirement for both testosterone and FSH to support normal spermatogenesis in men was revealed in studies by Matsumoto and colleagues (445-446) whereby gonadotropins were suppressed by the administration of testosterone until suppression of spermatogenesis occurred. They then introduced injections of hCG to stimulate Leydig cell function and to restore intratesticular testosterone concentrations which increased sperm counts but not to pre-treatment levels (Figure 12). These data suggested that, in association with undetectable FSH levels, increasing intratesticular androgen could partially restore sperm output (446). Using the same model, they initiated hFSH treatment when sperm counts were suppressed and showed that, in the presence of low intratesticular testosterone concentrations, FSH alone could partially restore sperm output (447). The latter study strongly suggests a role for FSH which appears to be able to synergise with low testosterone to stimulate sperm production in men.

 

Figure 12. The response in the sperm counts from normal volunteers to a suppression of FSH and LH by testosterone injections is shown. Note the recovery in sperm counts when hCG and hFSH were introduced singly into the treatment regime. Data from Matsumoto et. al. (reference 171, 172) and Bremner et. al. (reference 172).

Figure 12. The response in the sperm counts from normal volunteers to a suppression of FSH and LH by testosterone injections is shown. Note the recovery in sperm counts when hCG and hFSH were introduced singly into the treatment regime. Data from Matsumoto et. al. (reference 171, 172) and Bremner et. al. (reference 172).

Considerations for the Stimulation of Sperm Production for Fertility Treatment

 

Male infertility due to undetectable (azoospermia) or low (oligozoospermia) numbers of sperm in the ejaculate may occur in many clinical settings. Details of the approach to the treatment of men with reduced sperm counts are reviewed elsewhere (346, 448-449). Gonadotropic stimulation of sperm production is appropriate in men with gonadotropin deficiency, such as hypogonadotropic hypogonadism (HH) or acquired androgen deficiency, may be of limited benefit in some men with oligospermia (449) but is of no or minimal benefit in men with non-obstructive azoospermia due to primary testicular failure (448) in whom gonadotropic drive is already high.

 

As androgens are essential for the initiation of sperm production, the induction of spermatogenesis in HH acquired after puberty is achieved by the administration of hCG (as an LH substitute), 1000-2000 IU sc 2-3 times per week (449). Prolonged therapy is required to produce sperm in the ejaculate (346, 449), given that human spermatogenesis takes more than 2 months to produce sperm from immature spermatogonia. Treatment with hCG alone may be sufficient for the induction of spermatogenesis in men with larger testes due to potential residual FSH action (346). However, for many men, and particularly for those with congenital HH, the co-administration of FSH (75–150 IU sc 3 times per week) is needed for maximal stimulation of sperm output (346, 449). In men with congenital HH, FSH is needed to induce Sertoli cell maturation, whereas men with acquired HH and smaller testes benefit from the co-administration of FSH due to the well known synergistic actions of FSH and androgens on spermatogenesis as described above. It is also worth nothing that in some men, treatment may need to be particularly protracted (1-2 years) to enable pubertal maturation of the testis, for example the induction of spermatogenesis in Kallmann’s syndrome (449).

 

 

Considerations for the Suppression of Sperm Production for Contraception

As detailed in this chapter, both androgens and FSH co-operate and synergize to stimulate spermatogenesis. In a male hormonal contraceptive context, this means that adequate suppression of both androgens and FSH is required to halt sperm production. The most promising contraceptive strategies in terms of efficacy and rate of sperm count suppression are based on a combination of non-androgenic steroids (e.g. progestins) to suppress gonadotropins, and testosterone to maintain physiological androgen actions outside the testis (see extensive review in Endotext, Endocrinology of Male Reproduction, Chapter 15, Male Contraception (450).

 

The induction of azoospermia is seen as desirable for maximal contraceptive efficacy and acceptability, however no contraceptive regimen as yet is able to consistently induce azoospermia in all men (450). As discussed above, a very narrow dose range exists between testicular testosterone levels and sperm output, meaning that a “little testosterone goes a long way”. In addition, the presence of even low levels of FSH likely potentiates the action of residual androgen on spermatogenesis. In practice, this means that achieving the level of testosterone suppression needed for complete suppression of spermatogenesis may be difficult in some men. A minority of men (~5%) undergoing combined hormone-based therapies fail to achieve adequate sperm count suppression (450). The complete abolition of androgen production does not appear to be achievable because of LH-independent androgen secretion by Leydig cells (423) and the need to maintain extra-testicular androgen actions in men. A complete elimination of androgen action on spermatogenesis could theoretically be achieved via testis-specific enzyme or androgen receptor inhibition, however novel therapeutic tools to achieve this have not yet been identified (see Endotext, Endocrinology of Male Reproduction, Chapter 2, Androgen Physiology, Pharmacology and Abuse (145)).

 

REFERENCES

 

  1. de Kretser D, Temple-Smith P, Kerr J (1982) Anatomical and functional aspects of the male reproductive organs. Handbook of Urology, Vol XVI, Disturbances in Male Fertility. 16: 1-131
  2. Jarow JP (1990) Intratesticular arterial anatomy. J Androl. 11(3): 255-9
  3. Dahl EV, Herrick JF (1959) A vascular mechanism for maintaining testicular temperature by counter-current exchange. Surg Gynecol Obstet. 108(6): 697-705
  4. Clermont Y, Huckins C (1961) Microscopic anatomy of the sex cords and seminiferous tubules in growing and adult male albino rats. Am J Anat 108: 79-97
  5. de Kretser D, Kerr J (1994) The cytology of the testis, in The Physiology of Reproduction, Knobil, E. and Neill, J.D., Editors. Raven Press: New York. p. 1177-1290
  6. Sharpe R (1994) Regulation of spermatogenesis, in The Physiology of Reproduction, Knobil, E. and Neill, J.D., Editors. Raven Press: New York. p. 1363-1434
  7. Russell LD, Griswold MD (1993) The Sertoli Cell. Clearwater, Florida: Cache River Press
  8. Dym M, Fawcett DW (1971) Further observations on the numbers of spermatogonia, spermatocytes, and spermatids connected by intercellular bridges in the mammalian testis. Biol Reprod. 4(2): 195-215
  9. Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH, Nolte J, Wolf F, Li M, Engel W, Hasenfuss G (2006) Pluripotency of spermatogonial stem cells from adult mouse testis. Nature. 440(7088): 1199-203
  10. Conrad S, Renninger M, Hennenlotter J, et al. (2008) Generation of pluripotent stem cells from adult human testis. Nature. 456(7220): 344-9
  11. Meistrich ML, van Beek M (1993) Spermatogonial stem cells. Cell and Molecular Biology of the Testis. 266-295
  12. Clermont Y (1972) Kinetics of spermatogenesis in mammals: seminiferous epithelium cycle and spermatogonial renewal. Physiol Rev. 52(1): 198-236
  13. Kanatsu-Shinohara M, Ogonuki N, Inoue K, Miki H, Ogura A, Toyokuni S, Shinohara T (2003) Long-term proliferation in culture and germline transmission of mouse male germline stem cells. Biol Reprod. 69(2): 612-6
  14. Nagano M, Avarbock MR, Brinster RL (1999) Pattern and kinetics of mouse donor spermatogonial stem cell colonization in recipient testes. Biol Reprod. 60(6): 1429-36
  15. Ogawa T, Dobrinski I, Avarbock MR, Brinster RL (2000) Transplantation of male germ line stem cells restores fertility in infertile mice. Nat Med. 6(1): 29-34
  16. Ogawa T, Dobrinski I, Brinster RL (1999) Recipient preparation is critical for spermatogonial transplantation in the rat. Tissue Cell. 31(5): 461-72
  17. Chan F, Oatley MJ, Kaucher AV, Yang QE, Bieberich CJ, Shashikant CS, Oatley JM (2014) Functional and molecular features of the Id4+ germline stem cell population in mouse testes. Genes Dev. 28(12): 1351-62
  18. Oatley MJ, Kaucher AV, Racicot KE, Oatley JM (2011) Inhibitor of DNA binding 4 is expressed selectively by single spermatogonia in the male germline and regulates the self-renewal of spermatogonial stem cells in mice. Biol Reprod. 85(2): 347-56
  19. Nagano MC, Yeh JR (2013) The identity and fate decision control of spermatogonial stem cells: where is the point of no return? Curr Top Dev Biol. 102: 61-95
  20. Hara K, Nakagawa T, Enomoto H, Suzuki M, Yamamoto M, Simons BD, Yoshida S (2014) Mouse spermatogenic stem cells continually interconvert between equipotent singly isolated and syncytial states. Cell Stem Cell. 14(5): 658-72
  21. Nakagawa T, Sharma M, Nabeshima Y, Braun RE, Yoshida S (2010) Functional hierarchy and reversibility within the murine spermatogenic stem cell compartment. Science. 328(5974): 62-7
  22. Clermont Y (1969) Two classes of spermatogonial stem cells in the monkey (Cercopithecus aethiops). Am J Anat. 126(1): 57-71
  23. van Alphen MM, van de Kant HJ, de Rooij DG (1988) Depletion of the spermatogonia from the seminiferous epithelium of the rhesus monkey after X irradiation. Radiat Res. 113(3): 473-86
  24. Schlatt S, Weinbauer GF (1994) Immunohistochemical localization of proliferating cell nuclear antigen as a tool to study cell proliferation in rodent and primate testes. Int J Androl. 17(4): 214-22
  25. Schulze C (1979) Morphological characteristics of the spermatogonial stem cells in man. Cell Tissue Res. 198(2): 191-9
  26. Plant TM (2010) Undifferentiated primate spermatogonia and their endocrine control. Trends Endocrinol Metab. 21(8): 488-95
  27. Ramaswamy S, Razack BS, Roslund RM, Suzuki H, Marshall GR, Rajkovic A, Plant TM (2014) Spermatogonial SOHLH1 nucleocytoplasmic shuttling associates with initiation of spermatogenesis in the rhesus monkey (Macaca mulatta). Mol Hum Reprod. 20(4): 350-7
  28. Gassei K, Ehmcke J, Dhir R, Schlatt S (2010) Magnetic activated cell sorting allows isolation of spermatogonia from adult primate testes and reveals distinct GFRa1-positive subpopulations in men. J Med Primatol. 39(2): 83-91
  29. Shinohara T, Orwig KE, Avarbock MR, Brinster RL (2000) Spermatogonial stem cell enrichment by multiparameter selection of mouse testis cells. Proc Natl Acad Sci U S A. 97(15): 8346-51
  30. Stubbs L, Stern H (1986) DNA synthesis at selective sites during pachytene in mouse spermatocytes. Chromosoma. 93(6): 529-36
  31. Heller CH, Clermont Y (1964) Kinetics of the Germinal Epithelium in Man. Recent Prog Horm Res. 20: 545-75
  32. Vrooman LA, Nagaoka SI, Hassold TJ, Hunt PA (2014) Evidence for paternal age-related alterations in meiotic chromosome dynamics in the mouse. Genetics. 196(2): 385-96
  33. De Kretser DM (1969) Ultrastructural features of human spermiogenesis. Z Zellforsch Mikrosk Anat. 98(4): 477-505
  34. Eddy EM (1999) Role of heat shock protein HSP70-2 in spermatogenesis. Rev Reprod. 4(1): 23-30
  35. O'Donnell L, Nicholls PK, O'Bryan MK, McLachlan RI, Stanton PG (2011) Spermiation: The process of sperm release. Spermatogenesis. 1(1): 14-35
  36. Hermo L, Pelletier RM, Cyr DG, Smith CE (2010) Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 2: changes in spermatid organelles associated with development of spermatozoa. Microsc Res Tech. 73(4): 279-319
  37. Hermo L, Pelletier RM, Cyr DG, Smith CE (2010) Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 3: developmental changes in spermatid flagellum and cytoplasmic droplet and interaction of sperm with the zona pellucida and egg plasma membrane. Microsc Res Tech. 73(4): 320-63
  38. Oko RJ, Jando V, Wagner CL, Kistler WS, Hermo LS (1996) Chromatin reorganization in rat spermatids during the disappearance of testis-specific histone, H1t, and the appearance of transition proteins TP1 and TP2. Biol Reprod. 54(5): 1141-57
  39. Steger K, Klonisch T, Gavenis K, Drabent B, Doenecke D, Bergmann M (1998) Expression of mRNA and protein of nucleoproteins during human spermiogenesis. Mol Hum Reprod. 4(10): 939-45
  40. Eddy EM (1998) Regulation of gene expression during spermatogenesis. Semin Cell Dev Biol. 9(4): 451-7
  41. Russell LD, Russell JA, MacGregor GR, Meistrich ML (1991) Linkage of manchette microtubules to the nuclear envelope and observations of the role of the manchette in nuclear shaping during spermiogenesis in rodents. Am J Anat. 192(2): 97-120
  42. Kierszenbaum AL, Tres LL (2004) The acrosome-acroplaxome-manchette complex and the shaping of the spermatid head. Arch Histol Cytol. 67(4): 271-84
  43. O'Donnell L, O'Bryan MK (2014) Microtubules and spermatogenesis. Semin Cell Dev Biol. 30: 45-54
  44. Fawcett DW (1975) The mammalian spermatozoon. Dev Biol. 44(2): 394-436
  45. Kierszenbaum AL (2002) Intramanchette transport (IMT): managing the making of the spermatid head, centrosome, and tail. Mol Reprod Dev. 63(1): 1-4
  46. Carrera A, Gerton GL, Moss SB (1994) The major fibrous sheath polypeptide of mouse sperm: structural and functional similarities to the A-kinase anchoring proteins. Dev Biol. 165(1): 272-84
  47. Fulcher KD, Mori C, Welch JE, O'Brien DA, Klapper DG, Eddy EM (1995) Characterization of Fsc1 cDNA for a mouse sperm fibrous sheath component. Biol Reprod. 52(1): 41-9
  48. Mandal A, Naaby-Hansen S, Wolkowicz MJ, et al. (1999) FSP95, a testis-specific 95-kilodalton fibrous sheath antigen that undergoes tyrosine phosphorylation in capacitated human spermatozoa. Biol Reprod. 61(5): 1184-97
  49. Mei X, Singh IS, Erlichman J, Orr GA (1997) Cloning and characterization of a testis-specific, developmentally regulated A-kinase-anchoring protein (TAKAP-80) present on the fibrous sheath of rat sperm. Eur J Biochem. 246(2): 425-32
  50. Miki K, Eddy EM (1998) Identification of tethering domains for protein kinase A type Ialpha regulatory subunits on sperm fibrous sheath protein FSC1. J Biol Chem. 273(51): 34384-90
  51. Vijayaraghavan S, Liberty GA, Mohan J, Winfrey VP, Olson GE, Carr DW (1999) Isolation and molecular characterization of AKAP110, a novel, sperm-specific protein kinase A-anchoring protein. Mol Endocrinol. 13(5): 705-17
  52. Kirichok Y, Navarro B, Clapham DE (2006) Whole-cell patch-clamp measurements of spermatozoa reveal an alkaline-activated Ca2+ channel. Nature. 439(7077): 737-40
  53. Strunker T, Goodwin N, Brenker C, Kashikar ND, Weyand I, Seifert R, Kaupp UB (2011) The CatSper channel mediates progesterone-induced Ca2+ influx in human sperm. Nature. 471(7338): 382-6
  54. Lishko PV, Botchkina IL, Kirichok Y (2011) Progesterone activates the principal Ca2+ channel of human sperm. Nature. 471(7338): 387-91
  55. Okunade GW, Miller ML, Pyne GJ, et al. (2004) Targeted ablation of plasma membrane Ca2+-ATPase (PMCA) 1 and 4 indicates a major housekeeping function for PMCA1 and a critical role in hyperactivated sperm motility and male fertility for PMCA4. J Biol Chem. 279(32): 33742-50
  56. O'Bryan MK, Sebire K, Meinhardt A, Edgar K, Keah HH, Hearn MT, De Kretser DM (2001) Tpx-1 is a component of the outer dense fibers and acrosome of rat spermatozoa. Mol Reprod Dev. 58(1): 116-25
  57. Gibbs GM, Scanlon MJ, Swarbrick J, Curtis S, Gallant E, Dulhunty AF, O'Bryan MK (2006) The cysteine-rich secretory protein domain of Tpx-1 is related to ion channel toxins and regulates ryanodine receptor Ca2+ signaling. J Biol Chem. 281(7): 4156-63
  58. Holstein AF (1976) Ultrastructural observations on the differentiation of spermatids in man. Andrologia. 8(2): 157-65
  59. Russell LD (1991) The perils of sperm release-- 'let my children go'. Int J Androl. 14(5): 307-11
  60. Russell L (1993) Role in spermiation, in The Sertoli cell, Russell, L.D. and Griswold, M.D., Editors. Cache River Press: Clearwater, FL. p. 269-302
  61. O'Donnell L (2014) Mechanisms of spermiogenesis and spermiation and how they are disturbed. Spermatogenesis. 4(2): e979623
  62. Leblond CP, Clermont Y (1952) Definition of the stages of the cycle of the seminiferous epithelium in the rat. Ann N Y Acad Sci. 55(4): 548-73
  63. Parvinen M (1982) Regulation of the seminiferous epithelium. Endocr Rev. 3(4): 404-17
  64. Perey B, Clermont Y, LeBlond CP (1961) The wave of the seminiferous epithelium in the rat. . Am J Anat 108: 47-77
  65. Regaud C (1901) Études sur la structure des tubes seminiferes et sur la spermatogenese chez les mammiferes. . Arch Anat Microsc 4: 101-156
  66. Clermont Y (1963) The cycle of the seminiferous epithelium in man. Am J Anat. 112: 35-51
  67. Johnston DS, Wright WW, Dicandeloro P, Wilson E, Kopf GS, Jelinsky SA (2008) Stage-specific gene expression is a fundamental characteristic of rat spermatogenic cells and Sertoli cells. Proc Natl Acad Sci U S A. 105(24): 8315-20
  68. Clouthier DE, Avarbock MR, Maika SD, Hammer RE, Brinster RL (1996) Rat spermatogenesis in mouse testis. Nature. 381(6581): 418-21
  69. Timmons PM, Rigby PW, Poirier F (2002) The murine seminiferous epithelial cycle is pre-figured in the Sertoli cells of the embryonic testis. Development. 129(3): 635-47
  70. Sugimoto R, Nabeshima Y, Yoshida S (2012) Retinoic acid metabolism links the periodical differentiation of germ cells with the cycle of Sertoli cells in mouse seminiferous epithelium. Mech Dev. 128(11-12): 610-24
  71. Fawcett D (1975) Ultrastructure and function of the Sertoli cell. Handbook of Physiology, Section 7, Endocrinology. Vol 5, Male Reproductive System: 21-55
  72. Vogl AW (1988) Changes in the distribution of microtubules in rat Sertoli cells during spermatogenesis. Anat Rec. 222(1): 34-41
  73. Dym M, Fawcett DW (1970) The blood-testis barrier in the rat and the physiological compartmentation of the seminiferous epithelium. Biol Reprod. 3(3): 308-26
  74. Setchell BP, Waites GM (1970) Changes in the permeability of the testicular capillaries and of the 'blood-testis barrier' after injection of cadmium chloride in the rat. J Endocrinol. 47(1): 81-6
  75. Meng J, Greenlee AR, Taub CJ, Braun RE (2011) Sertoli cell-specific deletion of the androgen receptor compromises testicular immune privilege in mice. Biol Reprod. 85(2): 254-60
  76. McCabe MJ, Allan CM, Foo CF, Nicholls PK, McTavish KJ, Stanton PG (2012) Androgen Initiates Sertoli Cell Tight Junction Formation in the Hypogonadal (hpg) Mouse. Biol Reprod.
  77. Yan HH, Mruk DD, Cheng CY (2008) Junction restructuring and spermatogenesis: the biology, regulation, and implication in male contraceptive development. Curr Top Dev Biol. 80: 57-92
  78. Mruk DD, Cheng CY (2015) The Mammalian Blood-Testis Barrier: Its Biology and Regulation. Endocr Rev. 36(5): 564-91
  79. Gow A, Southwood CM, Li JS, Pariali M, Riordan GP, Brodie SE, Danias J, Bronstein JM, Kachar B, Lazzarini RA (1999) CNS myelin and sertoli cell tight junction strands are absent in Osp/claudin-11 null mice. Cell. 99(6): 649-59
  80. Hall PF, Mita M (1984) Influence of follicle-stimulating hormone on glucose transport by cultured Sertoli cells. Biol Reprod. 31(5): 863-9
  81. Jutte NH, Jansen R, Grootegoed JA, Rommerts FF, van der Molen HJ (1983) FSH stimulation of the production of pyruvate and lactate by rat Sertoli cells may be involved in hormonal regulation of spermatogenesis. J Reprod Fertil. 68(1): 219-26
  82. Robinson R, Fritz IB (1979) Myoinositol biosynthesis by Sertoli cells, and levels of myoinositol biosynthetic enzymes in testis and epididymis. Can J Biochem. 57(6): 962-7
  83. Kaur G, Thompson LA, Dufour JM (2014) Sertoli cells--immunological sentinels of spermatogenesis. Semin Cell Dev Biol. 30: 36-44
  84. Rebourcet D, O'Shaughnessy PJ, Monteiro A, Milne L, Cruickshanks L, Jeffrey N, Guillou F, Freeman TC, Mitchell RT, Smith LB (2014) Sertoli cells maintain Leydig cell number and peritubular myoid cell activity in the adult mouse testis. PLoS One. 9(8): e105687
  85. Hedger MP, Winnall WR (2012) Regulation of activin and inhibin in the adult testis and the evidence for functional roles in spermatogenesis and immunoregulation. Mol Cell Endocrinol. 359(1-2): 30-42
  86. Nicholls PK, Stanton PG, Chen JL, Olcorn JS, Haverfield JT, Qian H, Walton KL, Gregorevic P, Harrison CA (2012) Activin signaling regulates Sertoli cell differentiation and function. Endocrinology. 153(12): 6065-77
  87. Haverfield JT, Meachem SJ, Nicholls PK, Rainczuk KE, Simpson ER, Stanton PG (2014) Differential permeability of the blood-testis barrier during reinitiation of spermatogenesis in adult male rats. Endocrinology. 155(3): 1131-44
  88. Yan W (2015) Gene knockouts that affect Sertoli cell function, in Sertoli cell biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 437-469
  89. Chen C, Ouyang W, Grigura V, et al. (2005) ERM is required for transcriptional control of the spermatogonial stem cell niche. Nature. 436(7053): 1030-4
  90. Phillips BT, Gassei K, Orwig KE (2010) Spermatogonial stem cell regulation and spermatogenesis. Philos Trans R Soc Lond B Biol Sci. 365(1546): 1663-78
  91. Simorangkir DR, de Kretser DM, Wreford NG (1995) Increased numbers of Sertoli and germ cells in adult rat testes induced by synergistic action of transient neonatal hypothyroidism and neonatal hemicastration. J Reprod Fertil. 104(2): 207-13
  92. Simorangkir DR, Wreford NG, De Kretser DM (1997) Impaired germ cell development in the testes of immature rats with neonatal hypothyroidism. J Androl. 18(2): 186-93
  93. Haverfield JT, Stanton PG, Meachem SJ (2015) Adult Sertoli cell differentiation status in humans., in Sertoli cell biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 81-98
  94. Mazaud-Guittot S, Meugnier E, Pesenti S, Wu X, Vidal H, Gow A, Le Magueresse-Battistoni B (2010) Claudin 11 deficiency in mice results in loss of the Sertoli cell epithelial phenotype in the testis. Biol Reprod. 82(1): 202-13
  95. Tarulli GA, Stanton PG, Loveland KL, Meyts ER, McLachlan RI, Meachem SJ (2013) A survey of Sertoli cell differentiation in men after gonadotropin suppression and in testicular cancer. Spermatogenesis. 3(1): e24014
  96. Matson CK, Murphy MW, Sarver AL, Griswold MD, Bardwell VJ, Zarkower D (2011) DMRT1 prevents female reprogramming in the postnatal mammalian testis. Nature. 476(7358): 101-4
  97. Rebourcet D, O'Shaughnessy PJ, Pitetti JL, et al. (2014) Sertoli cells control peritubular myoid cell fate and support adult Leydig cell development in the prepubertal testis. Development. 141(10): 2139-49
  98. Cortes D, Muller J, Skakkebaek NE (1987) Proliferation of Sertoli cells during development of the human testis assessed by stereological methods. Int J Androl. 10(4): 589-96
  99. Yang Q-E, Oatley JM (2015) Early postnatal interactions between Sertoli and germ cells, in Sertoli cell biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 81-98
  100. Bagheri-Fam S, Argentaro A, Svingen T, Combes AN, Sinclair AH, Koopman P, Harley VR (2011) Defective survival of proliferating Sertoli cells and androgen receptor function in a mouse model of the ATR-X syndrome. Hum Mol Genet. 20(11): 2213-24
  101. Petersen C, Soder O (2006) The Sertoli cell--a hormonal target and 'super' nurse for germ cells that determines testicular size. Horm Res. 66(4): 153-61
  102. Cooke PS, Hess RA, Porcelli J, Meisami E (1991) Increased sperm production in adult rats after transient neonatal hypothyroidism. Endocrinology. 129(1): 244-8
  103. Boitani C, Stefanini M, Fragale A, Morena AR (1995) Activin stimulates Sertoli cell proliferation in a defined period of rat testis development. Endocrinology. 136(12): 5438-44
  104. Meehan T, Schlatt S, O'Bryan MK, de Kretser DM, Loveland KL (2000) Regulation of germ cell and Sertoli cell development by activin, follistatin, and FSH. Dev Biol. 220(2): 225-37
  105. Sheckter CB, McLachlan RI, Tenover JS, Matsumoto AM, Burger HG, de Kretser DM, Bremner WJ (1988) Stimulation of serum inhibin concentrations by gonadotropin-releasing hormone in men with idiopathic hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 67(6): 1221-4
  106. Christensen A (1975) Leydig cells, in Handbook of Physiology, Section 7, Endocrinology. p. 57-94
  107. Fawcett DW, Leak LV, Heidger PM, Jr. (1970) Electron microscopic observations on the structural components of the blood-testis barrier. J Reprod Fertil Suppl. 10: 105-22
  108. Miller SC (1982) Localization of plutonium-241 in the testis. An interspecies comparison using light and electron microscope autoradiography. Int J Radiat Biol Relat Stud Phys Chem Med. 41(6): 633-43
  109. Miller SC, Bowman BM, Rowland HG (1983) Structure, cytochemistry, endocytic activity, and immunoglobulin (Fc) receptors of rat testicular interstitial-tissue macrophages. Am J Anat. 168(1): 1-13
  110. Martin LJ (2016) Cell interactions and genetic regulation that contribute to testicular Leydig cell development and differentiation. Mol Reprod Dev. 83(6): 470-87
  111. Teerds KJ, Huhtaniemi IT (2015) Morphological and functional maturation of Leydig cells: from rodent models to primates. Hum Reprod Update. 21(3): 310-28
  112. Lording DW, De Kretser DM (1972) Comparative ultrastructural and histochemical studies of the interstitial cells of the rat testis during fetal and postnatal development. J Reprod Fertil. 29(2): 261-9
  113. Pelliniemi LJ, Niemi M (1969) Fine structure of the human foetal testis. I. The interstitial tissue. Z Zellforsch Mikrosk Anat. 99(4): 507-22
  114. Haider SG (2004) Cell biology of Leydig cells in the testis. Int Rev Cytol. 233: 181-241
  115. Wen Q, Cheng CY, Liu YX (2016) Development, function and fate of fetal Leydig cells. Semin Cell Dev Biol. 59: 89-98
  116. Huhtaniemi I (1977) Studies on steroidogenesis and its regulation in human fetal adrenal and testis. J Steroid Biochem. 8(5): 491-7
  117. Prince FP (1990) Ultrastructural evidence of mature Leydig cells and Leydig cell regression in the neonatal human testis. Anat Rec. 228(4): 405-17
  118. Shima Y, Matsuzaki S, Miyabayashi K, Otake H, Baba T, Kato S, Huhtaniemi I, Morohashi K (2015) Fetal Leydig Cells Persist as an Androgen-Independent Subpopulation in the Postnatal Testis. Mol Endocrinol. 29(11): 1581-93
  119. Christensen AK, Peacock KC (1980) Increase in Leydig cell number in testes of adult rats treated chronically with an excess of human chorionic gonadotropin. Biol Reprod. 22(2): 383-91
  120. Prince FP (2001) The triphasic nature of Leydig cell development in humans, and comments on nomenclature. J Endocrinol. 168(2): 213-6
  121. O'Shaughnessy PJ, Baker PJ, Johnston H (2006) The foetal Leydig cell-- differentiation, function and regulation. Int J Androl. 29(1): 90-5; discussion 105-8
  122. Bitgood MJ, Shen L, McMahon AP (1996) Sertoli cell signaling by Desert hedgehog regulates the male germline. Curr Biol. 6(3): 298-304
  123. Brokken LJ, Adamsson A, Paranko J, Toppari J (2009) Antiandrogen exposure in utero disrupts expression of desert hedgehog and insulin-like factor 3 in the developing fetal rat testis. Endocrinology. 150(1): 445-51
  124. Clark AM, Garland KK, Russell LD (2000) Desert hedgehog (Dhh) gene is required in the mouse testis for formation of adult-type Leydig cells and normal development of peritubular cells and seminiferous tubules. Biol Reprod. 63(6): 1825-38
  125. Li L, Wang Y, Li X, Liu S, Wang G, Lin H, Zhu Q, Guo J, Chen H, Ge HS, Ge RS (2016) Regulation of development of rat stem and progenitor Leydig cells by activin. Andrology.
  126. Odeh HM, Kleinguetl C, Ge R, Zirkin BR, Chen H (2014) Regulation of the proliferation and differentiation of Leydig stem cells in the adult testis. Biol Reprod. 90(6): 123
  127. Pierucci-Alves F, Clark AM, Russell LD (2001) A developmental study of the Desert hedgehog-null mouse testis. Biol Reprod. 65(5): 1392-402
  128. Canto P, Soderlund D, Reyes E, Mendez JP (2004) Mutations in the desert hedgehog (DHH) gene in patients with 46,XY complete pure gonadal dysgenesis. J Clin Endocrinol Metab. 89(9): 4480-3
  129. Wen Q, Zheng QS, Li XX, Hu ZY, Gao F, Cheng CY, Liu YX (2014) Wt1 dictates the fate of fetal and adult Leydig cells during development in the mouse testis. Am J Physiol Endocrinol Metab. 307(12): E1131-43
  130. Umehara T, Kawashima I, Kawai T, Hoshino Y, Morohashi KI, Shima Y, Zeng W, Richards JS, Shimada M (2016) Neuregulin 1 Regulates Proliferation of Leydig Cells to Support Spermatogenesis and Sexual Behavior in Adult Mice. Endocrinology. 157(12): 4899-4913
  131. Payne AH, Hardy MP, Russell LD (1996) The Leydig Cell Illinois: Cache River Press. 1-802
  132. Foresta C, Bettella A, Vinanzi C, Dabrilli P, Meriggiola MC, Garolla A, Ferlin A (2004) A novel circulating hormone of testis origin in humans. J Clin Endocrinol Metab. 89(12): 5952-8
  133. Zimmermann S, Steding G, Emmen JM, Brinkmann AO, Nayernia K, Holstein AF, Engel W, Adham IM (1999) Targeted disruption of the Insl3 gene causes bilateral cryptorchidism. Mol Endocrinol. 13(5): 681-91
  134. Ivell R, Wade JD, Anand-Ivell R (2013) INSL3 as a biomarker of Leydig cell functionality. Biol Reprod. 88(6): 147
  135. Anand-Ivell RJ, Relan V, Balvers M, Coiffec-Dorval I, Fritsch M, Bathgate RA, Ivell R (2006) Expression of the insulin-like peptide 3 (INSL3) hormone-receptor (LGR8) system in the testis. Biol Reprod. 74(5): 945-53
  136. Kawamura K, Kumagai J, Sudo S, Chun SY, Pisarska M, Morita H, Toppari J, Fu P, Wade JD, Bathgate RA, Hsueh AJ (2004) Paracrine regulation of mammalian oocyte maturation and male germ cell survival. Proc Natl Acad Sci U S A. 101(19): 7323-8
  137. Yuan FP, Li X, Lin J, Schwabe C, Bullesbach EE, Rao CV, Lei ZM (2010) The role of RXFP2 in mediating androgen-induced inguinoscrotal testis descent in LH receptor knockout mice. Reproduction. 139(4): 759-69
  138. Pathirana IN, Kawate N, Bullesbach EE, Takahashi M, Hatoya S, Inaba T, Tamada H (2012) Insulin-like peptide 3 stimulates testosterone secretion in mouse Leydig cells via cAMP pathway. Regul Pept. 178(1-3): 102-6
  139. Johansen ML, Anand-Ivell R, Mouritsen A, Hagen CP, Mieritz MG, Soeborg T, Johannsen TH, Main KM, Andersson AM, Ivell R, Juul A (2014) Serum levels of insulin-like factor 3, anti-Mullerian hormone, inhibin B, and testosterone during pubertal transition in healthy boys: a longitudinal pilot study. Reproduction. 147(4): 529-35
  140. Trabado S, Maione L, Bry-Gauillard H, et al. (2014) Insulin-like peptide 3 (INSL3) in men with congenital hypogonadotropic hypogonadism/Kallmann syndrome and effects of different modalities of hormonal treatment: a single-center study of 281 patients. J Clin Endocrinol Metab. 99(2): E268-75
  141. Rohayem J, Fricke R, Czeloth K, Mallidis C, Wistuba J, Krallmann C, Zitzmann M, Kliesch S (2015) Age and markers of Leydig cell function, but not of Sertoli cell function predict the success of sperm retrieval in adolescents and adults with Klinefelter's syndrome. Andrology. 3(5): 868-75
  142. Stanley E, Lin CY, Jin S, Liu J, Sottas CM, Ge R, Zirkin BR, Chen H (2012) Identification, Proliferation, and Differentiation of Adult Leydig Stem Cells. Endocrinology. DOI 10.1210/en.2012-1417
  143. Baird DT, Galbraith A, Fraser IS, Newsam JE (1973) The concentration of oestrone and oestradiol-17 in spermatic venous blood in man. J Endocrinol. 57(2): 285-8
  144. Rochira V, Madeo B, Diazzi C, Zirilli L, Santi D, Carani C. (2016) Estrogens and Male Reproduction, in www.ENDOTEXT.org, Endocrinology of Male Reproduction, Section Editor McLachlan R.I. MDTEXT.COM,INC, : South Dartmouth,MA 02748.
  145. Handelsman DJ. (2016) Androgen Physiology, Pharmacology and Abuse in www.ENDOTEXT.org, Endocrinology of Male Reproduction, Section Editor McLachlan R.I. MDTEXT.COM,INC, : South Dartmouth,MA 02748.
  146. Hodgson YM, de Kretser DM (1984) Acute responses of Leydig cells to hCG: evidence for early hypertrophy of Leydig cells. Mol Cell Endocrinol. 35(2-3): 75-82
  147. Waterman MR, Simpson ER (1989) Regulation of steroid hydroxylase gene expression is multifactorial in nature. Recent Prog Horm Res. 45: 533-63; discussion 563-6
  148. Wu FC, Irby DC, Clarke IJ, Cummins JT, de Kretser DM (1987) Effects of gonadotropin-releasing hormone pulse-frequency modulation on luteinizing hormone, follicle-stimulating hormone and testosterone secretion in hypothalamo/pituitary-disconnected rams. Biol Reprod. 37(3): 501-10
  149. Beattie MC, Adekola L, Papadopoulos V, Chen H, Zirkin BR (2015) Leydig cell aging and hypogonadism. Exp Gerontol. 68: 87-91
  150. Veldhuis JD, Liu PY, Keenan DM, Takahashi PY (2012) Older men exhibit reduced efficacy of and heightened potency downregulation by intravenous pulses of recombinant human LH: a study in 92 healthy men. Am J Physiol Endocrinol Metab. 302(1): E117-22
  151. Oury F, Sumara G, Sumara O, Ferron M, Chang H, Smith CE, Hermo L, Suarez S, Roth BL, Ducy P, Karsenty G (2011) Endocrine regulation of male fertility by the skeleton. Cell. 144(5): 796-809
  152. Karsenty G (2012) The mutual dependence between bone and gonads. J Endocrinol. 213(2): 107-14
  153. Karsenty G, Oury F (2012) Biology without walls: the novel endocrinology of bone. Annu Rev Physiol. 74: 87-105
  154. Aoki A, Fawcett DW (1978) Is there a local feedback from the seminiferous tubules affecting activity of the Leydig cells? Biol Reprod. 19(1): 144-58
  155. de Kretser DM (1987) Local regulation of testicular function. Int Rev Cytol. 109: 89-112
  156. O'Shaughnessy PJ, Morris ID, Huhtaniemi I, Baker PJ, Abel MH (2009) Role of androgen and gonadotrophins in the development and function of the Sertoli cells and Leydig cells: data from mutant and genetically modified mice. Mol Cell Endocrinol. 306(1-2): 2-8
  157. De Gendt K, Atanassova N, Tan KA, et al. (2005) Development and function of the adult generation of Leydig cells in mice with Sertoli cell-selective or total ablation of the androgen receptor. Endocrinology. 146(9): 4117-26
  158. Hazra R, Jimenez M, Desai R, Handelsman DJ, Allan CM (2013) Sertoli cell androgen receptor expression regulates temporal fetal and adult Leydig cell differentiation, function, and population size. Endocrinology. 154(9): 3410-22
  159. O'Shaughnessy PJ, Hu L, Baker PJ (2008) Effect of germ cell depletion on levels of specific mRNA transcripts in mouse Sertoli cells and Leydig cells. Reproduction. 135(6): 839-50
  160. Jegou B, Laws AO, de Kretser DM (1984) Changes in testicular function induced by short-term exposure of the rat testis to heat: further evidence for interaction of germ cells, Sertoli cells and Leydig cells. Int J Androl. 7(3): 244-57
  161. Lue Y, Hikim AP, Wang C, Im M, Leung A, Swerdloff RS (2000) Testicular heat exposure enhances the suppression of spermatogenesis by testosterone in rats: the "two-hit" approach to male contraceptive development. Endocrinology. 141(4): 1414-24
  162. Smith LB, O'Shaughnessy PJ, Rebourcet D (2015) Cell-specific ablation in the testis: what have we learned? Andrology. 3(6): 1035-49
  163. Risbridger GP, Kerr JB, de Kretser DM (1981) Evaluation of Leydig cell function and gonadotropin binding in unilateral and bilateral cryptorchidism; evidence for local control of Leydig cell function by the seminiferous tubule. Biol Reprod. 24(3): 534-40
  164. Risbridger GP, Kerr JB, Peake RA, de Kretser DM (1981) An assessment of Leydig cell function after bilateral or unilateral efferent duct ligation: further evidence for local control of Leydig cell function. Endocrinology. 109(4): 1234-41
  165. Andersson AM, Jorgensen N, Frydelund-Larsen L, Rajpert-De Meyts E, Skakkebaek NE (2004) Impaired Leydig cell function in infertile men: a study of 357 idiopathic infertile men and 318 proven fertile controls. J Clin Endocrinol Metab. 89(7): 3161-7
  166. van den Driesche S, Kolovos P, Platts S, Drake AJ, Sharpe RM (2012) Inter-relationship between testicular dysgenesis and Leydig cell function in the masculinization programming window in the rat. PLoS One. 7(1): e30111
  167. Hales DB (2002) Testicular macrophage modulation of Leydig cell steroidogenesis. J Reprod Immunol. 57(1-2): 3-18
  168. Welsh M, Moffat L, Belling K, de Franca LR, Segatelli TM, Saunders PT, Sharpe RM, Smith LB (2012) Androgen receptor signalling in peritubular myoid cells is essential for normal differentiation and function of adult Leydig cells. Int J Androl. 35(1): 25-40
  169. Clermont Y (1958) Contractile elements in the limiting membrane of the seminiferous tubules of the rat. Exp Cell Res. 15(2): 438-40
  170. Ross MH, Long IR (1966) Contractile cells in human seminiferous tubules. Science. 153(3741): 1271-3
  171. Maekawa M, Kamimura K, Nagano T (1996) Peritubular myoid cells in the testis: their structure and function. Arch Histol Cytol. 59(1): 1-13
  172. Rossi F, Ferraresi A, Romagni P, Silvestroni L, Santiemma V (2002) Angiotensin II stimulates contraction and growth of testicular peritubular myoid cells in vitro. Endocrinology. 143(8): 3096-104
  173. Tripiciano A, Filippini A, Ballarini F, Palombi F (1998) Contractile response of peritubular myoid cells to prostaglandin F2alpha. Mol Cell Endocrinol. 138(1-2): 143-50
  174. Tripiciano A, Filippini A, Giustiniani Q, Palombi F (1996) Direct visualization of rat peritubular myoid cell contraction in response to endothelin. Biol Reprod. 55(1): 25-31
  175. Losinno AD, Sorrivas V, Ezquer M, Ezquer F, Lopez LA, Morales A (2016) Changes of myoid and endothelial cells in the peritubular wall during contraction of the seminiferous tubule. Cell Tissue Res. 365(2): 425-35
  176. de Winter JP, Vanderstichele HM, Verhoeven G, Timmerman MA, Wesseling JG, de Jong FH (1994) Peritubular myoid cells from immature rat testes secrete activin-A and express activin receptor type II in vitro. Endocrinology. 135(2): 759-67
  177. Gnessi L, Emidi A, Jannini EA, Carosa E, Maroder M, Arizzi M, Ulisse S, Spera G (1995) Testicular development involves the spatiotemporal control of PDGFs and PDGF receptors gene expression and action. J Cell Biol. 131(4): 1105-21
  178. Verhoeven G, Hoeben E, De Gendt K (2000) Peritubular cell-Sertoli cell interactions: factors involved in PmodS activity. Andrologia. 32(1): 42-5
  179. Welsh M, Saunders PT, Atanassova N, Sharpe RM, Smith LB (2009) Androgen action via testicular peritubular myoid cells is essential for male fertility. FASEB J. 23(12): 4218-30
  180. Qian Y, Liu S, Guan Y, et al. (2013) Lgr4-mediated Wnt/beta-catenin signaling in peritubular myoid cells is essential for spermatogenesis. Development. 140(8): 1751-61
  181. Chen LY, Brown PR, Willis WB, Eddy EM (2014) Peritubular myoid cells participate in male mouse spermatogonial stem cell maintenance. Endocrinology. 155(12): 4964-74
  182. Chen LY, Willis WD, Eddy EM (2016) Targeting the Gdnf Gene in peritubular myoid cells disrupts undifferentiated spermatogonial cell development. Proc Natl Acad Sci U S A. 113(7): 1829-34
  183. Griswold MD (2015) ed.^eds. Sertoli cell biology. 2nd ed. Elsevier: Waltham, MA
  184. Lin YT, Capel B (2015) Cell fate commitment during mammalian sex determination. Curr Opin Genet Dev. 32: 144-52
  185. Yao HH, Ungewitter E, Franco H, Capel B (2015) Establishment of fetal Sertoli cells and their role in testis morphogenesis, in Sertoli cell biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 57-80
  186. Ohta K, Yamamoto M, Lin Y, Hogg N, Akiyama H, Behringer RR, Yamazaki Y (2012) Male differentiation of germ cells induced by embryonic age-specific Sertoli cells in mice. Biol Reprod. 86(4): 112
  187. Van Haaster LH, De Jong FH, Docter R, De Rooij DG (1992) The effect of hypothyroidism on Sertoli cell proliferation and differentiation and hormone levels during testicular development in the rat. Endocrinology. 131(3): 1574-6
  188. Hazra R, Corcoran L, Robson M, McTavish KJ, Upton D, Handelsman DJ, Allan CM (2013) Temporal role of Sertoli cell androgen receptor expression in spermatogenic development. Mol Endocrinol. 27(1): 12-24
  189. Meachem SJ, McLachlan RI, de Kretser DM, Robertson DM, Wreford NG (1996) Neonatal exposure of rats to recombinant follicle stimulating hormone increases adult Sertoli and spermatogenic cell numbers. Biol Reprod. 54(1): 36-44
  190. Fahrioglu U, Murphy MW, Zarkower D, Bardwell VJ (2007) mRNA expression analysis and the molecular basis of neonatal testis defects in Dmrt1 mutant mice. Sex Dev. 1(1): 42-58
  191. Welborn JP, Davis MG, Ebers SD, Stodden GR, Hayashi K, Cheatwood JL, Rao MK, MacLean JA, 2nd (2015) Rhox8 Ablation in the Sertoli Cells Using a Tissue-Specific RNAi Approach Results in Impaired Male Fertility in Mice. Biol Reprod. 93(1): 8
  192. Hess RA, Vogl AW (2015) Sertoli cell anatomy and cytoskeleton, in Sertoli cell biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 1-56
  193. Zimmermann C, Stevant I, Borel C, Conne B, Pitetti JL, Calvel P, Kaessmann H, Jegou B, Chalmel F, Nef S (2015) Research resource: the dynamic transcriptional profile of sertoli cells during the progression of spermatogenesis. Mol Endocrinol. 29(4): 627-42
  194. Hogarth CA (2015) Retinoic acid metabolism, signalling and function in the adult testis, in Sertoli cell biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 247-272
  195. Vernet N, Dennefeld C, Rochette-Egly C, Oulad-Abdelghani M, Chambon P, Ghyselinck NB, Mark M (2006) Retinoic acid metabolism and signaling pathways in the adult and developing mouse testis. Endocrinology. 147(1): 96-110
  196. Hogarth CA, Arnold S, Kent T, Mitchell D, Isoherranen N, Griswold MD (2015) Processive pulses of retinoic acid propel asynchronous and continuous murine sperm production. Biol Reprod. 92(2): 37
  197. Kent T, Arnold SL, Fasnacht R, Rowsey R, Mitchell D, Hogarth CA, Isoherranen N, Griswold MD (2016) ALDH Enzyme Expression Is Independent of the Spermatogenic Cycle, and Their Inhibition Causes Misregulation of Murine Spermatogenic Processes. Biol Reprod. 94(1): 12
  198. Vernet N, Dennefeld C, Klopfenstein M, Ruiz A, Bok D, Ghyselinck NB, Mark M (2008) Retinoid X receptor beta (RXRB) expression in Sertoli cells controls cholesterol homeostasis and spermiation. Reproduction. 136(5): 619-26
  199. Hasegawa K, Saga Y (2012) Retinoic acid signaling in Sertoli cells regulates organization of the blood-testis barrier through cyclical changes in gene expression. Development. 139(23): 4347-55
  200. Nicholls PK, Harrison CA, Rainczuk KE, Wayne Vogl A, Stanton PG (2013) Retinoic acid promotes Sertoli cell differentiation and antagonises activin-induced proliferation. Mol Cell Endocrinol. 377(1-2): 33-43
  201. O'Shaughnessy PJ (2014) Hormonal control of germ cell development and spermatogenesis. Semin Cell Dev Biol. 29: 55-65
  202. Smith LB, Walker WH, O'Donnell L (2015) Hormonal regulation of spermatogenesis through Sertoli cells by androgens and estrogens, in Sertoli cell biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 175-200
  203. O'Donnell L, Meachem SJ, Stanton PG, McLachlan RI (2006) Endocrine regulation of spermatogenesis, in Knobil and Neill's Physiology of Reproduction, Neill, J.D., Editor Elsevier: San Diego, CA. p. 1017-1069
  204. Ruwanpura SM, McLachlan RI, Meachem SJ (2010) Hormonal regulation of male germ cell development. J Endocrinol. 205(2): 117-31
  205. Pitetti JL, Calvel P, Romero Y, Conne B, Truong V, Papaioannou MD, Schaad O, Docquier M, Herrera PL, Wilhelm D, Nef S (2013) Insulin and IGF1 receptors are essential for XX and XY gonadal differentiation and adrenal development in mice. PLoS Genet. 9(1): e1003160
  206. Rodriguez I, Ody C, Araki K, Garcia I, Vassalli P (1997) An early and massive wave of germinal cell apoptosis is required for the development of functional spermatogenesis. EMBO J. 16(9): 2262-70
  207. Wreford NG, Rajendra Kumar T, Matzuk MM, de Kretser DM (2001) Analysis of the testicular phenotype of the follicle-stimulating hormone beta-subunit knockout and the activin type II receptor knockout mice by stereological analysis. Endocrinology. 142(7): 2916-20
  208. Grover A, Sairam MR, Smith CE, Hermo L (2004) Structural and functional modifications of Sertoli cells in the testis of adult follicle-stimulating hormone receptor knockout mice. Biol Reprod. 71(1): 117-29
  209. Matthiesson KL, McLachlan RI, O'Donnell L, Frydenberg M, Robertson DM, Stanton PG, Meachem SJ (2006) The relative roles of follicle-stimulating hormone and luteinizing hormone in maintaining spermatogonial maturation and spermiation in normal men. J Clin Endocrinol Metab. 91(10): 3962-9
  210. Nieschlag E, Simoni M, Gromoll J, Weinbauer GF (1999) Role of FSH in the regulation of spermatogenesis: clinical aspects. Clin Endocrinol (Oxf). 51(2): 139-46
  211. Simoni M, Weinbauer GF, Gromoll J, Nieschlag E (1999) Role of FSH in male gonadal function. Ann Endocrinol (Paris). 60(2): 102-6
  212. Walker WH (2010) Non-classical actions of testosterone and spermatogenesis. Philos Trans R Soc Lond B Biol Sci. 365(1546): 1557-69
  213. Toocheck C, Clister T, Shupe J, Crum C, Ravindranathan P, Lee TK, Ahn JM, Raj GV, Sukhwani M, Orwig KE, Walker WH (2016) Mouse Spermatogenesis Requires Classical and Nonclassical Testosterone Signaling. Biol Reprod. 94(1): 11
  214. Chang C, Chen YT, Yeh SD, Xu Q, Wang RS, Guillou F, Lardy H, Yeh S (2004) Infertility with defective spermatogenesis and hypotestosteronemia in male mice lacking the androgen receptor in Sertoli cells. Proc Natl Acad Sci U S A. 101(18): 6876-81
  215. De Gendt K, Swinnen JV, Saunders PT, et al. (2004) A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc Natl Acad Sci U S A. 101(5): 1327-32
  216. De Gendt K, Verhoeven G, Amieux PS, Wilkinson MF (2014) Genome-wide identification of AR-regulated genes translated in Sertoli cells in vivo using the RiboTag approach. Mol Endocrinol. 28(4): 575-91
  217. Abel MH, Baker PJ, Charlton HM, Monteiro A, Verhoeven G, De Gendt K, Guillou F, O'Shaughnessy PJ (2008) Spermatogenesis and Sertoli cell activity in mice lacking Sertoli cell receptors for follicle-stimulating hormone and androgen. Endocrinology. 149(7): 3279-85
  218. Saito K, O'Donnell L, McLachlan RI, Robertson DM (2000) Spermiation failure is a major contributor to early spermatogenic suppression caused by hormone withdrawal in adult rats. Endocrinology. 141(8): 2779-85
  219. Walker WH, Cheng J (2005) FSH and testosterone signaling in Sertoli cells. Reproduction. 130(1): 15-28
  220. Nicholls PK, Harrison CA, Walton KL, McLachlan RI, O'Donnell L, Stanton PG (2011) Hormonal regulation of sertoli cell micro-RNAs at spermiation. Endocrinology. 152(4): 1670-83
  221. Song HW, Wilkinson MF (2014) Transcriptional control of spermatogonial maintenance and differentiation. Semin Cell Dev Biol. 30: 14-26
  222. Manku G, Culty M (2015) Mammalian gonocyte and spermatogonia differentiation: recent advances and remaining challenges. Reproduction. 149(3): R139-57
  223. de Rooij DG (2009) The spermatogonial stem cell niche. Microsc Res Tech. 72(8): 580-5
  224. Oatley MJ, Racicot KE, Oatley JM (2011) Sertoli cells dictate spermatogonial stem cell niches in the mouse testis. Biol Reprod. 84(4): 639-45
  225. Giuili G, Tomljenovic A, Labrecque N, Oulad-Abdelghani M, Rassoulzadegan M, Cuzin F (2002) Murine spermatogonial stem cells: targeted transgene expression and purification in an active state. EMBO Rep. 3(8): 753-9
  226. Meng X, Lindahl M, Hyvonen ME, et al. (2000) Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science. 287(5457): 1489-93
  227. Nagano M, Ryu BY, Brinster CJ, Avarbock MR, Brinster RL (2003) Maintenance of mouse male germ line stem cells in vitro. Biol Reprod. 68(6): 2207-14
  228. Shinohara T, Avarbock MR, Brinster RL (1999) beta1- and alpha6-integrin are surface markers on mouse spermatogonial stem cells. Proc Natl Acad Sci U S A. 96(10): 5504-9
  229. Yomogida K, Yagura Y, Tadokoro Y, Nishimune Y (2003) Dramatic expansion of germinal stem cells by ectopically expressed human glial cell line-derived neurotrophic factor in mouse Sertoli cells. Biol Reprod. 69(4): 1303-7
  230. Oatley JM, Oatley MJ, Avarbock MR, Tobias JW, Brinster RL (2009) Colony stimulating factor 1 is an extrinsic stimulator of mouse spermatogonial stem cell self-renewal. Development. 136(7): 1191-9
  231. DeFalco T, Potter SJ, Williams AV, Waller B, Kan MJ, Capel B (2015) Macrophages Contribute to the Spermatogonial Niche in the Adult Testis. Cell Rep. 12(7): 1107-19
  232. Loveland KL, Schlatt S (1997) Stem cell factor and c-kit in the mammalian testis: lessons originating from Mother Nature's gene knockouts. J Endocrinol. 153(3): 337-44
  233. Vincent S, Segretain D, Nishikawa S, Nishikawa SI, Sage J, Cuzin F, Rassoulzadegan M (1998) Stage-specific expression of the Kit receptor and its ligand (KL) during male gametogenesis in the mouse: a Kit-KL interaction critical for meiosis. Development. 125(22): 4585-93
  234. Blume-Jensen P, Jiang G, Hyman R, Lee KF, O'Gorman S, Hunter T (2000) Kit/stem cell factor receptor-induced activation of phosphatidylinositol 3'-kinase is essential for male fertility. Nat Genet. 24(2): 157-62
  235. Hogarth CA, Griswold MD (2010) The key role of vitamin A in spermatogenesis. J Clin Invest. 120(4): 956-62
  236. Whitmore LS, Ye P (2015) Dissecting Germ Cell Metabolism through Network Modeling. PLoS One. 10(9): e0137607
  237. Griswold MD (2015) The initiation of spermatogenesis and the cycle of the seminiferous

epithelium, in Sertoli Cell Biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 233-246

  1. Ikami K, Tokue M, Sugimoto R, Noda C, Kobayashi S, Hara K, Yoshida S (2015) Hierarchical differentiation competence in response to retinoic acid ensures stem cell maintenance during mouse spermatogenesis. Development. 142(9): 1582-92
  2. Busada JT, Chappell VA, Niedenberger BA, Kaye EP, Keiper BD, Hogarth CA, Geyer CB (2015) Retinoic acid regulates Kit translation during spermatogonial differentiation in the mouse. Dev Biol. 397(1): 140-9
  3. Busada JT, Niedenberger BA, Velte EK, Keiper BD, Geyer CB (2015) Mammalian target of rapamycin complex 1 (mTORC1) Is required for mouse spermatogonial differentiation in vivo. Dev Biol. DBIO15155
  4. McLachlan RI, O'Donnell L, Meachem SJ, Stanton PG, de Kretser DM, Pratis K, Robertson DM (2002) Identification of specific sites of hormonal regulation in spermatogenesis in rats, monkeys, and man. Recent Prog Horm Res. 57: 149-79
  5. Schlatt S, Ehmcke J (2014) Regulation of spermatogenesis: an evolutionary biologist's perspective. Semin Cell Dev Biol. 29: 2-16
  6. Haywood M, Spaliviero J, Jimemez M, King NJ, Handelsman DJ, Allan CM (2003) Sertoli and germ cell development in hypogonadal (hpg) mice expressing transgenic follicle-stimulating hormone alone or in combination with testosterone. Endocrinology. 144(2): 509-17
  7. Shetty G, Wilson G, Huhtaniemi I, Boettger-Tong H, Meistrich ML (2001) Testosterone inhibits spermatogonial differentiation in juvenile spermatogonial depletion mice. Endocrinology. 142(7): 2789-95
  8. Marshall GR, Zorub DS, Plant TM (1995) Follicle-stimulating hormone amplifies the population of differentiated spermatogonia in the hypophysectomized testosterone-replaced adult rhesus monkey (Macaca mulatta). Endocrinology. 136(8): 3504-11
  9. Weinbauer GF, Behre HM, Fingscheidt U, Nieschlag E (1991) Human follicle-stimulating hormone exerts a stimulatory effect on spermatogenesis, testicular size, and serum inhibin levels in the gonadotropin-releasing hormone antagonist-treated nonhuman primate (Macaca fascicularis). Endocrinology. 129(4): 1831-9
  10. De Rooij DG (2015) The spermatogonial stem cell niche in mammals, in Sertoli Cell Biology, Griswold, M.D., Editor Elsevier: Waltham, MA. p. 99-122
  11. Tanaka T, Kanatsu-Shinohara M, Lei Z, Rao CV, Shinohara T (2016) The Luteinizing Hormone-Testosterone Pathway Regulates Mouse Spermatogonial Stem Cell Self-Renewal by Suppressing WNT5A Expression in Sertoli Cells. Stem Cell Reports. 7(2): 279-91
  12. Anderson EL, Baltus AE, Roepers-Gajadien HL, Hassold TJ, de Rooij DG, van Pelt AM, Page DC (2008) Stra8 and its inducer, retinoic acid, regulate meiotic initiation in both spermatogenesis and oogenesis in mice. Proc Natl Acad Sci U S A. 105(39): 14976-80
  13. Mark M, Jacobs H, Oulad-Abdelghani M, Dennefeld C, Feret B, Vernet N, Codreanu CA, Chambon P, Ghyselinck NB (2008) STRA8-deficient spermatocytes initiate, but fail to complete, meiosis and undergo premature chromosome condensation. J Cell Sci. 121(Pt 19): 3233-42
  14. Koubova J, Hu YC, Bhattacharyya T, Soh YQ, Gill ME, Goodheart ML, Hogarth CA, Griswold MD, Page DC (2014) Retinoic acid activates two pathways required for meiosis in mice. PLoS Genet. 10(8): e1004541
  15. Abby E, Tourpin S, Ribeiro J, et al. (2016) Implementation of meiosis prophase I programme requires a conserved retinoid-independent stabilizer of meiotic transcripts. Nat Commun. 7: 10324
  16. Hermo L, Pelletier RM, Cyr DG, Smith CE (2010) Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 1: background to spermatogenesis, spermatogonia, and spermatocytes. Microsc Res Tech. 73(4): 241-78
  17. Morelli MA, Cohen PE (2005) Not all germ cells are created equal: aspects of sexual dimorphism in mammalian meiosis. Reproduction. 130(6): 761-81
  18. Sanderson ML, Hassold TJ, Carrell DT (2008) Proteins involved in meiotic recombination: a role in male infertility? Syst Biol Reprod Med. 54(2): 57-74
  19. Baker SM, Bronner CE, Zhang L, et al. (1995) Male mice defective in the DNA mismatch repair gene PMS2 exhibit abnormal chromosome synapsis in meiosis. Cell. 82(2): 309-19
  20. Zhu D, Dix DJ, Eddy EM (1997) HSP70-2 is required for CDC2 kinase activity in meiosis I of mouse spermatocytes. Development. 124(15): 3007-14
  21. Alekseev OM, Richardson RT, O'Rand MG (2009) Linker histones stimulate HSPA2 ATPase activity through NASP binding and inhibit CDC2/Cyclin B1 complex formation during meiosis in the mouse. Biol Reprod. 81(4): 739-48
  22. Eto K, Shiotsuki M, Abe S (2013) Nociceptin induces Rec8 phosphorylation and meiosis in postnatal murine testes. Endocrinology. 154(8): 2891-9
  23. Eto K (2015) Nociceptin and meiosis during spermatogenesis in postnatal testes. Vitam Horm. 97: 167-86
  24. Bolcun-Filas E, Bannister LA, Barash A, Schimenti KJ, Hartford SA, Eppig JJ, Handel MA, Shen L, Schimenti JC (2011) A-MYB (MYBL1) transcription factor is a master regulator of male meiosis. Development. 138(15): 3319-30
  25. O'Hara L, Smith LB (2015) Androgen receptor roles in spermatogenesis and infertility. Best Pract Res Clin Endocrinol Metab. 29(4): 595-605
  26. Handelsman DJ (2011) Hormonal regulation of spermatogenesis: insights from constructing genetic models. Reprod Fertil Dev. 23(4): 507-19
  27. El Shennawy A, Gates RJ, Russell LD (1998) Hormonal regulation of spermatogenesis in the hypophysectomized rat: cell viability after hormonal replacement in adults after intermediate periods of hypophysectomy. J Androl. 19(3): 320-34; discussion 341-2
  28. Chalmel F, Rolland AD, Niederhauser-Wiederkehr C, Chung SS, Demougin P, Gattiker A, Moore J, Patard JJ, Wolgemuth DJ, Jegou B, Primig M (2007) The conserved transcriptome in human and rodent male gametogenesis. Proc Natl Acad Sci U S A. 104(20): 8346-51
  29. Elliott DJ, Grellscheid SN (2006) Alternative RNA splicing regulation in the testis. Reproduction. 132(6): 811-9
  30. Foulkes NS, Mellstrom B, Benusiglio E, Sassone-Corsi P (1992) Developmental switch of CREM function during spermatogenesis: from antagonist to activator. Nature. 355(6355): 80-4
  31. Mandel CR, Bai Y, Tong L (2008) Protein factors in pre-mRNA 3'-end processing. Cell Mol Life Sci. 65(7-8): 1099-122
  32. MacDonald CC, McMahon KW (2010) Tissue-specific mechanisms of alternative polyadenylation: testis, brain, and beyond. Wiley Interdiscip Rev RNA. 1(3): 494-501
  33. Li W, Park JY, Zheng D, Hoque M, Yehia G, Tian B (2016) Alternative cleavage and polyadenylation in spermatogenesis connects chromatin regulation with post-transcriptional control. BMC Biol. 14(1): 6
  34. Hogeveen KN, Sassone-Corsi P (2006) Regulation of gene expression in post-meiotic male germ cells: CREM-signalling pathways and male fertility. Hum Fertil (Camb). 9(2): 73-9
  35. Macho B, Brancorsini S, Fimia GM, Setou M, Hirokawa N, Sassone-Corsi P (2002) CREM-dependent transcription in male germ cells controlled by a kinesin. Science. 298(5602): 2388-90
  36. Zhang D, Penttila TL, Morris PL, Teichmann M, Roeder RG (2001) Spermiogenesis deficiency in mice lacking the Trf2 gene. Science. 292(5519): 1153-5
  37. Wu Y, Hu X, Li Z, et al. (2016) Transcription Factor RFX2 Is a Key Regulator of Mouse Spermiogenesis. Sci Rep. 6: 20435
  38. Kleene KC (2003) Patterns, mechanisms, and functions of translation regulation in mammalian spermatogenic cells. Cytogenet Genome Res. 103(3-4): 217-24
  39. Cullinane DL, Chowdhury TA, Kleene KC (2015) Mechanisms of translational repression of the Smcp mRNA in round spermatids. Reproduction. 149(1): 43-54
  40. Kleene KC (2016) Position-dependent interactions of Y-box protein 2 (YBX2) with mRNA enable mRNA storage in round spermatids by repressing mRNA translation and blocking translation-dependent mRNA decay. Mol Reprod Dev.
  41. Ren D, Navarro B, Perez G, Jackson AC, Hsu S, Shi Q, Tilly JL, Clapham DE (2001) A sperm ion channel required for sperm motility and male fertility. Nature. 413(6856): 603-9
  42. Escudier E, Duquesnoy P, Papon JF, Amselem S (2009) Ciliary defects and genetics of primary ciliary dyskinesia. Paediatr Respir Rev. 10(2): 51-4
  43. Chung SS, Wang X, Wolgemuth DJ (2009) Expression of retinoic acid receptor alpha in the germline is essential for proper cellular association and spermiogenesis during spermatogenesis. Development. 136(12): 2091-100
  44. Holdcraft RW, Braun RE (2004) Androgen receptor function is required in Sertoli cells for the terminal differentiation of haploid spermatids. Development. 131(2): 459-67
  45. O'Donnell L, McLachlan RI, Wreford NG, Robertson DM (1994) Testosterone promotes the conversion of round spermatids between stages VII and VIII of the rat spermatogenic cycle. Endocrinology. 135(6): 2608-14
  46. McLachlan RI, O'Donnell L, Stanton PG, Balourdos G, Frydenberg M, de Kretser DM, Robertson DM (2002) Effects of testosterone plus medroxyprogesterone acetate on semen quality, reproductive hormones, and germ cell populations in normal young men. J Clin Endocrinol Metab. 87(2): 546-56
  47. Heller CG, Clermont Y (1963) Spermatogenesis in man: an estimate of its duration. Science. 140(3563): 184-6
  48. Soumillon M, Necsulea A, Weier M, et al. (2013) Cellular source and mechanisms of high transcriptome complexity in the mammalian testis. Cell Rep. 3(6): 2179-90
  49. Luo LF, Hou CC, Yang WX (2016) Small non-coding RNAs and their associated proteins in spermatogenesis. Gene. 578(2): 141-57
  50. Siomi MC, Sato K, Pezic D, Aravin AA (2011) PIWI-interacting small RNAs: the vanguard of genome defence. Nat Rev Mol Cell Biol. 12(4): 246-58
  51. Fu Q, Wang PJ (2014) Mammalian piRNAs: Biogenesis, function, and mysteries. Spermatogenesis. 4: e27889
  52. Wang L, Xu C (2015) Role of microRNAs in mammalian spermatogenesis and testicular germ cell tumors. Reproduction. 149(3): R127-37
  53. de Mateo S, Sassone-Corsi P (2014) Regulation of spermatogenesis by small non-coding RNAs: role of the germ granule. Semin Cell Dev Biol. 29: 84-92
  54. Yadav RP, Kotaja N (2014) Small RNAs in spermatogenesis. Mol Cell Endocrinol. 382(1): 498-508
  55. Hogg K, Western PS (2015) Refurbishing the germline epigenome: Out with the old, in with the new. Semin Cell Dev Biol. 45: 104-13
  56. Jurka J, Kapitonov VV, Kohany O, Jurka MV (2007) Repetitive sequences in complex genomes: structure and evolution. Annu Rev Genomics Hum Genet. 8: 241-59
  57. Rayan NA, Del Rosario RC, Prabhakar S (2016) Massive contribution of transposable elements to mammalian regulatory sequences. Semin Cell Dev Biol.
  58. Bourc'his D, Bestor TH (2004) Meiotic catastrophe and retrotransposon reactivation in male germ cells lacking Dnmt3L. Nature. 431(7004): 96-9
  59. Pastor WA, Stroud H, Nee K, et al. (2014) MORC1 represses transposable elements in the mouse male germline. Nat Commun. 5: 5795
  60. Kuramochi-Miyagawa S, Watanabe T, Gotoh K, et al. (2008) DNA methylation of retrotransposon genes is regulated by Piwi family members MILI and MIWI2 in murine fetal testes. Genes Dev. 22(7): 908-17
  61. Aravin AA, Sachidanandam R, Girard A, Fejes-Toth K, Hannon GJ (2007) Developmentally regulated piRNA clusters implicate MILI in transposon control. Science. 316(5825): 744-7
  62. Carmell MA, Girard A, van de Kant HJ, Bourc'his D, Bestor TH, de Rooij DG, Hannon GJ (2007) MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline. Dev Cell. 12(4): 503-14
  63. Werner A, Piatek MJ, Mattick JS (2015) Transpositional shuffling and quality control in male germ cells to enhance evolution of complex organisms. Ann N Y Acad Sci. 1341: 156-63
  64. Ashe A, Sapetschnig A, Weick EM, et al. (2012) piRNAs can trigger a multigenerational epigenetic memory in the germline of C. elegans. Cell. 150(1): 88-99
  65. Schuster A, Skinner MK, Yan W (2016) Ancestral vinclozolin exposure alters the epigenetic transgenerational inheritance of sperm small noncoding RNAs. Environ Epigenet. 2(1)
  66. Meikar O, Vagin VV, Chalmel F, et al. (2014) An atlas of chromatoid body components. RNA. 20(4): 483-95
  67. Meikar O, Da Ros M, Korhonen H, Kotaja N (2011) Chromatoid body and small RNAs in male germ cells. Reproduction. 142(2): 195-209
  68. Yao C, Liu Y, Sun M, Niu M, Yuan Q, Hai Y, Guo Y, Chen Z, Hou J, He Z (2015) MicroRNAs and DNA methylation as epigenetic regulators of mitosis, meiosis and spermiogenesis. Reproduction. 150(1): R25-34
  69. Shirakawa T, Yaman-Deveci R, Tomizawa S, et al. (2013) An epigenetic switch is crucial for spermatogonia to exit the undifferentiated state toward a Kit-positive identity. Development. 140(17): 3565-76
  70. Papaioannou MD, Nef S (2010) microRNAs in the testis: building up male fertility. J Androl. 31(1): 26-33
  71. Winter J, Jung S, Keller S, Gregory RI, Diederichs S (2009) Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat Cell Biol. 11(3): 228-34
  72. Wu Q, Song R, Ortogero N, Zheng H, Evanoff R, Small CL, Griswold MD, Namekawa SH, Royo H, Turner JM, Yan W (2012) The RNase III Enzyme DROSHA Is Essential for MicroRNA Production and Spermatogenesis. J Biol Chem. 287(30): 25173-90
  73. Ro S, Park C, Sanders KM, McCarrey JR, Yan W (2007) Cloning and expression profiling of testis-expressed microRNAs. Dev Biol. 311(2): 592-602
  74. Han T, Manoharan AP, Harkins TT, Bouffard P, Fitzpatrick C, Chu DS, Thierry-Mieg D, Thierry-Mieg J, Kim JK (2009) 26G endo-siRNAs regulate spermatogenic and zygotic gene expression in Caenorhabditis elegans. Proc Natl Acad Sci U S A. 106(44): 18674-9
  75. Pavelec DM, Lachowiec J, Duchaine TF, Smith HE, Kennedy S (2009) Requirement for the ERI/DICER complex in endogenous RNA interference and sperm development in Caenorhabditis elegans. Genetics. 183(4): 1283-95
  76. Song R, Hennig GW, Wu Q, Jose C, Zheng H, Yan W (2011) Male germ cells express abundant endogenous siRNAs. Proc Natl Acad Sci U S A. 108(32): 13159-64
  77. Ortogero N, Schuster AS, Oliver DK, et al. (2014) A novel class of somatic small RNAs similar to germ cell pachytene PIWI-interacting small RNAs. J Biol Chem. 289(47): 32824-34
  78. Lim SL, Qu ZP, Kortschak RD, et al. (2015) HENMT1 and piRNA Stability Are Required for Adult Male Germ Cell Transposon Repression and to Define the Spermatogenic Program in the Mouse. PLoS Genet. 11(10): e1005620
  79. Reuter M, Berninger P, Chuma S, Shah H, Hosokawa M, Funaya C, Antony C, Sachidanandam R, Pillai RS (2011) Miwi catalysis is required for piRNA amplification-independent LINE1 transposon silencing. Nature. 480(7376): 264-7
  80. Di Giacomo M, Comazzetto S, Saini H, De Fazio S, Carrieri C, Morgan M, Vasiliauskaite L, Benes V, Enright AJ, O'Carroll D (2013) Multiple epigenetic mechanisms and the piRNA pathway enforce LINE1 silencing during adult spermatogenesis. Mol Cell. 50(4): 601-8
  81. Pantano L, Jodar M, Bak M, Ballesca JL, Tommerup N, Oliva R, Vavouri T (2015) The small RNA content of human sperm reveals pseudogene-derived piRNAs complementary to protein-coding genes. RNA. 21(6): 1085-95
  82. Goh WS, Falciatori I, Tam OH, Burgess R, Meikar O, Kotaja N, Hammell M, Hannon GJ (2015) piRNA-directed cleavage of meiotic transcripts regulates spermatogenesis. Genes Dev. 29(10): 1032-44
  83. Watanabe T, Cheng EC, Zhong M, Lin H (2015) Retrotransposons and pseudogenes regulate mRNAs and lncRNAs via the piRNA pathway in the germline. Genome Res. 25(3): 368-80
  84. Zhang P, Kang JY, Gou LT, et al. (2015) MIWI and piRNA-mediated cleavage of messenger RNAs in mouse testes. Cell Res. 25(2): 193-207
  85. Luk AC, Chan WY, Rennert OM, Lee TL (2014) Long noncoding RNAs in spermatogenesis: insights from recent high-throughput transcriptome studies. Reproduction. 147(5): R131-41
  86. Zhang C, Gao L, Xu EY (2016) LncRNA, a new component of expanding RNA-protein regulatory network important for animal sperm development. Semin Cell Dev Biol.
  87. Lin X, Han M, Cheng L, Chen J, Zhang Z, Shen T, Wang M, Wen B, Ni T, Han C (2016) Expression dynamics, relationships, and transcriptional regulations of diverse transcripts in mouse spermatogenic cells. RNA Biol. 13(10): 1011-1024
  88. Liang M, Li W, Tian H, Hu T, Wang L, Lin Y, Li Y, Huang H, Sun F (2014) Sequential expression of long noncoding RNA as mRNA gene expression in specific stages of mouse spermatogenesis. Sci Rep. 4: 5966
  89. Wen K, Yang L, Xiong T, et al. (2016) Critical roles of long noncoding RNAs in Drosophila spermatogenesis. Genome Res. 26(9): 1233-44
  90. Li L, Wang M, Wu X, Geng L, Xue Y, Wei X, Jia Y (2016) A long non-coding RNA interacts with Gfra1 and maintains survival of mouse spermatogonial stem cells. Cell Death Dis. 7: e2140
  91. Stewart KR, Veselovska L, Kelsey G (2016) Establishment and functions of DNA methylation in the germline. Epigenomics. 8(10): 1399-1413
  92. Ly L, Chan D, Trasler JM (2015) Developmental windows of susceptibility for epigenetic inheritance through the male germline. Semin Cell Dev Biol. 43: 96-105
  93. Soubry A, Hoyo C, Jirtle RL, Murphy SK (2014) A paternal environmental legacy: evidence for epigenetic inheritance through the male germ line. Bioessays. 36(4): 359-71
  94. Wei Y, Schatten H, Sun QY (2015) Environmental epigenetic inheritance through gametes and implications for human reproduction. Hum Reprod Update. 21(2): 194-208
  95. Skinner MK (2014) Endocrine disruptor induction of epigenetic transgenerational inheritance of disease. Mol Cell Endocrinol. 398(1-2): 4-12
  96. Skinner MK (2016) Endocrine disruptors in 2015: Epigenetic transgenerational inheritance. Nat Rev Endocrinol. 12(2): 68-70
  97. Anway MD, Rekow SS, Skinner MK (2008) Transgenerational epigenetic programming of the embryonic testis transcriptome. Genomics. 91(1): 30-40
  98. Anway MD, Cupp AS, Uzumcu M, Skinner MK (2005) Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science. 308(5727): 1466-9
  99. Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ (2010) Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature. 467(7318): 963-6
  100. Fullston T, Ohlsson Teague EM, Palmer NO, DeBlasio MJ, Mitchell M, Corbett M, Print CG, Owens JA, Lane M (2013) Paternal obesity initiates metabolic disturbances in two generations of mice with incomplete penetrance to the F2 generation and alters the transcriptional profile of testis and sperm microRNA content. FASEB J. 27(10): 4226-43
  101. Lambrot R, Xu C, Saint-Phar S, Chountalos G, Cohen T, Paquet M, Suderman M, Hallett M, Kimmins S (2013) Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat Commun. 4: 2889
  102. Cui X, Jing X, Wu X, Yan M, Li Q, Shen Y, Wang Z (2016) DNA methylation in spermatogenesis and male infertility. Exp Ther Med. 12(4): 1973-1979
  103. Rando OJ (2012) Daddy issues: paternal effects on phenotype. Cell. 151(4): 702-8
  104. Daxinger L, Whitelaw E (2012) Understanding transgenerational epigenetic inheritance via the gametes in mammals. Nat Rev Genet. 13(3): 153-62
  105. Conine CC, Moresco JJ, Gu W, Shirayama M, Conte D, Jr., Yates JR, 3rd, Mello CC (2013) Argonautes promote male fertility and provide a paternal memory of germline gene expression in C. elegans. Cell. 155(7): 1532-44
  106. Yuan S, Schuster A, Tang C, Yu T, Ortogero N, Bao J, Zheng H, Yan W (2016) Sperm-borne miRNAs and endo-siRNAs are important for fertilization and preimplantation embryonic development. Development. 143(4): 635-47
  107. Gapp K, Jawaid A, Sarkies P, Bohacek J, Pelczar P, Prados J, Farinelli L, Miska E, Mansuy IM (2014) Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat Neurosci. 17(5): 667-9
  108. Sharma U, Conine CC, Shea JM, et al. (2016) Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science. 351(6271): 391-6
  109. Hayes F, Dwyer A, Pitteloud N. (2013) Hypogonadotropic hypogondism (HH) and gonadotropin therapy, in www.ENDOTEXT.org, Endocrinology of Male Reproduction, Section Editor McLachlan R.I. MDTEXTCOM Inc.: South Dartmouth, MA
  110. Plant TM (2008) Hypothalamic control of the pituitary-gonadal axis in higher primates: key advances over the last two decades. J Neuroendocrinol. 20(6): 719-26
  111. Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M (2012) Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev. 92(3): 1235-316
  112. Mittelman-Smith MA, Williams H, Krajewski-Hall SJ, Lai J, Ciofi P, McMullen NT, Rance NE (2012) Arcuate kisspeptin/neurokinin B/dynorphin (KNDy) neurons mediate the estrogen suppression of gonadotropin secretion and body weight. Endocrinology. 153(6): 2800-12
  113. Tsutsui K, Ubuka T, Bentley GE, Kriegsfeld LJ (2012) Gonadotropin-inhibitory hormone (GnIH): discovery, progress and prospect. Gen Comp Endocrinol. 177(3): 305-14
  114. Clarke IJ (2011) Control of GnRH secretion: one step back. Front Neuroendocrinol. 32(3): 367-75
  115. Phillips DJ, de Kretser DM (1998) Follistatin: a multifunctional regulatory protein. Front Neuroendocrinol. 19(4): 287-322
  116. Jin JM, Yang WX (2014) Molecular regulation of hypothalamus-pituitary-gonads axis in males. Gene. 551(1): 15-25
  117. Hashimoto O, Nakamura T, Shoji H, Shimasaki S, Hayashi Y, Sugino H (1997) A novel role of follistatin, an activin-binding protein, in the inhibition of activin action in rat pituitary cells. Endocytotic degradation of activin and its acceleration by follistatin associated with cell-surface heparan sulfate. J Biol Chem. 272(21): 13835-42
  118. Sherins RJ, Loriaux DL (1973) Studies of the role of sex steroids in the feedback control of FSH concentrations in men. J Clin Endocrinol Metab. 36(5): 886-93
  119. Naftolin F, Ryan KJ, Petro Z (1971) Aromatization of androstenedione by the diencephalon. J Clin Endocrinol Metab. 33(2): 368-70
  120. Santen RJ (1975) Is aromatization of testosterone to estradiol required for inhibition of luteinizing hormone secretion in men? J Clin Invest. 56(6): 1555-63
  121. Santen RJ, Bardin CW (1973) Episodic luteinizing hormone secretion in man. Pulse analysis, clinical interpretation, physiologic mechanisms. J Clin Invest. 52(10): 2617-28
  122. Hayes FJ, Seminara SB, Decruz S, Boepple PA, Crowley WF, Jr. (2000) Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. J Clin Endocrinol Metab. 85(9): 3027-35
  123. Tilbrook AJ, de Kretser DM, Cummins JT, Clarke IJ (1991) The negative feedback effects of testicular steroids are predominantly at the hypothalamus in the ram. Endocrinology. 129(6): 3080-92
  124. Decker MH, Loriaux DL, Cutler GB, Jr. (1981) A seminiferous tubular factor is not obligatory for regulation of plasma follicle-stimulating hormone in the rat. Endocrinology. 108(3): 1035-9
  125. McCullagh DR (1932) Dual Endocrine Activity of the Testes. Science. 76(1957): 19-20
  126. de Kretser DM, Robertson DM (1989) The isolation and physiology of inhibin and related proteins. Biol Reprod. 40(1): 33-47
  127. Forage RG, Ring JM, Brown RW, McInerney BV, Cobon GS, Gregson RP, Robertson DM, Morgan FJ, Hearn MT, Findlay JK, et al. (1986) Cloning and sequence analysis of cDNA species coding for the two subunits of inhibin from bovine follicular fluid. Proc Natl Acad Sci U S A. 83(10): 3091-5
  128. Ling N, Ying SY, Ueno N, Esch F, Denoroy L, Guillemin R (1985) Isolation and partial characterization of a Mr 32,000 protein with inhibin activity from porcine follicular fluid. Proc Natl Acad Sci U S A. 82(21): 7217-21
  129. Mason AJ, Hayflick JS, Ling N, Esch F, Ueno N, Ying SY, Guillemin R, Niall H, Seeburg PH (1985) Complementary DNA sequences of ovarian follicular fluid inhibin show precursor structure and homology with transforming growth factor-beta. Nature. 318(6047): 659-63
  130. Robertson DM, Foulds LM, Leversha L, Morgan FJ, Hearn MT, Burger HG, Wettenhall RE, de Kretser DM (1985) Isolation of inhibin from bovine follicular fluid. Biochem Biophys Res Commun. 126(1): 220-6
  131. Ling N, Ying SY, Ueno N, Shimasaki S, Esch F, Hotta M, Guillemin R (1986) Pituitary FSH is released by a heterodimer of the beta-subunits from the two forms of inhibin. Nature. 321(6072): 779-82
  132. Vale W, Rivier J, Vaughan J, McClintock R, Corrigan A, Woo W, Karr D, Spiess J (1986) Purification and characterization of an FSH releasing protein from porcine ovarian follicular fluid. Nature. 321(6072): 776-9
  133. Robertson DM, Klein R, de Vos FL, McLachlan RI, Wettenhall RE, Hearn MT, Burger HG, de Kretser DM (1987) The isolation of polypeptides with FSH suppressing activity from bovine follicular fluid which are structurally different to inhibin. Biochem Biophys Res Commun. 149(2): 744-9
  134. Shimasaki S, Koga M, Esch F, Mercado M, Cooksey K, Koba A, Ling N (1988) Porcine follistatin gene structure supports two forms of mature follistatin produced by alternative splicing. Biochem Biophys Res Commun. 152(2): 717-23
  135. Ueno N, Ling N, Ying SY, Esch F, Shimasaki S, Guillemin R (1987) Isolation and partial characterization of follistatin: a single-chain Mr 35,000 monomeric protein that inhibits the release of follicle-stimulating hormone. Proc Natl Acad Sci U S A. 84(23): 8282-6
  136. Nakamura T, Takio K, Eto Y, Shibai H, Titani K, Sugino H (1990) Activin-binding protein from rat ovary is follistatin. Science. 247(4944): 836-8
  137. Rea MA, Marshall GR, Weinbauer GF, Nieschlag E (1986) Testosterone maintains pituitary and serum FSH and spermatogenesis in gonadotrophin-releasing hormone antagonist-suppressed rats. J Endocrinol. 108(1): 101-7
  138. Sun YT, Irby DC, Robertson DM, de Kretser DM (1989) The effects of exogenously administered testosterone on spermatogenesis in intact and hypophysectomized rats. Endocrinology. 125(2): 1000-10
  139. Jackson CM, Morris ID (1977) Gonadotrophin levels in male rats following impairment of Leydig cell function by ethylene dimethanesulphonate. Andrologia. 9(1): 29-35
  140. De Kretser DM, O'Leary PC, Irby DC, Risbridger GP (1989) Inhibin secretion is influenced by Leydig cells: evidence from studies using the cytotoxin ethane dimethane sulphonate (EDS). Int J Androl. 12(4): 273-80
  141. O'Leary P, Jackson AE, Averill S, de Kretser DM (1986) The effects of ethane dimethane sulphonate (EDS) on bilaterally cryptorchid rat testes. Mol Cell Endocrinol. 45(2-3): 183-90
  142. Le Gac F, de Kretser DM (1982) Inhibin production by Sertoli cell cultures. Mol Cell Endocrinol. 28(3): 487-98
  143. Steinberger A, Steinberger E (1976) Secretion of an FSH-inhibiting factor by cultured Sertoli cells. Endocrinology. 99(3): 918-21
  144. Klaij IA, Timmerman MA, Blok LJ, Grootegoed JA, de Jong FH (1992) Regulation of inhibin beta B-subunit mRNA expression in rat Sertoli cells: consequences for the production of bioactive and immunoreactive inhibin. Mol Cell Endocrinol. 85(3): 237-46
  145. Sharpe RM, Turner KJ, McKinnell C, Groome NP, Atanassova N, Millar MR, Buchanan DL, Cooke PS (1999) Inhibin B levels in plasma of the male rat from birth to adulthood: effect of experimental manipulation of Sertoli cell number. J Androl. 20(1): 94-101
  146. Anawalt BD, Bebb RA, Matsumoto AM, Groome NP, Illingworth PJ, McNeilly AS, Bremner WJ (1996) Serum inhibin B levels reflect Sertoli cell function in normal men and men with testicular dysfunction. J Clin Endocrinol Metab. 81(9): 3341-5
  147. Anderson RA, Wallace EM, Groome NP, Bellis AJ, Wu FC (1997) Physiological relationships between inhibin B, follicle stimulating hormone secretion and spermatogenesis in normal men and response to gonadotrophin suppression by exogenous testosterone. Hum Reprod. 12(4): 746-51
  148. Krummen LA, Toppari J, Kim WH, Morelos BS, Ahmad N, Swerdloff RS, Ling N, Shimasaki S, Esch F, Bhasin S (1989) Regulation of testicular inhibin subunit messenger ribonucleic acid levels in vivo: effects of hypophysectomy and selective follicle-stimulating hormone replacement. Endocrinology. 125(3): 1630-7
  149. Wallace EM, Groome NP, Riley SC, Parker AC, Wu FC (1997) Effects of chemotherapy-induced testicular damage on inhibin, gonadotropin, and testosterone secretion: a prospective longitudinal study. J Clin Endocrinol Metab. 82(9): 3111-5
  150. Risbridger GP, Clements J, Robertson DM, Drummond AE, Muir J, Burger HG, de Kretser DM (1989) Immuno- and bioactive inhibin and inhibin alpha-subunit expression in rat Leydig cell cultures. Mol Cell Endocrinol. 66(1): 119-22
  151. McLachlan RI, Matsumoto AM, Burger HG, de Kretser DM, Bremner WJ (1988) Relative roles of follicle-stimulating hormone and luteinizing hormone in the control of inhibin secretion in normal men. J Clin Invest. 82(3): 880-4
  152. Tena-Sempere M, Kero J, Rannikko A, Yan W, Huhtaniemi I (1999) The pattern of inhibin/activin alpha- and betaB-subunit messenger ribonucleic acid expression in rat testis after selective Leydig cell destruction by ethylene dimethane sulfonate. Endocrinology. 140(12): 5761-70
  153. Matthiesson KL, Robertson DM, Burger HG, McLachlan RI (2003) Response of serum inhibin B and pro-alphaC levels to gonadotrophic stimulation in normal men before and after steroidal contraceptive treatment. Hum Reprod. 18(4): 734-43
  154. de Winter JP, Vanderstichele HM, Timmerman MA, Blok LJ, Themmen AP, de Jong FH (1993) Activin is produced by rat Sertoli cells in vitro and can act as an autocrine regulator of Sertoli cell function. Endocrinology. 132(3): 975-82
  155. Lee W, Mason AJ, Schwall R, Szonyi E, Mather JP (1989) Secretion of activin by interstitial cells in the testis. Science. 243(4889): 396-8
  156. McFarlane JR, Foulds LM, Pisciotta A, Robertson DM, de Kretser DM (1996) Measurement of activin in biological fluids by radioimmunoassay, utilizing dissociating agents to remove the interference of follistatin. Eur J Endocrinol. 134(4): 481-9
  157. Mather JP, Attie KM, Woodruff TK, Rice GC, Phillips DM (1990) Activin stimulates spermatogonial proliferation in germ-Sertoli cell cocultures from immature rat testis. Endocrinology. 127(6): 3206-14
  158. Archambeault DR, Yao HH (2010) Activin A, a product of fetal Leydig cells, is a unique paracrine regulator of Sertoli cell proliferation and fetal testis cord expansion. Proc Natl Acad Sci U S A. 107(23): 10526-31
  159. Buzzard JJ, Farnworth PG, De Kretser DM, O'Connor AE, Wreford NG, Morrison JR (2003) Proliferative phase sertoli cells display a developmentally regulated response to activin in vitro. Endocrinology. 144(2): 474-83
  160. Mendis SH, Meachem SJ, Sarraj MA, Loveland KL (2011) Activin A balances Sertoli and germ cell proliferation in the fetal mouse testis. Biol Reprod. 84(2): 379-91
  161. de Winter JP, Themmen AP, Hoogerbrugge JW, Klaij IA, Grootegoed JA, de Jong FH (1992) Activin receptor mRNA expression in rat testicular cell types. Mol Cell Endocrinol. 83(1): R1-8
  162. Meinhardt A, O'Bryan MK, McFarlane JR, Loveland KL, Mallidis C, Foulds LM, Phillips DJ, de Kretser DM (1998) Localization of follistatin in the rat testis. J Reprod Fertil. 112(2): 233-41
  163. Michel U, Albiston A, Findlay JK (1990) Rat follistatin: gonadal and extragonadal expression and evidence for alternative splicing. Biochem Biophys Res Commun. 173(1): 401-7
  164. Tilbrook AJ, de Kretser DM, Dunshea FR, Klein R, Robertson DM, Clarke IJ, Maddocks S (1996) The testis is not the major source of circulating follistatin in the ram. J Endocrinol. 149(1): 55-63
  165. Phillips DJ, Hedger MP, McFarlane JR, Klein R, Clarke IJ, Tilbrook AJ, Nash AD, de Kretser DM (1996) Follistatin concentrations in male sheep increase following sham castration/castration or injection of interleukin-1 beta. J Endocrinol. 151(1): 119-24
  166. Dubey AK, Zeleznik AJ, Plant TM (1987) In the rhesus monkey (Macaca mulatta), the negative feedback regulation of follicle-stimulating hormone secretion by an action of testicular hormone directly at the level of the anterior pituitary gland cannot be accounted for by either testosterone or estradiol. Endocrinology. 121(6): 2229-37
  167. Ramaswamy S, Pohl CR, McNeilly AS, Winters SJ, Plant TM (1998) The time course of follicle-stimulating hormone suppression by recombinant human inhibin A in the adult male rhesus monkey (Macaca mulatta). Endocrinology. 139(8): 3409-15
  168. Robertson DM, Prisk M, McMaster JW, Irby DC, Findlay JK, de Kretser DM (1991) Serum FSH-suppressing activity of human recombinant inhibin A in male and female rats. J Reprod Fertil. 91(1): 321-8
  169. Tilbrook AJ, de Kretser DM, Clarke IJ (1993) Human recombinant inhibin A and testosterone act directly at the pituitary to suppress plasma concentrations of FSH in castrated rams. J Endocrinol. 138(2): 181-9
  170. Tilbrook AJ, De Kretser DM, Clarke IJ (1993) Human recombinant inhibin A suppresses plasma follicle-stimulating hormone to intact levels but has no effect on luteinizing hormone in castrated rams. Biol Reprod. 49(4): 779-88
  171. Roberts V, Meunier H, Vaughan J, Rivier J, Rivier C, Vale W, Sawchenko P (1989) Production and regulation of inhibin subunits in pituitary gonadotropes. Endocrinology. 124(1): 552-4
  172. Corrigan AZ, Bilezikjian LM, Carroll RS, Bald LN, Schmelzer CH, Fendly BM, Mason AJ, Chin WW, Schwall RH, Vale W (1991) Evidence for an autocrine role of activin B within rat anterior pituitary cultures. Endocrinology. 128(3): 1682-4
  173. Gospodarowicz D, Lau K (1989) Pituitary follicular cells secrete both vascular endothelial growth factor and follistatin. Biochem Biophys Res Commun. 165(1): 292-8
  174. Kogawa K, Nakamura T, Sugino K, Takio K, Titani K, Sugino H (1991) Activin-binding protein is present in pituitary. Endocrinology. 128(3): 1434-40
  175. Bilezikjian LM, Corrigan AZ, Blount AL, Vale WW (1996) Pituitary follistatin and inhibin subunit messenger ribonucleic acid levels are differentially regulated by local and hormonal factors. Endocrinology. 137(10): 4277-84
  176. Bilezikjian LM, Vaughan JM, Vale WW (1993) Characterization and the regulation of inhibin/activin subunit proteins of cultured rat anterior pituitary cells. Endocrinology. 133(6): 2545-53
  177. Gonzales GF, Risbridger GP, de Krester DM (1989) In vivo and in vitro production of inhibin by cryptorchid testes from adult rats. Endocrinology. 124(4): 1661-8
  178. Weinbauer GF, Bartlett JM, Fingscheidt U, Tsonis CG, de Kretser DM, Nieschlag E (1989) Evidence for a major role of inhibin in the feedback control of FSH in the male rat. J Reprod Fertil. 85(2): 355-62
  179. Jensen TK, Andersson AM, Hjollund NH, et al. (1997) Inhibin B as a serum marker of spermatogenesis: correlation to differences in sperm concentration and follicle-stimulating hormone levels. A study of 349 Danish men. J Clin Endocrinol Metab. 82(12): 4059-63
  180. Matzuk MM, Kumar TR, Bradley A (1995) Different phenotypes for mice deficient in either activins or activin receptor type II. Nature. 374(6520): 356-60
  181. Singh J, O'Neill C, Handelsman DJ (1995) Induction of spermatogenesis by androgens in gonadotropin-deficient (hpg) mice. Endocrinology. 136(12): 5311-21
  182. O’Donnell L, McLachlan RI (2012) The role of testosterone in spermatogenesis, in Testosterone: action, deficiency, substitution, Nieschlag, E., Behre, H.M., and Nieschlag, S., Editors. Cambridge University Press: New York, USA. p. 123-153
  183. Huang HF, Marshall GR, Rosenberg R, Nieschlag E (1987) Restoration of spermatogenesis by high levels of testosterone in hypophysectomized rats after long-term regression. Acta Endocrinol (Copenh). 116(4): 433-44
  184. Grino PB, Griffin JE, Wilson JD (1990) Testosterone at high concentrations interacts with the human androgen receptor similarly to dihydrotestosterone. Endocrinology. 126(2): 1165-72
  185. Handelsman DJ, Spaliviero JA, Simpson JM, Allan CM, Singh J (1999) Spermatogenesis without gonadotropins: maintenance has a lower testosterone threshold than initiation. Endocrinology. 140(9): 3938-46
  186. Zhang FP, Pakarainen T, Poutanen M, Toppari J, Huhtaniemi I (2003) The low gonadotropin-independent constitutive production of testicular testosterone is sufficient to maintain spermatogenesis. Proc Natl Acad Sci U S A. 100(23): 13692-7
  187. Bremner WJ, Millar MR, Sharpe RM, Saunders PT (1994) Immunohistochemical localization of androgen receptors in the rat testis: evidence for stage-dependent expression and regulation by androgens. Endocrinology. 135(3): 1227-34
  188. Van Roijen JH, Van Assen S, Van Der Kwast TH, De Rooij DG, Boersma WJ, Vreeburg JT, Weber RF (1995) Androgen receptor immunoexpression in the testes of subfertile men. J Androl. 16(6): 510-6
  189. Welsh M, Sharpe RM, Moffat L, Atanassova N, Saunders PT, Kilter S, Bergh A, Smith LB (2010) Androgen action via testicular arteriole smooth muscle cells is important for Leydig cell function, vasomotion and testicular fluid dynamics. PLoS One. 5(10): e13632
  190. Johnston DS, Russell LD, Friel PJ, Griswold MD (2001) Murine germ cells do not require functional androgen receptors to complete spermatogenesis following spermatogonial stem cell transplantation. Endocrinology. 142(6): 2405-8
  191. Tsai MY, Yeh SD, Wang RS, Yeh S, Zhang C, Lin HY, Tzeng CR, Chang C (2006) Differential effects of spermatogenesis and fertility in mice lacking androgen receptor in individual testis cells. Proc Natl Acad Sci U S A. 103(50): 18975-80
  192. Lim P, Robson M, Spaliviero J, McTavish KJ, Jimenez M, Zajac JD, Handelsman DJ, Allan CM (2009) Sertoli cell androgen receptor DNA binding domain is essential for the completion of spermatogenesis. Endocrinology. 150(10): 4755-65
  193. O'Donnell L, McLachlan RI, Wreford NG, de Kretser DM, Robertson DM (1996) Testosterone withdrawal promotes stage-specific detachment of round spermatids from the rat seminiferous epithelium. Biol Reprod. 55(4): 895-901
  194. O'Donnell L, Pratis K, Stanton PG, Robertson DM, McLachlan RI (1999) Testosterone-dependent restoration of spermatogenesis in adult rats is impaired by a 5alpha-reductase inhibitor. J Androl. 20(1): 109-17
  195. Meng J, Holdcraft RW, Shima JE, Griswold MD, Braun RE (2005) Androgens regulate the permeability of the blood-testis barrier. Proc Natl Acad Sci U S A. 102(46): 16696-700
  196. McCabe MJ, Allan CM, Foo CF, Nicholls PK, McTavish KJ, Stanton PG (2012) Androgen Initiates Sertoli Cell Tight Junction Formation in the Hypogonadal (hpg) Mouse. Biol Reprod. 87(2): 38
  197. Willems A, Batlouni SR, Esnal A, Swinnen JV, Saunders PT, Sharpe RM, Franca LR, De Gendt K, Verhoeven G (2010) Selective ablation of the androgen receptor in mouse Sertoli cells affects Sertoli cell maturation, barrier formation and cytoskeletal development. PLoS One. 5(11): e14168
  198. Zhengwei Y, Wreford NG, Royce P, de Kretser DM, McLachlan RI (1998) Stereological evaluation of human spermatogenesis after suppression by testosterone treatment: heterogeneous pattern of spermatogenic impairment. J Clin Endocrinol Metab. 83(4): 1284-91
  199. Weinbauer GF, Schlatt S, Walter V, Nieschlag E (2001) Testosterone-induced inhibition of spermatogenesis is more closely related to suppression of FSH than to testicular androgen levels in the cynomolgus monkey model (Macaca fascicularis). J Endocrinol. 168(1): 25-38
  200. Narula A, Gu YQ, O'Donnell L, Stanton PG, Robertson DM, McLachlan RI, Bremner WJ (2002) Variability in sperm suppression during testosterone administration to adult monkeys is related to follicle stimulating hormone suppression and not to intratesticular androgens. J Clin Endocrinol Metab. 87(7): 3399-406
  201. Allan CM, Handelsman DJ (2005) Transgenic models for exploring gonadotropin biology in the male. Endocrine. 26(3): 235-39
  202. Heckert L, Griswold MD (1993) Expression of the FSH receptor in the testis. Recent Prog Horm Res. 48: 61-77
  203. Rannikko A, Penttila TL, Zhang FP, Toppari J, Parvinen M, Huhtaniemi I (1996) Stage-specific expression of the FSH receptor gene in the prepubertal and adult rat seminiferous epithelium. J Endocrinol. 151(1): 29-35
  204. Allan CM, Garcia A, Spaliviero J, Zhang FP, Jimenez M, Huhtaniemi I, Handelsman DJ (2004) Complete Sertoli cell proliferation induced by follicle-stimulating hormone (FSH) independently of luteinizing hormone activity: evidence from genetic models of isolated FSH action. Endocrinology. 145(4): 1587-93
  205. O'Donnell L, Narula A, Balourdos G, Gu YQ, Wreford NG, Robertson DM, Bremner WJ, McLachlan RI (2001) Impairment of spermatogonial development and spermiation after testosterone-induced gonadotropin suppression in adult monkeys (Macaca fascicularis). J Clin Endocrinol Metab. 86(4): 1814-22
  206. McLachlan RI, O'Donnell L, Meachem SJ, Stanton PG, de K, Pratis K, Robertson DM (2002) Hormonal regulation of spermatogenesis in primates and man: insights for development of the male hormonal contraceptive. J Androl. 23(2): 149-62
  207. Meachem SJ, Wreford NG, Stanton PG, Robertson DM, McLachlan RI (1998) Follicle-stimulating hormone is required for the initial phase of spermatogenic restoration in adult rats following gonadotropin suppression. J Androl. 19(6): 725-35
  208. Matsumoto AM, Karpas AE, Paulsen CA, Bremner WJ (1983) Reinitiation of sperm production in gonadotropin-suppressed normal men by administration of follicle-stimulating hormone. J Clin Invest. 72(3): 1005-15
  209. Matsumoto AM, Paulsen CA, Bremner WJ (1984) Stimulation of sperm production by human luteinizing hormone in gonadotropin-suppressed normal men. J Clin Endocrinol Metab. 59(5): 882-7
  210. Bremner WJ, Matsumoto AM, Sussman AM, Paulsen CA (1981) Follicle-stimulating hormone and human spermatogenesis. J Clin Invest. 68(4): 1044-52
  211. Kumar R (2013) Medical management of non-obstructive azoospermia. Clinics (Sao Paulo). 68 Suppl 1: 75-9
  212. McLachlan RI (2013) Approach to the patient with oligozoospermia. J Clin Endocrinol Metab. 98(3): 873-80
  213. Handelsman DJ. (2015) Male contraception, in www.ENDOTEXT.org, Endocrinology of Male Reproduction, Section Editor McLachlan R.I. MDTEXTCOM Inc.: South Dartmouth, MA