Archives

Osteoporosis: Clinical Evaluation

ABSTRACT

 

The identification of a patient at high-risk of fracture should be followed by evaluation for factors contributing to low bone mass, skeletal fragility, falls, and fractures. Components of the evaluation include a bone density test, osteoporosis-directed medical history and physical exam, laboratory studies, and possibly skeletal imaging. A bone density test with dual-energy X-ray absorptiometry (DXA) is useful for diagnostic classification, assessment of fracture risk, and establishing a baseline for monitoring the skeletal effects of treatment. FRAX is a fracture risk algorithm that includes input of femoral neck bone mineral density measured by DXA. The DXA T-score, prior fracture history, and FRAX estimation of fracture risk are used with clinical practice guidelines to determine whether treatment is indicated. The medical history may reveal underlying causes of osteoporosis (e.g., nutritional deficiencies, gastric surgery, medications with adverse skeletal effects) and important risk factors for fracture (e.g., past history of fracture, family history of osteoporosis, or recent falls). Physical exam may show skeletal deformities due to unrecognized fractures (e.g., loss of height, kyphosis, or diminished rib-pelvis space), identify possible secondary causes of skeletal fragility (e.g., blue sclera with osteogenesis imperfecta, urticarial pigmentosa with systemic mastocytosis, dermatitis herpetiformis with celiac disease, or bone tenderness with osteomalacia), and help to recognize patients with poor balance and frailty that might lead to falls. Laboratory studies may show potentially reversible abnormalities (e.g., vitamin D deficiency, hypocalcemia, or impaired kidney function) that must be assessed and corrected, if possible, before starting pharmacological therapy. Disorders other than osteoporosis, requiring other types of treatment, may be found; for example, low serum alkaline phosphatase suggests hypophosphatasia, M-component may be due to myeloma, or hypocalciuria due to malabsorption with celiac disease. There are important safety considerations that can be derived from a pre-treatment assessment, as well. A patient with a blood clotting disorder should not be treated with raloxifene, a history of esophageal stricture is a contraindication for oral bisphosphonates, and previous skeletal radiation therapy precludes treatment with teriparatide or abaloparatide. Skeletal imaging may be helpful when a fracture, malignancy, or Paget’s disease of bone is suspected. Bone biopsy is rarely performed in clinical practice, but may be helpful in some situations, such as when it is necessary to determine the underlying bone disease in a patient with severe chronic kidney disease.

 

INTRODUCTION

 

Osteoporosis is a common systemic skeletal disease characterized by low bone strength that results in an increased risk of fracture (1). Fractures are associated with serious clinical consequences, including pain, disability, loss of independence, and death, as well as high healthcare costs. Early identification and intervention with patients at high risk for fracture is needed to reduce the burden of osteoporotic fractures (2). Management of a patient with a confirmed diagnosis of osteoporosis or low bone mass (osteopenia) includes assessment of fracture risk, evaluation for secondary causes of skeletal fragility, decisions on initiation of treatment, and identification of all relevant clinical factors that may influence patient management. This is a review of the key components in the care of patients prior to treatment.

 

DIAGNOSIS OF OSTEOPOROSIS

 

The World Health Organization (WHO) diagnostic classification (Table 1) (3) is made by bone mineral density (BMD) testing with dual-energy X-ray absorptiometry (DXA) using the T-score, calculated by subtracting the mean BMD (in g/cm2) of a young-adult reference population from the patient’s BMD and dividing by the standard deviation (SD) of the young-adult reference population. The International Society for Clinical Densitometry (ISCD) recommends that BMD be measured at the lumbar spine (ideally L1-L4), total hip, and femoral neck, with the 33% radius (1/3 radius) being measured when the lumbar spine and/or hip cannot be measured (e.g., obese patient who exceeds weight limit of table), is invalid (e.g., patient with lumbar laminectomy or bilateral total hip replacements), or when the patient has hyperparathyroidism (4). Osteoporosis cannot be diagnosed by BMD measurement at skeletal sites other than lumbar spine, total hip, femoral neck, and 33% radius or with technologies other than DXA, except for total hip and femoral neck T-scores calculated from 2D projections of quantitative computed tomography (QCT) data. The quality of DXA instrument maintenance, acquisition, analysis, interpretation, and reporting is important in obtaining valid results that can be used for making appropriate clinical decisions (4-6). In a patient with a fragility fracture, a clinical diagnosis of osteoporosis may be considered independently of BMD results, assuming other causes of skeletal fragility (e.g., osteomalacia, multiple myeloma) are not responsible for the fracture. Establishing a diagnosis of osteoporosis is clinically useful because it facilitates communication among healthcare providers and patients concerning a disease with potentially serious consequences; in some countries, such as the United States (US), a diagnosis is necessary in order to select a numerical code for submission of insurance claims for reimbursement for medical services. The US Bone Health & Osteoporosis Foundation (BHOF) (7) recommends that osteoporosis be diagnosed in postmenopausal women and men over the age ≥ 50 years in any of the following circumstances: T-score ≤ −2.5 at the lumbar spine, femoral neck, total hip, or 33% radius; low-trauma fracture of the hip, spine, and/or forearm; or T-score between -1.0 and -2.5 with FRAX 10-year probability of major osteoporotic fracture ≥ 20% or 10-year probability of hip fracture ≥ 3%.

 

Table 1. World Health Organization Criteria for Classification of Patients with Bone Mineral Density Measured by Dual-Energy X-ray Absorptiometry (3)

Classification

T-score

Normal

-1.0 or greater

Low bone mass (osteopenia)

Between 1-.0 and -2.5

Osteoporosis

-2.5 and below

Severe osteoporosis

-2.5 and below + fragility fracture

 

The BHOF indications for BMD testing in the US (7), which are similar to the ISCD Official Positions (4) are listed in Table 2. BMD testing should be done when it is likely to have an influence on patient management decisions. Other organizations and other countries with different economic resources and health care priorities have used a variety of methodologies to develop alternative recommendations (8-10).

 

Table 2. The BHOF Recommends that Bone Mineral Density Testing be Considered at DXA Facilities using Accepted Quality Assurance Procedures for the Following individuals (7).

Women age ≥ 65 years and men age ≥ 70 years

Postmenopausal women and men age 50-69 years, based on risk profile

Postmenopausal women and men age ≥ 50 years with history of adult-age fracture

Adults with a condition (e.g., rheumatoid arthritis, organ transplant) or taking a medication (e.g., glucocorticoids, aromatase inhibitors, androgen deprivation therapy) associated with low bone mass or bone loss

 

FRACTURE RISK ASSESSMENT

 

There is a robust correlation between BMD and fracture risk, with approximately a 2-fold increase in fracture risk for every 1 SD decrease in BMD (11). However, many or most patients with a hip fracture have a T-score better than -2.5 (12); although fracture risk is higher in patients with very low BMD, there are numerically many more patients with a T-score better than -2.5 than with a T-score ≤ -2.5, therefore there are numerically more fractures in those with higher T-scores. The presence of clinical risk factors (CRFs) that are independent of BMD, particularly advancing age, prior fracture, and recency/number/severity of fracture(s), can identify patients at high-risk for fracture by providing information on fracture risk that is complementary to BMD. The BHOF has provided an extensive list of CRFs (summarized in Table 3) for osteoporosis and fractures. Since most fractures occur with a fall, it is helpful to recognize risk factors for falling (summarized in Table 4) so that appropriate interventions can be made, when possible, to reduce the chances of falling.

 

Table 3. Conditions, Diseases, and Medications that Cause or Contribute to Osteoporosis and Fractures (adapted from guidelines of the BHOF (7)).

Lifestyle Factors

Low Calcium Intake

Vitamin D Insufficiency

Excess Vitamin A

Excessive Thinness

High Salt Intake

Immobilization

Inadequate Physical Activity

 

Smoking

Frequent Falling

 

 

Genetic Factors

Cystic Fibrosis

Homocystinuria

Osteogenesis Imperfecta

Ehlers-Danlos Syndrome

Hypophosphatasia

Gaucher’s Disease

Idiopathic Hypercalciuria

Porphyria

Glycogen storage diseases

Marfan Syndrome

Riley-Day Syndrome

Hemochromatosis

Menkes Steely Hair Syndrome

Parental History of Hip Fracture

Androgen Insensitivity

Turner’s & Klinefelter’s Syndromes

Endocrine Disorders

Obesity

Diabetes Mellitus

Hyperthyroidism

Cushing’s Syndrome

Hyperparathyroidism

Hypogonadism

Panhypopituitarism

Female athlete triad

Anorexia Nervosa

Hyperprolactinemia

Premature Menopause

Androgen Insensitivity

Gastrointestinal Disorders

Celiac Disease

Inflammatory Bowel Disease

Primary Biliary Cirrhosis

Gastric Bypass

Malabsorption

GI Surgery

Pancreatic Disease

 

Hematologic Disorders

Hemophilia

Monoclonal Gammopathies

Systemic Mastocytosis

Leukemia

Lymphoma

Sickle Cell Disease

Thalassemia

Multiple Myeloma

Rheumatic and Autoimmune Diseases

Ankylosing Spondylitis

Systemic Lupus

Rheumatoid Arthritis

Multiple Sclerosis

Muscular Dystrophy

Parkinson’s Disease

Spinal Cord Injury

Stroke

Epilepsy

 

 

 

Miscellaneous Conditions and Diseases

Chronic Obstructive Pulmonary Disease

Weight Loss

Amyloidosis

End Stage Renal Disease

Parenteral Nutrition

Chronic Metabolic Acidosis

Hyponatremia

Post-Transplant Bone Disease

Congestive Heart Failure

Idiopathic Scoliosis

Prior Fracture as an Adult

Depression

HIV/AIDS

Sarcoidosis

 

 

Medications

Anticoagulants (heparin)

Cancer Chemotherapy

Gonadotropin Releasing Hormone Agonists

Anticonvulsants

Lithium

Aromatase Inhibitors

Depo-medroxyprogesterone

Barbiturates

Glucocorticoids (> 5mg of prednisone or equivalent for > 3 months)

Cyclosporine A

Tacrolimus

Aluminum-containing Antacids

Proton Pump Inhibitors

Tamoxifen (premenopausal)

Selective Serotonin Reuptake Inhibitors

Thiazolidinediones

 

Table 4. Risk Factors for Falls Adapted from Guidelines of the BHOF (7).

Environmental Risk Factors: lack of assistive devices in bathrooms, loose throw rugs, low level lighting, obstacles in the walking path, stairs, slippery outdoor conditions

Medical Risk Factors: advanced age, anxiety and agitation, arrhythmias, dehydration, depression, female gender, impaired transfer and mobility, malnutrition, orthostatic hypotension, poor vison and use of bifocals, previous fall, reduced mental acuity and diminished cognitive skills, urgent urinary incontinence, Vitamin D insufficiency (serum 25-OH-D < 30 ng/mL [75 nmol/L]), medications causing over-sedation (narcotic analgesics, anticonvulsants, psychotropics), diabetes

Neurological and Musculoskeletal Risk Factors: kyphosis, poor balance, reduced proprioception, weak muscles

Psychological Risk Factors: fear of falling

The presence of any of these risk factors should trigger consideration of further evaluation and treatment to reduce the risk of falls and fall-related injuries.

 

VERTEBRAL FRACTURE ASSESSMENT (VFA)

 

VFA is a method for imaging the thoracic and lumbar spine by DXA for the purpose of detecting vertebral fracture deformities. Identification of a previously unrecognized vertebral fracture may alter diagnostic classification, change estimation of fracture risk, and influence treatment decisions (13). VFA compares favorably with standard radiographs of the spine, with good correlation for detecting moderate (grade 2) and severe (grade 3) vertebral fractures, a smaller dose of ionizing irradiation, greater patient convenience (i.e., it may be done at the same visit and with the same instrument as BMD testing by DXA), and lower cost. In a study of women age 65 years and older, using the Genant semi-quantitative (SC) method of classifying vertebral deformities (14), the sensitivity of VFA for diagnosing moderate and severe vertebral fractures was 87-93%, with a specificity of 93-95% (15). Indications for vertebral imaging are listed in Table 5. Optimal use of DXA and VFA requires training and adherence to well established quality standards (4).

 

Table 5. ISCD Indications for Lateral Spine Imaging by Standard Radiography or Vertebral Fracture Assessment (VFA) (4)

Vertebral imaging is indicated when the T-score is < -1.0 and one or more of the following is present:

Women ≥ 70 years of age or men ≥ 80 years of age

Historical height loss > 4 cm (1.5 inches)

Self-reported but undocumented prior vertebral fracture

Glucocorticoid therapy equivalent to ≥ 5 mg of prednisone or equivalent per day for ≥ 3 months

 

QUALITY OF DXA AND VFA

 

DXA and VFA should be performed by well-trained and experienced staff operating an instrument that has been maintained and calibrated according to the manufacturer’s standards. Precision assessment and least significant change (LSC) calculation by each DXA technologist are required in order to make quantitative comparisons of serial BMD measurements. The LSC is the smallest change in BMD that is statistically significant, usually with a 95% level of confidence. The use of the correct scan modes, proper patient positioning, consistent vertebral body labeling, and bone edge detection are among the essential elements for serial comparisons of BMD. VFA should be done by a technologist properly trained in acquisition techniques and interpreted by a clinician familiar with methods of diagnosing vertebral fractures using this technology. Bone densitometry facilities should be supervised by a clinician who knows current methods for BMD measurement and fully understands the standards for quality control, interpretation, and reporting of the findings. Poor quality studies may result in inappropriate clinical decisions, generate unnecessary healthcare expenses, and be harmful to patients (5). Assurances of high quality DXA can be attained through education, training, and certification of DXA technologists and interpreters by organizations such as the ISCD. DXA facilities should understand and adhere to ISCD Official Positions (4) and DXA Best Practices (6, 14); facility accreditation (15) provides assurance of adherence to DXA quality standards.

 

TECHNOLOGIES FOR ASSESSMENT OF SKELETAL HEALTH

 

Dual-energy X-ray Absorptiometry (DXA)

 

Devices that measure or estimate BMD differ according to their clinical utility, cost, portability, and use of ionizing radiation. DXA is the “gold standard” method for measuring bone density in clinical practice. There is a strong correlation between mechanical strength and BMD measured by DXA biomechanical studies (16). In observational studies of untreated patients, there is a robust relationship between fracture risk and BMD measured by DXA (11). The WHO diagnostic classification of osteoporosis is based primarily on reference data obtained by DXA (3), and femoral neck BMD provides input into the FRAX algorithm. Most randomized clinical trials showing reduction in fracture risk with pharmacological therapy have selected study participants according to BMD measured by DXA (17). There is a relationship between BMD increases with drug therapy and fracture risk reduction (18, 19). Accuracy and precision of DXA are excellent (20). Radiation exposure with DXA is very low (21). BMD of the 33% (one-third) radius, measured either by a dedicated peripheral DXA (pDXA) device or a central DXA instrument with appropriate software, may be used for diagnostic classification with the WHO criteria and to assess fracture risk, but is generally not clinically useful in monitoring the effects of treatment. DXA measures bone mineral content (BMC in grams [g]) and bone area (cm2), then calculates areal BMD in g/cm2 and derives parameters, such as the T-score and Z-score. DXA is used for diagnostic classification, assessment of fracture risk, and for monitoring changes in BMD over time.

 

Quantitative Ultrasound (QUS)

 

QUS devices emit inaudible high frequency sound waves in the ultrasonic range, typically between 0.1 and 1.0 megahertz (MHz). The sound waves are produced and detected by means of high-efficiency piezoelectric transducers, which must have good acoustical contact with the skin over the bone being tested. Technical differences among QUS systems are great, with different instruments using variable frequencies, different transducer sizes, and sometimes measuring different regions of interest, even at the same skeletal site. The calcaneus is the skeletal site most often tested, although other bones, including the radius, tibia, and finger phalanges, can be used. Commercial QUS systems usually measure two parameters- the speed of sound (SOS) and broadband ultrasound attenuation (BUA). A proprietary value, such as the “quantitative ultrasound index” (QUI) with the Hologic Sahara or “stiffness index” with the GE Healthcare Achilles Express, may be calculated from a combination of these measurements. SOS varies according to the type of bone, with a typical range of 3000-3600 meters per second (m/sec) with cortical bone and 1650-2300 m/sec for trabecular bone (22). A higher bone density is associated with a higher SOS. BUA, reported as decibels per megahertz (dB/MHz), is a measurement of the loss of energy, or attenuation, of the sound wave as it passes through bone. As with SOS, a higher bone density is associated with a higher BUA. Values obtained from calculations using ultrasound parameters may be used to generate an estimated BMD and a T-score. The T-score derived from a QUS measurement is not the same as a T-score from a DXA. QUS cannot be used for diagnostic classification and is not clinically useful to monitor the effects of therapy (23).

 

Radiofrequency Echographic Multi Spectrometry (REMS) assesses bone health and fracture risk with an ultrasound scan of the lumbar spine and proximal femur, thereby overcoming the limitation of QUS of only measuring peripheral skeletal sites. REMS technology uses a portable device with a transducer that transmits ultrasound waves to the target axial skeletal site and a receiver that captures the resultant back-scattered waveforms with B-mode image reconstruction of the region of interest. There are studies that support a strong correlation between REMS and DXA measurements of BMD (24). Potential clinical applications include its use in frail patients with limited mobility, bedside measurements in hospitalized patients, and special populations such as pregnant women and children.

 

Quantitative Computed Tomography (QCT) and Peripheral QCT (pQCT)

 

QCT and pQCT measure trabecular and cortical volumetric BMD at the axial skeleton and peripheral skeletal sites, respectively. QCT is a useful research tool to enhance understanding of the pathophysiology of osteoporosis and the mechanism of action of pharmacological agents used to treat osteoporosis. QCT predicts fracture risk, with the correlation varying according to skeletal site and bone compartment measured, type of fracture predicted, and population assessed (4). The ISCD Official Positions state that “spinal trabecular BMD as measured by QCT has at least the same ability to predict vertebral fractures as AP spinal BMD measured by central DXA in postmenopausal women with lack of sufficient evidence to support this position in men; pQCT of the forearm at the ultra-distal radius predicts hip, but not spine, fragility fractures in postmenopausal women with lack of sufficient evidence to support this position in men (4).” QCT is more expensive than DXA and QUS and uses higher levels of ionizing radiation than DXA. T-scores by QCT are typically lower than with DXA (27), thereby overestimating the prevalence of osteoporosis, with the exception of total hip and femoral neck T-scores calculated from 2D projections of QCT data, which are similar to DXA-derived T-scores at the same regions of interest and may be used for diagnosis of osteoporosis in accordance with the WHO criteria. T-scores and femoral neck BMD derived from 2D projections of QCT data may also be used as input for the FRAX algorithm to estimate 10-year fracture probabilities.

 

Other Technologies of Interest

 

Pulse-echo ultrasonography (PEUS) uses a portable handheld ultrasound device to estimate the thickness of cortical bone at peripheral skeletal sites. When connected to a computer with proprietary software, a value can be generated that that is correlated with hip BMD measured by DXA, with the potential benefit of identifying patients who are likely or unlikely to have osteoporosis (25).  

 

Biomechanical CT (BCT) is an opportunistic analysis of data from pre-existing CT scans of the hip and/or spine that provides DXA-equivalent T-scores for the hip, QCT-equivalent vBMD at the spine, and an estimate of bone strength with finite element analysis (FEA) (26).  

 

3D-Shaper is software that can be added to a DXA system using statistical modelling to reconstruct the 3D shape and density distribution of the proximal femur from 2D DXA data scans. A recent study found that this technology provided an estimation of femur strength that was similar to that derived from QCT FEA (27).

 

FRACTURE RISK ASSESSMENT TOOL (FRAX® and FRAXplus)

 

The combination of BMD and clinical risk factors (CRFs) predicts fracture risk better than BMD or CRFs alone (28,29) (2). A fracture risk assessment tool (FRAX) combines CRFs and femoral neck BMD in a computer-based algorithm that estimates the 10-year probability of hip fracture and major osteoporotic fracture (i.e., clinical spine, hip, proximal humerus, and distal forearm fracture). FRAX can be accessed online at http://www.shef.ac.uk/FRAX (Figure 1), on most software versions of DXA systems, and on smartphones. FRAX is based on analysis of data from 12 large prospective observational studies in about 60,000 untreated men and women in different world regions, having over 250,000 person-years of observation and more than 5,000 reported fractures reported.

Figure 1. FRAX online for US Caucasian patients. This example shows a 65-year-old woman who has no clinical risk factors for fracture and a femoral neck BMD of 0.582 g/cm2 with a Hologic instrument. The 10-year probability of major osteoporotic fracture is 11% and the 10-year probability of hip fracture is 2.2%. These levels do not meet the Bone Health & Osteoporosis Foundation guidelines for initiation of pharmacological therapy in the US (7). Image reproduced with permission from Eugene McCloskey, University of Sheffield, Sheffield, UK.

 

The input for FRAX is the patient’s age, sex, height, weight, a “yes” or “no” response indicating the presence or absence for each of 7 CRFs: 1. previous ‘spontaneous’ or fragility fracture as an adult; 2. parent with hip fracture; 3. current tobacco smoking; 4. ever use of chronic glucocorticoids at least 5 mg prednisolone for at least 3 months; 5. confirmed rheumatoid arthritis; 6. secondary osteoporosis, such as type 1 diabetes, osteogenesis imperfecta in adults, untreated longstanding hyperthyroidism and hypogonadism, or premature menopause (note: this is a “dummy” risk factor that has no effect on the fracture risk calculation unless no femoral neck BMD value is entered); 7. alcohol intake greater than 3 units per day, with a unit of alcohol defined as equivalent to a glass of beer, an ounce of spirits or a medium-sized glass of wine), and if available, femoral neck BMD and trabecular bone score (TBS). Since the introduction of FRAX, upgrades have been introduced to correct errors, enhance its usability, and incorporate new data that have become available.

 

Benefits of FRAX

 

The use of FRAX provides a quantitative estimation of fracture risk that is based on robust data in large populations of men and women with ethnic and geographic diversity. Expression of fracture risk as a probability provides greater clinical utility than relative risk. When combined with cost-utility analysis, a fracture risk level at which it is cost-effective to treat may be derived. FRAX can be used to estimate fracture probability without femoral neck BMD, allowing it to be used when DXA in unavailable or inaccessible. FRAX is incorporated into many clinical practice guidelines.

 

Limitations of FRAX

 

To generate a valid FRAX output, the responses to CRF questions must be correct; for example, an incorrect entry of self-reported rheumatoid arthritis or use of glucocorticoids could skew the results toward overestimation of fracture risk. FRAX may underestimate or overestimate fracture risk due to dichotomized (yes or no) input for CRFs that in reality are associated with a range of risk that varies according to dose, duration of exposure, or severity; for example, fracture risk may be underestimated when a patient is on high-dose glucocorticoid therapy or has had multiple recent fragility fractures, even when a “yes” response is entered for these CRFs. FRAX is validated only in untreated patients and may overestimate fracture risk when the patient is being treated; the NOF(BHOF)/ISCD guidance on FRAX suggests that “untreated” may be interpreted as never treated or if previously treated, no bisphosphonate for the past 2 years (unless it is an oral agent taken for less than 2 months); and no estrogen, raloxifene, calcitonin, or denosumab for the past 1 year (7). In this context, calcium and vitamin D do not constitute treatment. FRAX in the US allows input for 4 ethnicities (Caucasian, Black, Hispanic, Asian); it is not clear how to use FRAX for patients of other ethnicities or a mix of these ethnicities. Answering “yes” for the category of secondary osteoporosis has no effect on the fracture risk calculation as long as a value for femoral neck BMD is entered. The range of error for a fracture probability generated by FRAX is unknown but may be substantial in some cases.

 

Some important risk factors, such as falls and frailty, are not directly entered into FRAX, although they are indirectly included insofar as they are a component of aging. FRAX may underestimate fracture risk when the lumbar spine BMD is substantially lower than femoral neck BMD, as may occur in about 15% of patients (30). Despite the limitations of FRAX, it is a helpful clinical tool when used with a good understanding of factors that may result in underestimation or overestimation of fracture risk. FRAX may enhance discussion of risk with the patient and help to identify those who are at sufficiently high for fracture to benefit from therapy.

 

FRAXplus

 

FRAXplus (https://www.fraxplus.org/) is an updated version of FRAX that addresses some of the limitations of traditional FRAX, allowing input for these additional rick factors: recency of osteoporotic fracture, high exposure to oral glucocorticoids, type 2 diabetes, concurrent data on lumbar spine BMD, trabecular bone score, falls history, and hip axis length. For patients with fracture risk that is close to the intervention threshold for the applicable clinical practice guideline, the use of an additional risk factor with FRAXplus might influence the decision to treat or not treat with a pharmacological agent.

 

MEDICAL HISTORY

 

A thorough medical history may identify risk factors for osteoporosis and fractures, suggesting that a bone density test and/or further evaluation is indicated. The medical history may also reveal symptoms of potentially correctable causes of skeletal fragility (e.g., gluten intolerance with celiac disease) or co-morbidities that could influence treatment decisions (e.g., esophageal stricture suggests that oral bisphosphonates should not be given). A history of falls is a predictor of future falls, with that risk potentially modifiable though appropriate interventions. Finally, some symptoms may trigger further evaluation for the presence of fractures (e.g., historical height loss or development of kyphotic posture suggests the possibility of vertebral fractures that may warrant spine imaging). Table 6 provides examples of helpful information that might be obtained from a thoughtful interactive discussion with the patient.

 

Medical History for Patients with Osteoporosis

 

A thorough review of systems and history of relevant familial disorders, previous surgical procedures, medications, dietary supplements, food intolerances, and lifestyle provides helpful information in the management of patients with osteoporosis. Such historical information may play a role in determining who should have a bone density test, assessing fracture risk, providing input for FRAX, evaluating for secondary causes of osteoporosis, selecting the most appropriate treatment to reduce fracture risk, and finding factors contributing to suboptimal response to therapy. Listed here are key components of the skeletal health history and examples of the potential impact on patient care.

 

Table 6. Clinical Utility of the Medical History

Clinical Utility

Medical History

Assist in determining who need a bone density test

See Table 3

Assessing fracture risk

See Table 3 and 4

Input for FRAX

Age, sex, weight, height, previous fracture, parent with hip fracture, current tobacco smoking, ever use of glucocorticoids, rheumatoid arthritis, secondary osteoporosis, alcohol intake 3 or more units per day, and if available, femoral neck bone mineral density and trabecular bone score

Evaluating for secondary causes of osteoporosis

See Table 3

Selecting most appropriate treatment

Identify co-morbidities of clinical significance. For example, high-risk of breast cancer favors raloxifene use, while history of thrombophlebitis suggests that raloxifene should not be used; esophageal stricture is a contraindication for oral bisphosphonate use; a patient with a skeletal malignancy should not be treated with teriparatide.

Factors contributing to suboptimal response to therapy

Compliance and persistence to therapy; adequacy of calcium and vitamin D; comorbidities listed in Table 3.

 

PHYSICAL EXAM

 

Findings of importance on the physical exam of a patient with osteoporosis may be the sequelae of old fractures (e.g., kyphosis due to old vertebral fractures), a consequence of a recent fracture (e.g., localized vertebral spinous process tenderness with a new vertebral fracture), or abnormalities suggestive of a secondary cause of osteoporosis (e.g., thyromegaly with thyrotoxicosis). An accurate measurement of height with a wall-mounted stadiometer is a helpful office tool for evaluating patients at risk for fracture. A height loss of 1.5 inches (4.0 cm) or more compared to the historical maximum (28, 29) or a loss of 0.75 inches (2.0 cm) or more compared to a previous measured height (30) suggests a high likelihood of vertebral fracture. Body weight measurement is part of the osteoporosis evaluation because low body weight (less than 127 lbs) (31), low BMI (20 kg/m2 or less) (32), and weight loss of 5% or more ((33)36) are associated with increased risk of fracture. Localized tenderness of the spine, kyphosis, or diminished distance between the lower ribs and the pelvic brim may be the result of one or more vertebral fractures. Abnormalities of gait, posture, balance, muscle strength, or the presence of postural hypotension or impaired level of consciousness may be associated with increased risk of falling. Bone tenderness may be caused by osteomalacia. Atrophic testicles suggest hypogonadism. Patients should be observed for stigmata of hyperthyroidism or Cushing’s syndrome. Blue sclera, hearing loss, and yellow-brown teeth are suggestive of osteogenesis imperfecta. Joint hypermobility and skin fragility could be due to Ehlers-Danlos syndrome. Urticaria pigmentosa may occur with systemic mastocytosis. Table 7 shows examples of abnormal physical exam findings with osteoporosis.

 

Table 7. Focused Physical Examination in a Patient with Osteoporosis

Component of physical exam

Example of finding of potential skeletal importance

Potential clinical implications for skeletal health

Vital signs

Low body weight or body mass index

Anorexia nervosa

Loss of height

Vertebral fracture

Loss of weight

Malignancy, malabsorption

Skin

Urticaria pigmentosa

Dermatitis herpetiformis

Systemic mastocytosis

Celiac disease

Striae, acne

Cushing’s syndrome, exogenous glucocorticoids

Head

Cranial dysostosis

Hypophosphatasia

Eyes

Blue sclera

Osteogenesis imperfect

Ears

Hearing loss

Osteogenesis imperfecta, sclerosteosis

Nose

Anosmia

Kallmann syndrome

Throat

Poor dentition

Increased risk of osteonecrosis of the jaw

Neck

Thyromegaly

Thyrotoxicosis

Lungs

Decreased breath sounds

Chronic obstructive pulmonary disease

Heart

Aortic insufficiency

Marfan’s syndrome

Musculoskeletal

Kyphosis

Vertebral fractures

Spinous process tenderness

Acute vertebral fracture

Decreased space between lower ribs and pelvis

Vertebral fractures

Tender bones

Osteomalacia

Inflammatory joint disease

Rheumatoid arthritis

Hypermobility of joints

Ehlers-Danlos syndrome

Muscle weakness

Vitamin D deficiency, osteomalacia

Abdomen

Hepatomegaly

Chronic liver disease

Surgical scars

Bariatric surgery, gastrectomy

Genitalia

Testicular atrophy

Hypogonadism

Neurological

Poor balance

High fall risk, vitamin D deficiency

Dementia

Poor adherence to therapy, high fall risk

This table provides examples of findings on physical exam that may be helpful in the evaluation of skeletal health. It is not intended to show all findings of importance.

 

EVALUATION FOR SECONDARY CAUSES OF OSTEOPOROSIS

 

The possibility of previously unrecognized causes of skeletal fragility should be considered in every patient with osteoporosis (34), understanding that some patients with a T-score ≤ -2.5 may have a skeletal disease other than osteoporosis and some patients with osteoporosis have contributing disorders and conditions other than estrogen deficiency and advancing age that can be corrected. Collectively, these contributing factors are sometimes called secondary causes of osteoporosis. After an initial medical history is taken and physical exam is performed, appropriate laboratory testing and imaging may provide information that is critical for ongoing patient care.

 

The reported prevalence of secondary osteoporosis varies depending on the study population, the extent of the medical evaluation, and definitions for laboratory abnormalities. It is likely that many or most patients with osteoporosis have clinically significant contributing factors that may influence patient management. In a study of North American women receiving osteoporosis therapy, it was found that 52% had vitamin D inadequacy, defined as serum 25-hydroxyvitamin D (25-OH-D) levels less than 30 ng/ml (35). In another study of patients referred to an osteoporosis clinic, over 60% were found to have elements of secondary osteoporosis when vitamin D deficiency was very conservatively defined as serum 25-OH-D level less than 12.5 ng/ml (36, 37). In the same study, the number of patients with secondary osteoporosis was much higher when vitamin D inadequacy was more appropriately defined as serum 25-OH-D less than 33 ng/ml (38, 39).

 

It has been proposed by some that a bone density that is less than expected compared to an age- and sex-matched population, as represented by a low Z-score (e.g., less than -2.0), suggests a high likelihood of secondary osteoporosis and should be one of the triggers for further investigation (40, 41). While there may be some merit to this concept, there are few if any studies validating the use of a Z-score cutoff for this purpose. Since secondary causes of osteoporosis are common, a more effective strategy is to screen all patients with osteoporosis for contributing factors (42). The results of a metabolic evaluation may identify previously unrecognized diseases and conditions that require treatment in addition to, or instead of, standard osteoporosis pharmacological therapy.

 

Depending on the patient population being studied, different causes of secondary osteoporosis may predominate. Calcium deficiency, vitamin D deficiency, and sedentary lifestyle are common contributing factors for all patients. In women referred to an osteoporosis clinic with previously recognized medications or diseases contributing to osteoporosis, the most common were history of glucocorticoid use (36%), premature ovarian failure (21%), history of unintentional weight loss (10%), history of alcoholism (10%), and history of liver disease (10%) (36). When patients without previously recognized contributing factors were evaluated at the same specialty clinic, most (55%) were found to have vitamin D deficiency or insufficiency (serum 25-OH-D less than 33 ng/ml) (39), while 10% had hypercalciuria, 8% had malabsorption, and 7% had primary or secondary hyperparathyroidism (36). In men, the most common secondary causes of osteoporosis are long-term glucocorticoid use, hypogonadism, and alcoholism (43, 44). The increasing use of aromatase inhibitor therapy for breast cancer in women and androgen deprivation therapy for prostate cancer in men (45) is now recognized as an important factor in the development of osteoporosis in these patients. Other common causes for low BMD and fractures include multiple myeloma (46), gastric bypass surgery (47) and gastric resection (48). Treatable but easily missed secondary causes of osteoporosis include asymptomatic primary hyperparathyroidism (49), subclinical hyperthyroidism (50), mild Cushing’s syndrome (51), and malabsorption due to unrecognized celiac disease (52). Table 8 lists some of the causes of low BMD by category.

 

Table 8. Causes of Low Bone Mineral Density

Inherited

Nutritional

Endocrine

Drugs

Other

Osteogenesis imperfecta

Malabsorption

Hypogonadism

Glucocorticoids

Multiple myeloma

Homocystinuria

Chronic liver disease

Hyperthyroidism

Anticonvulsants

Rheumatoid arthritis

Marfan’s syndrome

Alcoholism

Hyperparathyroidism

Long-term heparin

Systemic mastocytosis

Hypophosphatasia

Calcium deficient diet

Cushing’s syndrome

Excess thyroid

Immobilization

 

Vitamin D deficiency

Eating disorder

GnRH agonists

 
     

Aromatase inhibitors

 

 

Although a variety of testing strategies have been proposed as screening for all patients with osteoporosis, a minimal cost-effective work-up for all patients consists of a complete blood count (CBC), serum calcium, phosphorus, creatinine with calculated or measured creatinine clearance, alkaline phosphatase, 24-hour urinary calcium, and serum 25-OH-D. Other laboratory tests may be indicated according to the patient’s clinical profile and the practice setting. A summary of useful common and uncommon laboratory studies with comments on their possible skeletal significance is provided below.

 

CLINICAL CASE

 

A 52-year-old postmenopausal woman with a history of irritable bowel syndrome (IBS) and a family history of osteoporosis (mother with hip fracture) is found to have osteoporosis on a DXA study. Evaluation for secondary causes of osteoporosis is unremarkable except for mild iron deficiency anemia (a long-standing problem, previously attributed to heavy menses) and a low 24-hour urinary calcium of 30 mg, with adequate calcium intake and normal renal function. Serum 25-OH-D is 29 ng/ml. Additional work-up shows a high titer of IgA endomysial antibodies consistent with celiac disease. This diagnosis is confirmed by a small bowel biopsy showing villous atrophy. She is started on a gluten-free diet, resulting in resolution of her “IBS” symptoms and correction of her anemia. One year later, with no pharmacological therapy for osteoporosis, there is a statistically significant BMD increase of 9% at the lumbar spine.

 

Celiac disease may result in osteoporosis due to calcium malabsorption, even in the absence of gastrointestinal symptoms. Treatment is strict lifelong adherence to a gluten-free diet, which may sometimes be followed by a substantial increase in BMD, as seen in this patient. A 24-hour urinary calcium is an inexpensive screening test for calcium malabsorption that should be considered a routine part of the initial evaluation of osteoporosis.

 

BASIC BLOOD TESTS

 

CBC- Anemia may be seen in patients with myeloma or malnutrition

 

Sedimentation rate- May be elevated with myeloma and rheumatic diseases.

 

Calcium- Among the many causes of hypercalcemia are primary and secondary hyperparathyroidism, hyperthyroidism, renal failure, vitamin D intoxication, and Paget’s disease of bone. Hypocalcemia may be seen with vitamin D deficiency and hyperphosphatemia.

 

Phosphorus- Hyperphosphatemia may occur with hypoparathyroidism, renal failure, and possibly with bisphosphonate therapy. Hypophosphatemia may be seen with primary or secondary hyperparathyroidism, vitamin D deficiency, tumor induced osteomalacia, and X-linked hypophosphatemia.

 

Alkaline phosphatase- High values can be seen with healing fractures, osteomalacia, and Paget’s disease, as well as occurring normally in growing children. Low values occur with hypophosphatasia, a rare genetic disorder that causes impaired mineralization of bone and dental tissue.

 

Vitamin D- The test that best reflects vitamin D stores is the serum 25-OH-D. While there is no consensus on the optimal range of serum 25-OH-D, a reasonable target for good skeletal health is approximately 30-50 ng/ml. This is likely to maximize intestinal absorption of calcium and minimize serum PTH levels. Interpretation of serum 25-OH-D levels is confounded by assay variability (59). Serum 1,25-(OH)2-D3 is usually not helpful in the evaluation of osteoporosis patients, unless there are concerns regarding renal conversion of 25-OH-D to 1,25-(OH)2-D3. Deficiency or insufficiency of vitamin D is very common and play a role in the pathogenesis of osteoporosis and osteomalacia.

 

Creatinine- Chronic kidney disease may cause an elevated creatinine level and renal osteodystrophy. Elderly patients with small muscle mass may have impaired renal function with a “normal” serum creatinine. An estimated glomerular filtration rate can be calculated using one of many formulae, such as that of Cockcroft and Gault (53) or modification of diet in renal disease study equation (54). Impaired renal function not only has adverse skeletal effects but also raises considerations regarding the type and dose of pharmacologic agents used.

 

TSH- Hyperthyroidism from any cause, including excess thyroid replacement, can usually be recognized by a low TSH. High bone turnover associated hyperthyroidism is associated with loss of bone mass.

 

Liver enzymes- Abnormalities may be caused by chronic liver disease, which is a risk factor for osteoporosis.

 

BASIC URINE TESTS

 

Urinalysis- Proteinuria may occur with multiple myeloma or chronic kidney disease. Abnormal cells may suggest kidney disease.

 

24-hour urine for calcium- A well-collected 24-hour urine for calcium is a helpful screening test for identifying patients with common disorders of calcium metabolism. The “normal” range of urinary calcium is not well established and varies according to many dietary factors and estrogen status in women (55). As a “rule of thumb,” urinary calcium may be considered elevated when it is greater than 250 mg per 24 hours in women; greater than 300 mg per 24 hours in men; or greater than 4 mg/kg body weight per 24 hours in either sex. It has been proposed that hypercalciuria can be easily classified as “renal” (renal calcium leak), “resorptive” (excess skeletal loss of calcium) or “absorptive” (increased intestinal absorption of calcium) (56). However, in clinical practice, these distinctions are not so easily established. Idiopathic hypercalciuria, perhaps the most common type of hypercalciuria (57), may be diagnosed if there are no underlying medical disorders (e.g., hyperparathyroidism, vitamin D toxicity, Paget’s disease of bone, multiple myeloma, sarcoidosis) and no obvious dietary excesses (e.g., calcium, sodium, protein, carbohydrates, alcohol) or deficiencies (e.g., phosphate, potassium) that are associated with hypercalciuria. In the absence of dietary calcium deficiency, vitamin D deficiency, malabsorption, liver disease, or chronic renal failure, low urinary calcium (less than 50 mg per 24 hours in women or men) is suggestive of calcium malabsorption and warrants further investigation. Celiac disease is a common (58) cause of asymptomatic malabsorption in osteoporosis that is treatable with a gluten-free diet.

 

ADDITIONAL STUDIES IN SELECTED PATIENTS

 

Celiac antibodies- Anti-endomysial antibody and tissue transglutaminase antibody are currently the serological markers of choice, with a higher sensitivity and specificity than anti-gliadin antibody and anti-reticulin antibody. If a serological marker is abnormal, or if there is a high clinical suspicion for celiac disease, the patient should be referred for endoscopy and small bowel biopsy.

 

Intact PTH- This may be elevated in patients with primary hyperparathyroidism or with secondary hyperparathyroidism due to disorders such as chronic kidney disease, vitamin D deficiency, or calcium malabsorption.

 

Serum protein electrophoresis and serum kappa/lambda light chain ratio- These are helpful tests to screen for possible multiple myeloma. Abnormal results may require further evaluation by an oncologist.

 

Overnight 1 mg dexamethasone suppression test or 24-hour urinary free cortisol- This is helpful to evaluate patients with suspected Cushing’s syndrome.

 

Serum total or free testosterone level- May be helpful in the assessment of men with osteoporosis.

 

Serum homocysteine- Elevated circulating homocysteine levels are associated with an increased risk of fractures (59). It is unknown whether reduction of homocysteine levels by increasing dietary intake of folic acid and vitamins B6 and B12 reduces the risk of fracture.

 

Serum tryptase and 24-hour urine for N-methylhistamine- Systemic mastocytosis is a rare cause of osteoporosis that can be diagnosed by a biopsy of typical skin lesions of urticaria pigmentosa, when present. Patients with systemic mastocytosis may sometimes present with osteoporosis and no other manifestations of the disease (60). When this disorder is suspected but skin lesions are not present, the finding of an elevated serum tryptase and/or urinary N-methyl histamine can be helpful, especially during or soon after a symptomatic episode of histamine release. However, normal values do not exclude the diagnosis. Bone marrow aspiration or biopsy, or non-decalcified double tetracycline labeled transiliac bone biopsy, may be necessary to confirm the diagnosis.

 

Serum bicarbonate- Renal tubular acidosis (RTA) has been associated with osteoporosis (61). With distal (type I) RTA, the serum bicarbonate is usually less than 15 mmol/l with a urine pH greater than 5.5 despite having systemic acidosis. This is due to the impaired ability of the distal nephron to secrete hydrogen ions effectively, which is a hallmark of the condition.

 

BONE TURNOVER MARKERS

 

Bone turnover markers (BTMs) are noninvasive laboratory tests of serum and urine that are readily available in clinical practice. While BTMs cannot be used to diagnose osteoporosis or determine the cause to osteoporosis, they have been very helpful in research to understand the pathophysiology of osteoporosis and other skeletal diseases and the mechanism of action of interventions used in the treatment of osteoporosis. In clinical practice, BTMs offer the potential of predicting fracture risk independently of BMD and may be useful in monitoring the metabolic effects of therapy (62). Drugs that are approved for the management of osteoporosis modulate bone remodeling in ways that are reflected by changes in BTMs. A decrease in BTMs with antiresorptive therapy is predictive of a subsequent increase in BMD (63)and reduction in fracture risk (64). The magnitude of BTM decrease with antiresorptive therapy is significantly associated with the level of fracture risk reduction, although the proportion of treatment effect due to the reduction in BTMs appears to vary according to the type of drug used (65). Teriparatide and abaloparatide, analogs of PTH and PTHrP, respectfully, are bone forming drugs associated with an increase in bone remodeling, with bone formation markers rising sooner and greater than bone resorption makers. Romosozumab is bone forming drug that uncouples bone resorption and formation, with an initial increase in bone formation markers and decrease in bone resorption markers.

 

Markers of bone resorption are mostly fragments of type I collagen, the main component of the organic bone matrix, which are released during osteoclastic bone resorption. These are measured in the serum or urine, with those available for clinical use including N-telopeptide of type I collagen (NTX), C-telopeptide of type I collagen (CTX), deoxypyridinoline (DPD), and pyridinoline (PYD). Bone formation markers are proteins secreted by osteoblasts or byproducts of type I collagen production by osteoblasts. They are measured in the serum and include bone specific alkaline phosphatase (BSAP), N-terminal propeptide of type I collagen (P1NP), and osteocalcin. CTX and P1NP have been proposed as the reference BTMs for clinical trials (66) and for clinical practice (67).

 

Clinical use of BTMs requires knowledge of their limitations as well as benefits. BTMs are subject to pre-analytical (biological) and analytical variability (62). Uncontrollable sources of pre-analytical variability include age, sex, menopausal status, pregnancy, lactation, fractures, co-existing diseases (e.g., diabetes mellitus, impaired renal function, and liver disease), drugs (e.g., glucocorticoids, anticonvulsants, and gonadotropin hormone releasing agonists), and immobility. Controllable pre-analytical sources of variability include time of day (circadian variability), fasting status, and exercise. Analytical sources of variability include specimen processing (e.g., collection, handling, and storage). Between-laboratory variability may be large (reported to be as much as a 7.3-fold difference), casting doubt on the validity of comparing specimens sent to different labs (68). Reference ranges for BTMs are not well established and may vary according to the population tested, the type of BTM, and the circumstances under which it is collected and processed.

 

In order to compare BTMs measurements longitudinally, it would be ideal to know the least significant change (LSC) and use this in a manner similar to what should be (but is probably not) common practice with DXA. However, the standards for calculating an LSC for a BTM are not as clear as with DXA, and the opportunity to do precision assessment for a BTM may not present itself. The Belgian Bone Club suggests using an estimated LSC by assuming an LSC of about 30% for serum BTMs and about 50-60% for urine BTMs (69). While the LSC for BTMs is almost always greater than for DXA, the magnitude of likely change is greater than DXA, with the “signal to noise ratio” that may be as good or even better than DXA. One strategy for the use of BTMs to monitor patients on antiresorptive therapy is to use absolute values rather that percent changes, as follows: treatment effect can be considered optimal when serum CTX has decreased by 100 ng/L or is below 280 ng/L, or when P1NP has decreased by 10 mcg/L or is less than 35 mcg/L (70).

 

A significant change of a BTM level in the appropriate direction following therapy is evidence that the patient is taking the drug regularly, taking it correctly, and that it is being absorbed and having the expected effect in modulating bone remodeling. Failure to achieve such a change in the BTM level is cause for concern and suggests that evaluation and possibly a reconsideration of treatment strategies. The use of BTMs allows assessment of drug effect sooner than with DXA, so that evaluation and corrective action, if needed, can be taken early in the course of therapy rather than later. Monitoring BTMs, especially in association with regular contact by a healthcare provider, may improve persistence with therapy (70). Despite the well-described limitations of BTMs, there is emerging support for their use in clinical practice, particularly in the assessment of response to therapy (62). Clinicians who are familiar with the benefits and limitations of BTMs may find them a helpful tool, in association with BMD testing, for managing patients with osteoporosis.

 

IMAGING STUDIES

 

Standard X-rays are used to diagnose fractures of all types and may sometimes suggest secondary causes of osteoporosis. Pseudofractures (Looser’s zones) are radiolucent lines running perpendicular to the bone cortex that may be seen in patients with osteomalacia. These probably represent stress fractures that have healed with poorly mineralized osteoid. Punctate radiolucencies may be seen in bone X-rays of patients with systemic mastocytosis. Primary hyperparathyroidism may cause bone cysts, subperiosteal bone resorption, brown tumors, and demineralization (‘salt and pepper’ pattern) of the skull. MRI, CT scanning, or nuclear imaging may be used to detect stress fractures that are not visible on X-ray. MRI of the spine is commonly used prior to vertebroplasty or kyphoplasty to determine the age of the fracture, the likelihood of the fracture being from causes other than osteoporosis, and whether there is retropulsion of bony fragments than could impair neurological function.

 

BONE BIOPSY

 

Non-decalcified double tetracycline labeled iliac crest bone biopsy is rarely used in clinical practice but may be helpful with difficult diagnostic problems. In the evaluation of renal osteodystrophy, a bone biopsy can distinguish between high turnover and low turnover bone disease, and possibly be an aid in the selection of therapy. With infiltrative disorders of bone, such as systemic mastocytosis, a bone biopsy or bone marrow aspiration may sometimes be the only way to make the diagnosis. In patients who are not responding to therapy as expected, or in patients with unusual presentations of osteoporosis, a bone biopsy may be indicated. Bone biopsies are required by the FDA for safety monitoring in clinical trials of osteoporosis drugs.

 

SUMMARY

 

Osteoporosis is a common skeletal disease with serious clinical consequences. Effective management of skeletal health includes appropriate selection of patients for bone density testing and assessment of risk factors for fracture. Prior to treatment, and when response to treatment is suboptimal, patients should be evaluated for secondary causes of osteoporosis. All reversible factors should be corrected and treatment should be individualized based on the clinical circumstances.

 

REFERENCES

 

  1. NIH Consensus Development Panel. Osteoporosis prevention, diagnosis, and therapy. JAMA. 2001;285(6):785-95.
  2. US Department of Health and Human Services. Bone Health and Osteoporosis: A Report of the Surgeon General. Rockville, MD: US Department of Health and Human Services, Office of the Surgeon General; 2004.
  3. WHO Study Group on Assessment of Fracture Risk and its Application to Screening for Postmenopausal Osteoporosis. Technical Report Series 843: Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Geneva: World Health Organization; 1994.
  4. Shuhart C, Cheung A, Gill R, Gani L, Goel H, Szalat A. Executive Summary of the 2023 Adult Position Development Conference of the International Society for Clinical Densitometry: DXA Reporting, Follow-up BMD Testing and Trabecular Bone Score Application and Reporting. J Clin Densitom. 2024;27(1):101435.
  5. Lewiecki EM, Binkley N, Petak SM. DXA Quality Matters. J Clin Densitom. 2006;9(4):388-92.
  6. Lewiecki EM, Binkley N, Morgan SL, Shuhart CR, Camargos BM, Carey JJ, et al. Best Practices for Dual-Energy X-ray Absorptiometry Measurement and Reporting: International Society for Clinical Densitometry Guidance. J Clin Densitom. 2016;19(2):127-40.
  7. LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ, et al. The clinician's guide to prevention and treatment of osteoporosis. Osteoporos Int. 2022;33(10):2049-102.
  8. Qaseem A, Hicks LA, Etxeandia-Ikobaltzeta I, Shamliyan T, Cooney TG, Clinical Guidelines Committee of the American College of P, et al. Pharmacologic Treatment of Primary Osteoporosis or Low Bone Mass to Prevent Fractures in Adults: A Living Clinical Guideline From the American College of Physicians. Ann Intern Med. 2023;176(2):224-38.
  9. Gregson CL, Armstrong DJ, Bowden J, Cooper C, Edwards J, Gittoes NJL, et al. UK clinical guideline for the prevention and treatment of osteoporosis. Archives of osteoporosis. 2022;17(1):58.
  10. Kanis JA, Harvey NC, McCloskey E, Bruyere O, Veronese N, Lorentzon M, et al. Algorithm for the management of patients at low, high and very high risk of osteoporotic fractures. Osteoporos Int. 2020;31(1):1-12.
  11. Marshall D, Johnell O, Wedel H. Meta-analysis of how well measures of bone mineral density predict occurrence of osteoporotic fractures. BMJ. 1996;312(7041):1254-9.
  12. Wainwright SA, Marshall LM, Ensrud KE, Cauley JA, Black DM, Hillier TA, et al. Hip fracture in women without osteoporosis. J Clin Endocrinol Metab. 2005;90(5):2787-93.
  13. Lewiecki EM, Laster AJ. Clinical applications of vertebral fracture assessment by dual-energy X-ray absorptiometry. J Clin Endocrinol Metab. 2006;91(11):4215-22.
  14. Khan AA, Slart R, Ali DS, Bock O, Carey JJ, Camacho P, et al. Osteoporotic Fractures: Diagnosis, Evaluation, and Significance From the International Working Group on DXA Best Practices. Mayo Clin Proc. 2024;99(7):1127-41.
  15. International Society for Clinical Densitometry. Facility AccreditationMarch 11, 2018. Available from: http://www.iscd.org/accreditation/.
  16. Lotz JC, Cheal EJ, Hayes WC. Fracture prediction for the proximal femur using finite element models: Part I--Linear analysis. J Biomech Eng. 1991;113(4):353-60.
  17. Cranney A, Guyatt G, Griffith L, Wells G, Tugwell P, Rosen C, et al. Summary of meta-analyses of therapies for postmenopausal osteoporosis. Endocr Rev. 2002;23(4):570-8.
  18. Black DM, Bauer DC, Vittinghoff E, Lui LY, Grauer A, Marin F, et al. Treatment-related changes in bone mineral density as a surrogate biomarker for fracture risk reduction: meta-regression analyses of individual patient data from multiple randomised controlled trials. Lancet Diabetes Endocrinol. 2020;8(8):672-82.
  19. Bouxsein ML, Eastell R, Lui LY, Wu LA, de Papp AE, Grauer A, et al. Change in Bone Density and Reduction in Fracture Risk: A Meta-Regression of Published Trials. J Bone Miner Res. 2019;34(4):632-42.
  20. Hangartner TN. A study of the long-term precision of dual-energy X-ray absorptiometry bone densitometers and implications for the validity of the least-significant-change calculation. Osteoporos Int. 2007;18(4):513-23.
  21. Njeh CF, Fuerst T, Hans D, Blake GM, Genant HK. Radiation exposure in bone mineral density assessment. Applied Radiation & Isotopes. 1999;50(1):215-36.
  22. Bonnick SL. Bone densitometry in clinical practice- application and interpretation. 2nd ed ed. Totowa, N.J: Humana Press; 2004 2004.
  23. Lewiecki EM, Richmond B, Miller PD. Uses and misuses of quantitative ultrasonography in managing osteoporosis. CleveClin J Med. 2006;73(8):742-52.
  24. Fuggle NR, Reginster JY, Al-Daghri N, Bruyere O, Burlet N, Campusano C, et al. Radiofrequency echographic multi spectrometry (REMS) in the diagnosis and management of osteoporosis: state of the art. Aging Clin Exp Res. 2024;36(1):135.
  25. Lewiecki EM. Assessment of Skeletal Strength: Bone Density Testing and Beyond. Endocrinol Metab Clin North Am. 2021;50(2):299-317.
  26. Keaveny TM, Clarke BL, Cosman F, Orwoll ES, Siris ES, Khosla S, et al. Biomechanical Computed Tomography analysis (BCT) for clinical assessment of osteoporosis. Osteoporos Int. 2020;31(6):1025-48.
  27. Qasim M, Lopez Picazo M, Ruiz Wills C, Noailly J, Di Gregorio S, Del Rio Barquero LM, et al. 3D-DXA Based Finite Element Modelling for Femur Strength Prediction: Evaluation Against QCT. J Clin Densitom. 2024;27(2):101471.
  28. Vogt TM, Ross PD, Palermo L, Musliner T, Genant HK, Black D, et al. Vertebral fracture prevalence among women screened for the Fracture Intervention Trial and a simple clinical tool to screen for undiagnosed vertebral fractures. Fracture Intervention Trial Research Group. Mayo Clin Proc. 2000;75(9):888-96.
  29. Gunnes M, Lehmann EH, Mellstrom D, Johnell O. The relationship between anthropometric measurements and fractures in women. Bone. 1996;19(4):407-13.
  30. Siminoski K, Jiang G, Adachi JD, Hanley DA, Cline G, Ioannidis G, et al. Accuracy of height loss during prospective monitoring for detection of incident vertebral fractures. Osteoporos Int. 2005;16(4):403-10.
  31. Margolis KL, Ensrud KE, Schreiner PJ, Tabor HK. Body size and risk for clinical fractures in older women. Study of Osteoporotic Fractures Research Group. AnnIntern Med. 2000;133(2):123-7.
  32. de Laet C, Kanis JA, Oden A, Johanson H, Johnell O, Delmas P, et al. Body mass index as a predictor of fracture risk: a meta-analysis. OsteoporosInt. 2005;16(11):1330-8.
  33. Ensrud KE, Ewing SK, Stone KL, Cauley JA, Bowman PJ, Cummings SR. Intentional and unintentional weight loss increase bone loss and hip fracture risk in older women. J AmGeriatrSoc. 2003;51(12):1740-7.
  34. Lewiecki EM. Evaluation of the osteoporosis patient. In: Dempster DW, Cauley JA, Bouxsein ML, Cosman F, editors. Marcus and Feldman’s Osteoporosis. 2. Fifth ed. San Diego, CA, USA: Elsevier; 2021. p. 1475-500.
  35. Holick MF, Siris ES, Binkley N, Beard MK, Khan A, Katzer JT, et al. Prevalence of Vitamin D inadequacy among postmenopausal North American women receiving osteoporosis therapy. J Clin Endocrinol Metab. 2005;90(6):3215-24.
  36. Tannenbaum C, Clark J, Schwartzman K, Wallenstein S, Lapinski R, Meier D, et al. Yield of laboratory testing to identify secondary contributors to osteoporosis in otherwise healthy women. J Clin Endocrinol Metab. 2003;87:4431-7.
  37. Wagman RB, Marcus R. Beyond bone mineral density-navigating the laboratory assessment of patients with osteoporosis. J Clin Endocrinol Metab. 2002;87(10):4429-30.
  38. Barzel US. Recommended testing in patients with low bone density. J Clin Endocrinol Metab. 2003;88(3):1404-5.
  39. Luckey MM, Tannenbaum C. Authors' response: Recommended testing in patients with low bone density. J Clin Endocrinol Metab. 2003;88(3):1405.
  40. Harper KD, Weber TJ. Secondary osteoporosis - Diagnostic considerations. Endocrinol Metab Clin North Am. 1998;27(2):325-48.
  41. Eastell R. Treatment of postmenopausal osteoporosis. NEnglJMed. 1998;338(11):736-46.
  42. Favus MJ. Postmenopausal osteoporosis and the detection of so-called secondary causes of low bone density. J Clin Endocrinol Metab. 2005;90(6):3800-1.
  43. Looker AC, Orwoll ES, Johnston CCJ, Lindsay RL, Wahner HW, Dunn WL, et al. Prevalence of low femoral bone density in older U.S. adults from NHANES III. JBone MinerRes. 1997;12:1761-8.
  44. Orwoll ES, Klein RF. Osteoporosis in men. EndocrRev. 1995;16:87-116.
  45. Bedatsova L, Drake MT. The skeletal impact of cancer therapies. Br J Clin Pharmacol. 2019;85(6):1161-8.
  46. Rasch S, Lund T, Asmussen JT, Lerberg Nielsen A, Faebo Larsen R, Osterheden Andersen M, et al. Multiple Myeloma Associated Bone Disease. Cancers (Basel). 2020;12(8).
  47. Yu EW, Lee MP, Landon JE, Lindeman KG, Kim SC. Fracture Risk After Bariatric Surgery: Roux-en-Y Gastric Bypass Versus Adjustable Gastric Banding. J Bone Miner Res. 2017;32(6):1229-36.
  48. Adachi Y, Shiota E, Matsumata T, Iso Y, Yoh R, Kitano S. Osteoporosis after gastrectomy: bone mineral density of lumbar spine assessed by dual-energy X-ray absorptiometry. CalcifTissue Int. 2000;66(2):119-22.
  49. Bilezikian JP, Silverberg SJ, Bandeira F, Cetani F, Chandran M, Cusano NE, et al. Task Force #8: Management of Primary Hyperparathyroidism. J Bone Miner Res. 2022;Epub.
  50. Földes J, Tarján G, Szathmari M, Varga F, Krasznai I, Horvath C. Bone mineral density in patients with endogenous subclinical hyperthyroidism:Is this thyroid status a risk factor for osteoporosis. ClinEndocrinol(Oxf). 1993;39:521-7.
  51. Mancini T, Doga M, Mazziotti G, Giustina A. Cushing's Syndrome and Bone. Pituitary. 2004;7:249-52.
  52. Katz S, Weinerman S. Osteoporosis and gastrointestinal disease. Gastroenterol Hepatol (N Y). 2010;6(8):506-17.
  53. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31-41.
  54. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, et al. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. AnnInternMed. 2006;145(4):247-54.
  55. Heaney RP, Recker RR, Ryan RA. Urinary calcium in perimenopausal women: normative values. Osteoporos Int. 1999;9(1):13-8.
  56. Power ML, Heaney RP, Kalkwarf HJ, Pitkin RM, Repke JT, Tsang RC, et al. The role of calcium in health and disease. Am J Obstet Gynecol. 1999;181(6):1560-9.
  57. Pacifici R. Idiopathic hypercalciuria and osteoporosis--distinct clinical manifestations of increased cytokine-induced bone resorption? J Clin Endocrinol Metab. 1997;82(1):29-31.
  58. Fasano A, Berti I, Gerarduzzi T, Not T, Colletti RB, Drago S, et al. Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: a large multicenter study. ArchInternMed. 2003;163(3):286-92.
  59. McLean RR, Jacques PF, Selhub J, Tucker KL, Samelson EJ, Broe KE, et al. Homocysteine as a predictive factor for hip fracture in older persons. NEnglJ Med. 2004;350(20):2042-9.
  60. Lidor C, Frisch B, Gazit D, Gepstein R, Hallel T, Mekori YA. Osteoporosis as the sole presentation of bone marrow mastocytosis. J Bone Miner Res. 1990;5(8):871-6.
  61. Weger M, Deutschmann H, Weger W, Kotanko P, Skrabal F. Incomplete renal tubular acidosis in 'primary' osteoporosis. Osteoporos Int. 1999;10(4):325-9.
  62. Brown JP, Don-Wauchope A, Douville P, Albert C, Vasikaran SD. Current use of bone turnover markers in the management of osteoporosis. Clin Biochem. 2022;109-110:1-10.
  63. Greenspan SL, Parker RA, Ferguson L, Rosen HN, Maitland-Ramsey L, Karpf DB. Early changes in biochemical markers of bone turnover predict the long-term response to alendronate therapy in representative elderly women: a randomized clinical trial. J Bone Miner Res. 1998;13:1431-8.
  64. Eastell R, Barton I, Hannon RA, Chines A, Garnero P, Delmas PD. Relationship of early changes in bone resorption to the reduction in fracture risk with risedronate. J Bone Miner Res. 2003;18:1051-6.
  65. Bouxsein ML, Delmas PD. Considerations for development of surrogate endpoints for antifracture efficacy of new treatments in osteoporosis: a perspective. J Bone Miner Res. 2008;23(8):1155-67.
  66. Vasikaran S, Cooper C, Eastell R, Griesmacher A, Morris HA, Trenti T, et al. International Osteoporosis Foundation and International Federation of Clinical Chemistry and Laboratory Medicine Position on bone marker standards in osteoporosis. Clin Chem Lab Med. 2011;49(8):1271-4.
  67. Lee J, Vasikaran S. Current recommendations for laboratory testing and use of bone turnover markers in management of osteoporosis. Ann Lab Med. 2012;32(2):105-12.
  68. Seibel MJ, Lang M, Geilenkeuser WJ. Interlaboratory variation of biochemical markers of bone turnover. Clin Chem. 2001;47(8):1443-50.
  69. Bergmann P, Body JJ, Boonen S, Boutsen Y, Devogelaer JP, Goemaere S, et al. Evidence-based guidelines for the use of biochemical markers of bone turnover in the selection and monitoring of bisphosphonate treatment in osteoporosis: a consensus document of the Belgian Bone Club. Int J Clin Pract. 2009;63(1):19-26.
  70. Fontalis A, Eastell R. The challenge of long-term adherence: The role of bone turnover markers in monitoring bisphosphonate treatment of osteoporosis. Bone. 2020;136:115336.

 

Management of Diabetes and Hyperglycemia in Hospitalized Patients

ABSTRACT

 

Diabetes is the most prevalent metabolic disorder, and in 2021, the International Diabetes Federation estimated that it affected 537 million adults globally. In 2024, the United States Centers for Disease Control reported that 38.1 million adult Americans, or 14.7% of the adult population, have diabetes. Patients with diabetes have a 3-4-fold greater chance ofhospitalization compared to those without diabetes. In 2020, in the U.S., there were over 7.86 million hospital discharges for adults listed as having diabetes. Hyperglycemia, defined as a blood glucose greater than 140 mg/dl (7.8 mmol/l), isreported in 22-46% of non-critically ill hospitalized patients. Extensive data indicates that inpatient hyperglycemia, in patients with or without a prior diagnosis of diabetes, is associated with an increased risk of complications and mortality. In 2025, the American Diabetes Association (ADA) recommends that once therapy is initiated, a glycemic goal of 140–180 mg/dL (7.8–10.0 mmol/L) is recommended for most critically ill (ICU) individuals with hyperglycemia. More stringent individualized glycemic goals may be appropriate for selected critically ill individuals if they are achieved without significant hypoglycemia. However, for non-critically ill (non-ICU) individuals, a glycemic goal of 100-180 mg/dL (5.6-10.0 mmol/L) is recommended, if achieved without significant hypoglycemia. Insulin remains the best way to control hyperglycemia in the inpatient setting, especially in critically ill patients. Intravenously administered insulin is the preferredmethod to achieve the recommended glycemic target in the ICU. In 2025, the ADA changed its recommendations on using SGLT2 inhibitors in inpatients. They now suggest that in people with type 2 diabetes and heart failure, SGLT2 inhibitors may be started or continued if there are no contraindications (which include prolonged fasting or post-operative recovery). The use of GLP-1 receptor agonists was not recommended in previous guidelines because of the need for more safety and efficacy studies in the inpatient setting. However, increasing evidence indicates that treatment with oral agents such as DPP4 inhibitors, alone or combined with basal insulin, is safe and effective in general medicine andsurgery patients with mild to moderate hyperglycemia.

 

INTRODUCTION

 

Diabetes is the most prevalent metabolic disorder, affecting more than 537 million adults globally and is projected to rise to almost 800 million (10.9% of the adult population) by 2045 (1). In the United States, data from the National Diabetes Statistics Report in 2023 estimated that 38.4 million people of all ages or 14.7% of all U.S. adults had diabetes (2). The percentage of the population with diagnosed diabetes is expected to rise, with one study projecting that as many as onein three U.S. adults will have diabetes during their lifetime (3). People with diabetes have a 35% greater chance of referral for elective operations and a 3-4-fold greater chance of hospitalization compared to those without diabetes (4-7). Data from the US and Scotland estimate that of those individuals with a discharge diagnosis of diabetes, 30% will require two or more hospitalizations in any given year (5; 6; 8). In 2020, in the U.S., there were over 7.86 million hospital discharges for adults listed as having diabetes, (i.e., diabetes as either a principal diagnosis for hospitalization or as a secondary diagnosis, coexisting condition) (9). Data from the USA suggest that the prevalence of diabetes in the adult inpatient population has increased by 2.5% annually from 17.1% to 27.3% between 2000 and 2018 (10). In the UK, the annual National Diabetes Inpatient Audit suggested that the prevalence of diabetes amongst inpatients had risen from15% in 2010 to almost 20% in 2019 (11). In addition, those hospitalized with a diagnosis of diabetes stay in the hospitalfor longer than those without a diagnosis of diabetes admitted for the same condition (12; 13).

 

Diabetes was the 8th leading cause of death in the United States in 2021, accounting for 31.1 deaths per 100,000 of thepopulation (2). A further 120.3 per 100,000 people had diabetes listed as a contributing factor towards the cause of death (2). Not only does diabetes have a significant economic impact on those living with the condition, but it also imposes a substantial burden on the economy, with a total estimated cost of treating people diagnosed with diabetes in the UnitedStates in 2022 of $413 billion – or 25% of all health care spending in the US (14). This included $306.6 billion in directmedical costs. It is estimated that a further cost of $96.5 billion is incurred due to reduced productivity (14). Data from Ireland estimated that the overall cost of treating diabetes represented between 12 and 14% of the annual health budget. The cost per admission for someone with type 1 or type 2 diabetes was €4,027 and €5,026, respectively (15). Globally,diabetes care costs have been estimated at $1.3 trillion, rising to an estimated $2.1-2.5 trillion by 2030 (16; 17). This represents a rise in spending on diabetes as a proportion of global gross domestic product from 1.8% in 2015 to 2.2% in 2030 (17). Other than the costs of diabetes medications, the most significant component of this medical expenditure is hospital inpatient care (13; 18).

 

Hyperglycemia is defined as a blood glucose concentration greater than 140 mg/dl (7.8 mmol/l) (19-21). It is not just found in those with a pre-existing diagnosis of diabetes but in those with stress hyperglycemia or previously undiagnosed diabetes. The prevalence has been reported to be 22% to 46% in non-critically ill hospitalized patients (8; 19). Extensiveobservational and trial data indicate that inpatient hyperglycemia, in patients with or without a prior diagnosis of diabetes,is associated with an increased risk of complications and mortality, a longer hospital stay, a higher admission rate to the intensive care unit (ICU), and a higher need for transitional or nursing home care after hospital discharge (8; 22; 23).

 

Several studies and meta-analyses have shown that attempting ‘tight’ glycemic control using intensive insulin therapy isassociated with an increased risk of hypoglycemia (24-28), which has been associated with increased morbidity andmortality in hospitalized patients (19; 29-34). Thus, while insulin therapy is recommended for managing hyperglycemia inhospitalized patients, the concern about hypoglycemia has led leading professional organizations worldwide to recommend targets that avoid the risk of hypoglycemia (20; 27; 35-38).

 

This chapter reviews the pathophysiology of hyperglycemia during illness, the mechanisms for increased complicationsand mortality due to hyperglycemia and hypoglycemia, and the evidence supporting different therapies and approaches for the management of inpatient diabetes and hyperglycemia in critical care, general medicine, and surgical settings.

 

PREVALENCE OF DIABETES AND HYPERGLYCEMIA IN THE HOSPITALIZED PATIENT

 

Observational studies have reported a prevalence of hyperglycemia and diabetes ranging from 38% to 40% in hospitalized patients (8) and in 70-80% of those with diabetes who have a critical illness or cardiac surgery (39-41). A2017 report using point-of-care bedside glucose tests data in almost 3.5 million people (653,359 ICU and 2,831,436 non-ICU) from 575 hospitals in the United States reported a prevalence of hyperglycemia (defined as a glucose level >180mg/dl [10.0 mmol/l]) of 32.2% in ICU patients and in 32.0% of non-ICU patients (39). A study of 893 people across 69 ICUs in France reported a prevalence of hyperglycemia (>180 mg/dl [10 mmol/l]) of 45% (42). Other USA data suggest that between 2000 and 2018, the prevalence of diabetes amongst adult inpatients increased by 2.5% per year from 17.1% to 27.3% (10), and that over 33% of all hospital discharges in 2020 had diabetes listed as a diagnosis (9). However, this does not include those individuals who develop stress hyperglycemia. The American Diabetes Association (ADA) and American Association of Clinical Endocrinologists (AACE) consensus on inpatient hyperglycemia defined stress hyperglycemia or hospital-related hyperglycemia as any blood glucose concentration >140 mg/dl (>7.8 mmol/l) in patients without a prior history of diabetes (19; 20). The data from the US included those with newly identified diabetes or stress hyperglycemia as well as those with a prior diagnosis of diabetes (39). Although stress hyperglycemia typicallyresolves as the acute illness or surgical stress abates, a significant proportion (up to 60% in some reports) develop confirmed diabetes at 6-12 months after discharge (43; 44). A guide from the UK on the management of ‘diabetes at the front door’, also recommends that any individual without diabetes who presents acutely unwell should have a capillary glucose measurement and blood/urine ketone measurement taken, but that if it is high on admission (i.e. >140mg/dl [7.8 mmol/l]) and subsequently goes down to normal, then a diagnosis of stress hyperglycemia should be made and documented to the primary care team (21).

 

Measurement of HbA1c is indicated in people with hyperglycemia without a history of diabetes to differentiate betweenstress-induced hyperglycemia and previously undiagnosed diabetes (21; 45-48). The ADA also recommends that an HbA1c be done in those with diabetes who have not had it measured in the preceding 3 months (48). The Endocrine Society and the UK Joint British Diabetes Societies for Inpatient Care (JBDS) recommendations indicate that people hospitalized with elevated blood glucose >140 mg/dl (7.8 mmol/l) and an HbA1c of 6.5% (48 mmol/mol) or higher can be identified as having diabetes (19; 21). Given the increasing prevalence of diabetes, the UK has also produced a calculator to help teams work out their optimal staffing levels (49).

 

PATHOPHYSIOLOGY OF HYPERGLYCEMIA DURING ILLNESS

 

In subjects without diabetes during the fasted state, plasma glucose is maintained between 70 – 100 mg/dl (3.9 – 5.6 mmol/l) by a finely regulated balance between glucose production from the liver and kidneys and glucose utilization inperipheral tissues. Maintenance of near-normal glucose concentration is essential for cardiovascular and central nervous system function because the brain can neither synthesize nor store glucose (50; 51).

 

Systemic glucose balance is maintained by dynamic, minute-to-minute regulation of endogenous glucose production and glucose utilization by peripheral tissues (52). Glucose production is accomplished by gluconeogenesis or glycogenolysisprimarily in the liver and, to a lesser degree, by the kidneys (53; 54). Gluconeogenesis results from converting non-carbohydrate precursors such as lactate, alanine, and glycerol to glucose in the liver (55). Excess glucose is polymerized into glycogen, mainly stored in the liver and muscle. Hyperglycemia develops because of three processes: 1) increased gluconeogenesis, 2) accelerated glycogenolysis, and 3) impaired glucose utilization by peripheral tissues (Figure 1).

 

Figure 1. Pathogenesis of hyperglycemia. Hyperglycemia results from increased hepatic glucose production and impaired glucose utilization in peripheral tissues. Reduced insulin and excess counter-regulatory hormones (glucagon, cortisol, catecholamines, and growth hormone) increase lipolysis and protein breakdown (proteolysis) and impair glucose uptake by peripheral tissues. Hyperglycemia causes osmotic diuresis, leading to volume depletion, decreasing glomerular filtration rate, and worsening hyperglycemia. At the cellular level, increased blood glucose concentrations result in mitochondrial injury by generating reactive oxygen species and endothelial dysfunction by inhibiting nitric oxide production. Hyperglycemia increases levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin [IL]-6, leading to immune system dysfunction. These changes can eventually lead to an increased risk of infection, impaired wound healing, multiple organ failure, prolonged hospital stay, and death. Adapted from ref (25).

 

From the quantitative standpoint, inappropriately increased hepatic glucose production represents the major pathogenic disturbance. Increased hepatic glucose production results from the high availability of gluconeogenic precursors. These include the amino acids alanine and glutamine, which result from accelerated proteolysis and decreased proteinsynthesis; lactate, which results from increased muscle glycogenolysis; glycerol, which results from increased lipolysis;and the increased activity of gluconeogenic enzymes (phosphoenol pyruvate carboxykinase, fructose-1,6-bisphosphatase, and pyruvate carboxylase) (53; 55).

 

Glucose metabolism is maintained by an interaction of glucoregulatory hormones – insulin and counter-regulatory hormones (glucagon, cortisol, epinephrine, norepinephrine, and growth hormone). Insulin controls hepatic glucose production by suppressing hepatic gluconeogenesis and glycogenolysis. Depending on the concentration in the circulation, insulin inhibits glycogenolysis and protein breakdown and, at higher concentrations, promotes protein anabolism ininsulin-sensitive tissues such as muscle, glucose uptake, and glycogen synthesis (52; 56; 57). In addition, insulin is a potent inhibitor of lipolysis, free fatty acid oxidation, and ketogenesis (56-58).

 

Counter-regulatory hormones also play an essential role in regulating glucose production and utilization. Glucagon is themost important glycogenolytic hormone, and therefore regulates hepatic glucose production in healthy individuals and in every state of hyperglycemia (53). During stress, excess concentration of counter-regulatory hormones results in altered carbohydrate metabolism by inducing insulin resistance, increasing hepatic glucose production, and reducing peripheralglucose utilization. In addition, high epinephrine levels stimulate glucagon secretion and inhibit insulin release bypancreatic β-cells (59; 60).

 

The development of hyperglycemia results in an inflammatory state characterized by an elevation of pro-inflammatory cytokines and increased oxidative stress markers (61-63). Circulating levels of TNF-α, IL-6, IL1-ß, IL-8, and C-reactiveprotein are significantly increased two- to fourfold on admission in people with severe hyperglycemia compared withcontrol subjects, and levels returned to normal levels after insulin treatment and resolution of hyperglycemic crises (61).Raised concentrations of TNF-α lead to insulin resistance at the level of the insulin receptor and through altered regulationof the insulin-signaling pathway (62; 64). In addition, preventing insulin-mediated activation of phosphatidylinositol 3-kinase TNF-α reduces insulin-stimulated glucose uptake in peripheral tissues (62; 64; 65).

 

CONSEQUENCES OF HYPERGLYCEMIA IN THE HOSPITALIZED PATIENTS

 

A large body of literature, including observational and prospective randomized clinical trials, in people with and withoutdiabetes, as well as those who are critically or non-critically ill has shown a strong association between hyperglycemia (in particular, a blood glucose >200mg/dl [11.0mmol/l]) and poor clinical outcomes, such as mortality, infections, and hospital complications compared to those with a glucose concentration of <100mg/dl (5.6mmol/l) (5; 66-76). This association correlates with the severity of hyperglycemia prior to or on admission and during the hospital stay (72; 77-79). Of interest, increasing evidence indicates an increased risk of complications and mortality in patients without a history of diabetes(stress-induced) compared to patients with a known diagnosis of diabetes (8; 69; 75; 77; 80; 81). It is not clear if stresshyperglycemia is the direct cause of poor outcomes or if it is a general marker of the severity of illness. However, there are data to show that those without a prior history of diabetes have fewer point-of-care glucose concentrations measured compared to those with diabetes, even when glucose concentrations are just as high (75; 82). In those who had diabetes, having more point-of-care tests increases contact with the ward staff, suggesting that impending complications may be picked up sooner, resulting in lower mortality. These data correlate with other work that also shows that those with lower preoperative HbA1c lower the number of post-operative glucose checks in a general surgical population (83).

 

The mechanisms implicated in the detrimental effects of hyperglycemia during acute illnesses are not entirely understood.Current evidence indicates that severe hyperglycemia results in impaired neutrophil granulocyte function, high circulating free fatty acids, and overproduction of pro-inflammatory cytokines and reactive oxygen species (ROS) that can result in direct cellular damage and endothelial and immune dysfunction (84; 85).

 

The majority of evidence linking hyperglycemia and poor outcomes comes from studies in the ICU. Falciglia et al., in a retrospective study of over 250,000 veterans admitted to various ICUs, reported that hyperglycemia is an independentrisk factor for mortality and complications (77). In a nonrandomized, prospective study, Furnary et al. followed 3,554 people with diabetes who underwent coronary artery bypass graft. These were treated with either intermittent subcutaneous insulin (SCI) or with a continuous intravenous insulin infusion (CIII). The group treated with SCI achieved an average blood glucose of 214 mg/dl (11.9 mmol/l), compared to 177 mg/dl (9.8 mmol/l) in the CIII group. The CIII group had significantly fewer deep sternal wound infections and a 50% lower risk-adjusted mortality (73; 86). In other ICU studies, patients with blood glucose levels >200 mg/dl (>11.1 mmol/l) were shown to have higher mortality comparedto those with blood glucose levels <200 mg/dl (<11.1 mmol/) (72; 75). Importantly however, once again it has been shown that it was those people who were not previously known to have diabetes yet who developed hyperglycemia on the ICU who fared worse (75; 87). This was confirmed by another ICU study looking at almost 350,000 people, looking at the outcomes of those with sepsis (88). These authors showed that having hyperglycemia without a prior diagnosis of diabetes was associated with an increased stay in hospital and ICU and greater 90-day mortality (88). However, there was no difference in outcomes for those with diabetes unless they had experienced severe hypoglycemia (<40 mg/dl [2.2 mmol/l]), in which case mortality rose (OR 2.95 95%CI 1.19-7.32) (88). Another ICU study randomized 9230 people who were not given early parenteral nutrition to liberal glucose control (insulin only started if glucose rose to >215 mg/dl [>11.9 mmol/l]), or tight glucose control with glucose concentrations maintained between 80 and 110 mg/dl (4.4 – 6.1 mmol/l). These authors showed no differences in outcome, including length of time in ICU, infection rates, time on respiratory or hemodynamic support, or mortality. The only differences were lower severe acute kidney injury incidence and cholestatic liver dysfunction in the tight glycemic control arm (89). 

 

The association of hyperglycemia and poor outcomes also applies to those not in ICU but admitted to general medicine, surgery, or mental health services. In such individuals, hyperglycemia is associated with poor hospital outcomes, including prolonged hospital stay, infections, disability after hospital discharge, and death (5; 8; 66; 67; 81; 90). In a study of 1,886 patients admitted to a community hospital, mortality in the general floors was significantly higher in patients withnewly diagnosed hyperglycemia and with known diabetes compared to subjects with normal glucose values (10% vs. 1.7% vs. 0.8%, respectively, p < 0.01) (8). In a prospective cohort multicenter study of 2,471 patients with community-acquired pneumonia, those with an admission glucose level of >198 mg/dl (>11.0 mmol/l) had a greater risk of mortality and complications than those with glucose <198 mg/dl (<11.0 mmol/l) (91). The risk of complications increased by 3% foreach 18 mg/dl (1.0 mmol/l) increase in admission glucose (91). In a retrospective study of 348 patients with chronic obstructive pulmonary disease and respiratory tract infection, the relative risk of death was 2.1 in those with a bloodglucose of 126-160 mg/dl (7.0-8.9 mmol/l), and 3.4 for those with a blood glucose of >162 mg/dl (9.0 mmol/l) compared topatients with a blood glucose of 108 mg/dl (6.0 mmol/l) (92). Similar data from a systematic review and meta-analysis from 38 studies of people who needed hospitalization for community-acquired pneumonia showed that in those without a prior diagnosis of diabetes, hyperglycemia was associated with an almost doubling of the need for ICU admission (crude OR 1.82, 95% CI 1.17 to 2.84) and in-hospital mortality (adjusted OR 1.28, 95% CI 1.09 to 1.50) (81). Those people already known to have diabetes had no increased risk of either. 

 

General surgery patients with hyperglycemia during the perioperative period are also at increased risk for adverse outcomes. Reviews of diabetes and the risk of surgical site infection across a variety of surgical specialties have shown that high peri-operative glucose is associated with an increased risk of infection (93; 94). In a case-control study, elevated preoperative glucose levels increased the risk of postoperative mortality in patients undergoing elective non-cardiac non-vascular surgery (95). Patients with glucose levels of 110-200 mg/dl (5.6-11.1 mmol/l) and those with glucose levels of >200 mg/dl (>11.1 mmol/l) had, respectively, 1.7-fold and  2.1-fold increased mortality compared to those with glucose levels <5.6 mmol/l (<110 mg/dl) (95). In another study, patients with glucose levels >220 mg/dl (>12.2 mmol/l) on the first postoperative day had a rate of infection 2.7 times higher than those who had serum glucose levels <220 mg/dl (<12.2 mmol/l) (96). Other authors showed an increase of postoperative infection rate by 30% for every 40mg/dl (2.2 mmol/l) rise in postoperative glucose level above 110 mg/dl (6.1 mmol/l) (96). Further, a study looking at perioperativeglycemic control and the effect on surgical site infections in people with diabetes undergoing foot and ankle surgery showed that 11.9% of those with a serum glucose ≥200 mg/dl (11.1 mmol/l) during the admission developed a surgicalsite infection versus only 5.2% of those with a serum glucose <200 mg/dl (11.1 mmol/l) (odds ratio = 2.45; 95% CI 1.09-5.52, P = 0.03) (97). Lastly, a prospective randomized study looking at the impact of glycemic control at 1-year post livertransplant showed that in those randomized to glycemic control of blood glucose below 140 mg/dl (7.8 mmol/l), any infection within one year occurred in 35 of the 82 patients (42.7%) versus 54 of 82 (65.9%) in those randomized to glycemic control of 180 mg/dl (10.0 mmol/l) (P = 0.0046) (98).

 

Emerging evidence suggests that early intervention and the use of technology allowing proactive identification of people at risk help to reduce hospital-acquired infection rates, episodes of hyper- and hypoglycemia, and, in some cases, length of stay (99-102). A meta-analysis also shows that improving peri-operative glycemic control reduced postoperative infection rates (103).

 

In summary, despite a large amount of work having been done, and the numerous data showing the association – but not causation – between hyperglycemia and poor outcomes, and because there remain a very few robust intervention studies showing a benefit of glycemic control, the optimal blood glucose concentration for people on ICU has yet to be determined (104; 105).

 

GLYCEMIC TARGETS IN THE ICU AND NON-ICU SETTINGS

 

The American Diabetes Association (ADA) and American Association of Clinical Endocrinology (AACE) task force on inpatient glycemic control and other groups recommended differing glycemic targets in the ICU setting (20) (Table 1). These guidelines suggest targeting a BG level between 140 and 180 mg/dl (7.8 and 10.0 mmol/l) for the majority of ICU patients and a lower glucose target between 110 and 140 mg/dl (6.1 and 7.8 mmol/l) in selected ICU patients (i.e., centers with extensive experience and appropriate nursing support, cardiac surgical patients, patients with stableglycemic control without hypoglycemia). Glucose targets >180 mg/dl (>10.0 mmol/l) or <110 mg/dl (<6.1 mmol/l) are not recommended in ICU patients. There is an argument that lowering glucose thresholds for hospital patients will likely be associated with harm (32). Still, an equally persuasive argument suggests that implementing the thresholds advocated by national and organizational guidelines has led to safer care (106).

 

The Society of Critical Care Medicine (SCCM) guidelines for the management of hyperglycemia in critically ill (ICU)patients recently “recommended against” titrating an insulin infusion to a lower glucose target of 80–139 mg/dL (4.4–7.7 mmol/L) as compared with a higher BG target range of 140–200 mg/dL (7.8–11.1 mmol/L) to reduce the risk of hypoglycemia (107). They also recommended that clinicians should initiate glycemic management protocols and procedures to treat persistent hyperglycemia greater than or equal to 180 mg/dL (10 mmol/L) to maintain target glucose below <180 mg/dl (<10.0 mmol/l) in critically ill adults (107). They also suggest that the insulin regimen and monitoringsystem be designed to avoid and detect hypoglycemia (blood glucose <70 mg/dl [<3.9 mmol/l]) and to minimize glycemicvariability.

 

Table 1. Major Guidelines for Treatment of Hyperglycemia in a Hospital Setting

 

ICU

Non-ICU

ADA/AACE (20; 108)

Initiate insulin therapy for persistent hyperglycemia (glucose >180 mg/dl [>10 mmol/l]).Treatment goal: For most people, target a glucose level between 140 – 180 mg/dl (7.8 – 10.0 mmol/l].More stringent goals (110 – 140 mg/dl [6.1 – 7.8 mmol/l]) or 100 – 180 mg/dL (5.6 –10.0 mmol/L), may be appropriate for selected patients and are acceptable if they can be achieved without significant hypoglycemia No specific guidelines.Insulin therapy should be initiated for the treatment of persistent hyperglycemia ≥180 mg/dL (10.0 mmol/L) and targeted to a glucose range of 140 –180 mg/dL (7.8 – 10.0 mmol/L) for most critically ill patients.
More stringent goals, such as 110–140 mg/dL (6.1–7.8 mmol/L), may be appropriate for selected patients (e.g., critically ill postsurgical patients or patients with cardiac surgery) as long as they can be achieved without significant hypoglycemia.Less stringent targets (e.g., >250 mg/dL (13.9 mmol/L) maybe appropriate in people withsevere comorbidities or end of life care.

ACP (27)

Recommends against intensiveinsulin therapy in those with orwithout diabetes in surgical / medical ICUs

Treatment goal: target glucosebetween 140 – 200 mg/dl (7.8 – 11.0 mmol/l), in people with or without diabetes, in surgical / medical ICUs

 

Critical Care Society (107)

BG >180 mg/dl (>10.0 mmol/l) should trigger insulin therapy.

Treatment goal: maintainglucose <180 mg/dl (<10.0 mmol/l) for most adults in ICU.

Maintain glucose levels <180mg/dl (10.0 mmol/l) while avoiding hypoglycemia.

 

Endocrine Society (19; 109)

 

Pre-meal glucose target <140mg/dl (<7.8mmol/l) and random blood glucose <180 mg/dl (<10.0 mmol/l). Those with insulin treated diabetes aim for a target glucose of 100 – 180 mg/dL (5.6 – 10 mmol/L). A lower target range may beappropriate in people able toachieve and maintain glycemiccontrol without hypoglycemia. Aglucose of <180 – 200 mg/dl(<10.0 – 11.0 mmol/l) isappropriate in those withterminal illness and/or withlimited life expectancy or at high risk for hypoglycemia.

Adjust antidiabetic therapywhen glucose falls <100 mg/dl (<5.6 mmol/l) to avoidhypoglycemia.

Society of ThoracicSurgeons (110)

Continuous insulin infusionpreferred over SC or intermittent intravenousboluses.

Treatment goal: Recommendglucose <180 mg/dl (<10.0 mmol/l) during surgery (≤110 mg/dl [≤6.1 mmol/l] in fasting and pre-meal states)

 

Joint British Diabetes Society for Inpatient Care(111)

 

Target blood glucose levels inmost people of between 108 – 180 mg/dl (6.0 – 10 mmol/l) with an acceptable range of between 72 – 216 mg/dl (4.0 – 12.0 mmol/l).

AACE/ADA, American Association of Endocrinologists and American Diabetes Association joint guidelines; ACP, American College of Physicians; ADA, American Diabetes Association; ICU, intensive care unit; SC, subcutaneous.

 

In the non-ICU setting, the Endocrine Society and the ADA/AACE Practice Guidelines recommended a pre-meal glucose of <140 mg/dl (<7.8 mmol/l) and a random BG of <180 mg/dl (<10.0 mmol/l) for the majority of non-critically ill patients treated with insulin (19; 20; 35; 109). More recently, the American Diabetes Association has recommended that targetglucose for most general medicine and surgery patients in non-ICU settings should be between 140 – 180 mg/dl (7.8 – 10.0 mmol/l) (108). In contrast, higher glucose ranges (>200 mg/dl [>11.1 mmol/l]) may be acceptable in people who are terminally ill or in those with severe comorbidities as a way of avoiding symptomatic hyperglycemia (19; 112).

 

Guidelines from the JBDS in the UK published over the last few years aim for target blood glucose concentrations in most people between 108 – 180 mg/dl (6.0 – 10.0 mmol/l) with an acceptable range of between 72 – 216 mg/dl (4.0 –12.0 mmol/l) (111). Table 1 summarizes the currently available guidelines for managing hyperglycemia in the hospitalsetting.

 

EVIDENCE FOR CONTROLLING HYPERGLYCEMIA IN ICU AND NON – ICU SETTINGS

 

In 2001, the Leuven surgical ICU study promoted intensive glycemic control in the critical care setting (113). This study randomized 1,548 people admitted to the surgical ICU (63% cardiac cases, 13% with diabetes, with most receiving earlyparenteral nutrition). Individuals were randomized to either conventional therapy, with target glucose between 180 – 200 mg/dl (10.0 – 11.1 mmol/l), or intensive treatment to target glucose between 80 – 110 mg/dl (4.4 – 6.1 mmol/l). Those in the conventional arm had a mean daily glucose average of 153 mg/dl (8.5 mmol/l), and those in the intensive arm had an average glucose of 103 mg/dl (5.7 mmol/l). Those in the intensive group had significantly less bacteremia, fewer antibioticrequirements, lower length of ventilator dependency, lower number of ICU days, and an overall 34% reduction in mortality(113). Following a similar study design, the same group of investigators randomized people to a medical ICU (18% withdiabetes) and reported that intensive insulin therapy (mean daily glucose of 111 mg/dl [6.2 mmol/l]) resulted in less ICU and total hospital complications in those with three days of insulin treatment (114). These two studies together, based on the positive outcomes on morbidity and mortality, suggested a glycemic target in the ICU of 140 – 180 mg/dl (7.8 – 10.0mmol/l) (20). There was also a realization that while lower targets may be appropriate for selected individuals, a target of <110 mg/dl (<6.1 mmol/l) was not recommended (20).

 

Many well-designed randomized controlled trials and meta-analyses have shown that such low glucose targets aredifficult to achieve, even in environments with high staff-to-patient ratios, without increasing the risk for severe hypoglycemia (24; 115-117). In addition, these and other studies failed to show improvement in clinical outcomes andhave even shown increased mortality risk with intensive glycemic control, targeting glucose concentrations of (80 – 110 mg/dl [4.4 – 6.1 mmol/l]) versus conventional glycemic control (140 – 200 mg/dl [7.8 – 11.0 mmol/l]) (Table 2) (29; 115-118). Most of these studies showed no differences in clinical outcomes between groups but had an increased risk of severe hypoglycemia in the intensively treated arms.  One study in ICU patients was the Normoglycemia in Intensive Care Evaluation and Surviving Using Glucose Algorithm Regulation (NICE-SUGAR) trial, which randomized over 6104subjects to receive either conventional glycemic control to target glucose <180 mg/dl [<10.0 mmol/l]) or intensiveglycemic control (target 81 – 108 mg/dl [4.5 – 6.0 mmol/l]). This study also reported no difference in hospital mortality butfound increased mortality at 90 days of follow-up (24.9% vs. 27.5%, p=0.02) (24). In a subsequent analysis of the trial, the NICE-SUGAR investigators reported a higher frequency of hypoglycemia in the intensive arm (6.8% vs. 0.5%), and those with hypoglycemia had a ~2-fold increase in mortality compared to patients without hypoglycemia (29). More recently, Gunst et al. recently published the results of a multicenter, randomized trial involving 9230 patients in medical and surgical ICUs (89). 4622 patients were assigned to liberal glucose control, where insulin was initiated only when the blood glucose level was above 215 mg per deciliter (11.9 mmol per liter), and 4608 patients were assigned to tight glucose control, targeting a glucose level between 80 and 110 mg per deciliter. The primary outcome, the duration of time in ICU care, did not differ significantly between the two trial groups. The hazard ratio for earlier discharge alive with tight glucose control was 1.00 with a 95% confidence interval of 0.96 to 1.04. Effective glycemic separation between the groups was observed, with a median absolute difference of -28 mg/dl (-1.6 mmol/l) in daily blood glucose levels. Additionally, the safety outcome, mortality within 90 days after randomization, was 10.1% in the liberal-control group and 10.5% in the tight-control group. The incidence of other secondary outcomes, including severe hypoglycemia, time to discharge alive from the hospital, use of respiratory support, or in-hospital mortality, were no different between intensive and relaxed glycemic targets, except for a trend in lower rates of liver and kidney injury in the tight control group.  Umpierrez recently summarized the data on mortality and outcomes of ICU RCTs (119).

 

Increasing evidence indicates that high pre-admission glycemic control – as measured by HbA1c >8.0% (64mmol/mol) is associated with lower mortality than those with an HbA1c <6.5% (48mmol/mol) (120). Whether this is due to an increased risk of hypoglycemia in the low HbA1c group or an increased frequency of monitoring or bedside vigilance in those with higher glucose or preadmission HbA1c remains unknown (75; 83).

 

Table 2. Clinical Trials of Intensive Glycemic Control in ICU Populations

Study

Setting

Population

Percentage with diabetes

Clinical Outcome

Malmberg, 1994 (121)

CCU

People with diabetes with suspected or confirmed acute MI

100

28% decreasemortality after 1 year

Furnary, 1999 (73)

CCU

People with diabetes undergoing CABG

100

65% decrease indeep sternal woundinfection rate

Van den Berghe,2001 (113)

Surgical ICU

Mixed, with CABG

13

34% decrease in mortality

Furnary, 2003 (86)

CCU

People with diabetes undergoing CABG

100

50% decrease inadjusted mortalityrate

Krinsley, 2003 (72)

Medical and surgical ICU

Mixed

22.4

27% decrease inmortality

Lazar, 2004 (122)

Operating room and ICU

People with diabetes undergoing CABG

100

 

60% decrease of post - operative atrialfibrillation

Van den Berghe,2006 (114)

Medical ICU

Mixed

17

18% decreasemortality

Gandhi, 2007 (123)

OperatingRoom

Mixed, undergoing cardiac surgery

19.6

No difference inmortality; increase instroke rate in the intensive treatmentarm

VISEP, 2008 (115)

Medical ICU

Mixed, admitted withsepsis

30

No differences in 28-day or 90-daymortality, end-organ failure, length of stay

De La Rosa, 2008(116)

Medical and surgical ICU

Mixed

12

No differences in 28-day mortality orinfection rate

Glucontrol, 2009 (124)

Medical and surgical ICU

Mixed

18

No difference in 28-day mortality

NICE-SUGAR,2009/2012 (24; 29)

Medical and surgical ICU

Mixed

20

No difference in 90-day mortality

Boston Children’s(SPECS), 2012 (125; 126)

Cardiac ICU

Cardiac surgery,people without diabetes

0

No differences in 30-day mortality, length of stay, in the cardiac ICU, length ofhospital, duration ofmechanicalventilation andvasoactive support,or measures of organfailure

ChiP, 2014 (127)

Pediatric ICU

Criticalillness/injury/majorsurgery, those without diabetes.

0

No difference in 30-day mortality.Increasedhypoglycemia in theintensive treatedgroup

CGAO–REA, 2014 (128; 129)

Medical ICU

Mixed

23

No difference in 90-day mortality. Increasedhypoglycemia in theintensive treatedgroup

Okabayashi, 2014 (130)

 

Surgical ICU

Mixed

25.3

Decreased surgicalsite infection in theintensive treatedgroup

Umpierrez (GLUCOCABG) 2015

Surgical ICU

CABG

50%

No difference in mortality

 

Gunst et al

ICU

ICU

 

XX

No difference in mortality

 

MI, myocardial infarction, ICU – Intensive Care Unit, CABG – Coronary artery bypass graft. Mixed-study enrolled those with and without diabetes.

 

The GLUCO-CABG trial was a randomized open-label clinical study that included those with and without diabetesundergoing CABG who experienced perioperative hyperglycemia, defined as a BG >140 mg/dl (>7.8 mmol/l) 6069 (70). A total of 302 people between 18 and 80 years of age were randomized to the intensive glycemic control group (target BG 100 – 140 mg/dl [5.6 – 7.8 mmol/l]) or the control group (BG 141 – 180 mg/dl [7.9 – 10.0 mmol/l]) in the ICU. Aftertransitioning from the ICU to the telemetry floor, patients were managed with a single treatment protocol to maintain a glucose target of <140 mg/dl (<7.8 mmol/l) before meals during the hospital stay. The primary outcome included differences between intensive and conservative glucose control on a composite of perioperative complications, includingsternal wound infection, bacteremia, respiratory failure, pneumonia, acute kidney injury, major adverse cardiovascular events including acute coronary syndrome, stroke, heart failure, and cardiac arrhythmias (70). The mean BG during the ICU stay was 132±14 mg/dl (7.3±0.8 mmol/l) in the intensive and 152±17 mg/dl (8.4±1.0 mmol/l) in the conservative group. Intensive glucose treatment resulted in a 20% reduction in perioperative complications compared to theconservative group (42% vs. 52%). Of interest, there were no differences in the rate of complications among patients with diabetes treated with intensive or conservative regimens (42% vs. 52%, p=0.08); however, intensive treatment wasassociated with a significantly lower rate of complications compared to the conservative group in those without diabetes (34% vs. 55%, p=0.008) (70). Hospitalization costs were lower in the intensive group (median [IQR] $36,681 [28,488 – 46,074] vs. $40,913 [31,464 – 56,629], p=0.04), with an average total cost savings of $3,654 per case compared to conservative glucose control (131).

 

To date, few large studies have been conducted to determine if improved control in those not in ICU may result inreduced morbidity and mortality in general medical and surgical patients – indeed, until recently, for most people in hospital with diabetes while there are observational data to show that dysglycemia is harmful, there were little data to show that improving glycemic control helps (132). A randomized controlled trial from 2011 reported that improved glucose control using a basal-bolus regimen may reduce hospital complications in general surgery patients (71). Improving glucose control with a basal-bolus regimen significantly reduced the frequency of composite complications, including postoperative wound infection, pneumonia, bacteremia, and acute renal and respiratory failure (71). In that study, treatment with basal-bolus insulin reduced average total inpatient costs per day by 14% or $751 compared totreatment with a correction bolus dose insulin alone (133). Another study from Australia has shown that in a randomized study of 1371 surgical inpatients, 680 with even a single glucose value >200 mg/dl (11.1 mmol/l) received early intervention from the diabetes inpatient team (134). This led to reductions in glucose of a modest -5.4 mg/dl (-0.3 mmol/l), which still equated to a 4.6%, statistically significant reduction in hospital-acquired infections compared to those receiving standard care (134).

 

HYPOGLYCEMIA

 

Hypoglycemia is the most common side effect of treatment of all types of diabetes and stress hyperglycemia in thehospital setting. It presents a significant barrier to satisfactory long-term glycemic control. Hypoglycemia results from animbalance between glucose supply, glucose utilization, and current insulin levels. Hypoglycemia is defined as a lower-than-normal level of blood glucose. Hypoglycemia is defined as any glucose level <70 mg/dl (<3.9 mmol/l) (108; 135) for hospital inpatients. Level 1 hypoglycemia is a glucose concentration of 54 – 70 mg/dL (3.0 – 3.9 mmol/L). Level 2 hypoglycemia is a blood glucose concentration of <54 mg/dL (3.0 mmol/L) (108). Severe hypoglycemia has been defined by many as <40 mg/dl (<2.2 mmol/l) (136), but a newer definition, Level 3 hypoglycemia, is a glucose concentration low enough where the individual requires third-party assistance to aid recovery (108). The UK JBDS guideline suggests that the lower limit of glucose in the inpatient population should be 108 mg/dl (6.0 mmol/l), and that the range between 72 – 108 mg/dl (4.0 – 6.0 mmol/l) be designated ‘looming’ hypoglycemia, to alert ward staff to take action because of the possibility that lower glucose levels may be associated with harm (137). The exception to this is those people on a diet only or those people on an insulin pump / closed loop system who can self-manage their diabetes while in the hospital.

 

The incidence of severe hypoglycemia in the different trials ranged between 5% and 28%, depending on the intensity ofglycemic control in the ICU (138). Rates from subcutaneous insulin trials in non-critically ill patients range from less than 1% to 33% (71; 139; 140). In 2017, the UK National Diabetes Inpatient Audit (NaDIA) data showed that 18% of peoplewith diabetes in hospital experienced one or more hypoglycemic episodes with a blood glucose <72mg/dl (<4.0 mmol/l) –down from 26% in 2011, with 7% (1 in 14) of all inpatients experiencing episodes requiring third party assistance to administer rescue therapy (141). The NaDIA data also showed that those with type 1 diabetes had the highestprevalence, with 25% experiencing a severe hypoglycemic episode (141). Furthermore, 1.3% (1 in 80) of those in hospitals with diabetes required some form of injectable rescue treatment (i.e., IV glucose or IM glucagon), down from2.1% in 2011 (141). The same data showed that the highest proportion of episodes occurred overnight (28%) between 05:00 and 09.00 AM when snack availability was likely the lowest (140; 141).

 

Table 3 lists some key factors that predict the likelihood of someone experiencing a hypoglycemic event whilehospitalized. These also include older age, greater illness severity, diabetes, and the use of oral glucose-loweringmedications and/or insulin (137; 142-145). In-hospital processes of care that contribute to the risk for hypoglycemia include unexpected changes in nutritional intake that are not accompanied by associated changes in the glycemic management regimen. Examples include (but are not limited to) cessation of nutrition for procedures, an adjustment in the amount of nutritional support, interruption of the established routine for glucose monitoring, deviations from the established glucose control protocols, and failure to adjust therapy when glucose is trending down, or steroid therapy isbeing tapered (137; 146; 147). A common cause of inpatient hypoglycemia is when handwritten insulin prescriptions lead to errors, including misreading, e.g., when ‘U’ is used for units (i.e., 4U becoming 40 units) or confusing the insulin name with the dose (e.g., Humalog Mix25 becoming Humalog 25 units) (148). Electronic prescribing has been associated with a lower rate of prescription errors (141).

 

However, other factors may also be involved, such as concurrent use of drugs with hypoglycemic agents, e.g., warfarin, quinine, salicylates, fibrates, sulphonamides (including co-trimoxazole), monoamine oxidase inhibitors, NSAIDs, probenecid, somatostatin analogs, or selective serotonin reuptake inhibitors. Secondary causes of inpatienthypoglycemia include loss of counter-regulatory hormone function, e.g., Addison’s disease, growth hormone deficiency,hypothyroidism, or hypopituitarism.

 

Table 3. Common Risk Factors for Developing Hypoglycemia in the Hospital

Prior episode of hypoglycemia

Older age

Chronic kidney disease

Congestive heart failure

Liver Failure

Sepsis

Malnutrition

Erratic eating patterns / Nutritional interruptions / Lack of access to carbohydrates

Malignancies

Insulin regimen

Type 1 diabetes

Mental status changes

Certain concomitants use of medications

Duration of diabetes

 

The development of hypoglycemia is associated with adverse hospital outcomes (29; 30; 117; 118; 124; 144; 149-155). Turchin et al. examined data from 4,368 admission episodes for people with diabetes, of which one-third were on regular insulin therapy (30). Patients experiencing inpatient hypoglycemia experienced a 66% increased risk of death within one year and spent 2.8 days longer in the hospital compared to those not experiencing hypoglycemia. A 2019 systematic review and meta-analysis of hospital-acquired hypoglycemia in non-ICU patients suggested that adults exposed to glucose levels <72 mg/dl (<4.0 mmol/l) experienced a mean increased length of hospital stay of 4.1 days (95% CI 2.36 – 5.79) compared to those who did not experience hypoglycemia (144). The same dataset suggested an increased relative risk of in-hospital mortality for non-ICU patients of 2.09 (95% CI 1.64 – 2.67) (144). There was a non-significant reduction in mortality for those in ICU of 0.75 (95% CI 0.49 – 1.16) (144). The odds ratio (95% confidence interval) for mortality associated with one or more episodes was 2.28 (1.41-3.70, p=0.0008) among a cohort of 5,365 patients admitted to amixed medical-surgical ICU (142). In a larger cohort of over 6,000 patients, hypoglycemia was associated with longer ICU stays and greater hospital mortality, especially for patients with more than one episode of hypoglycemia (29). These data strengthen the argument to have potentially less strict glycemic targets for those not on ICU (32; 137). For example, if an individual has a glucose of 75 mg/dl (4.2 mmol/l), and is on an intravenous insulin infusion, by the time their bedside capillary glucose is next measured, they may have a glucose well below 72 mg/dl (4.0 mmol/l), thus they have come to potential harm. Indeed, data published from previous NaDIA surveys and NaDIA Harms using data from over 100 hospitals across the UK showed several serious adverse events, including seizures, permanent cerebral damage, cardiac arrests, and deaths. Insulin therapy was implicated in several of these events (33; 34; 156; 157). The counterargument is that there are initiatives to reduce the risk of developing inpatient hypoglycemia and having national guidance has led to improved patient care overall (106; 158). As with the outpatient population, the increased use of technology may help avoid hypoglycemia (159).

 

Hypoglycemia has been associated with adverse cardiovascular outcomes, such as increased myocardial contractility, prolonged QT interval (possibly due to the rapid drop in potassium concentrations due to the increased circulating epinephrine and norepinephrine), ischemic electrocardiogram changes and repolarization abnormalities, angina, arrhythmias, increased inflammation, and sudden death, (51; 160-162). The mechanisms for the poor outcome have yet to be entirely understood. Still, hypoglycemia has been associated with increases in pro-inflammatory cytokines (TNFα,IL-1β, IL-6, and IL-8), markers of lipid peroxidation, acute changes in endothelial dysfunction with associated vasoconstriction, increased blood coagulability, cellular adhesion, and oxidative stress (163; 164).

 

Despite these observations, the direct causal effect of iatrogenic hypoglycemia on outcome is still debatable. Kosiborod et al. reported that spontaneous hypoglycemia, but not insulin–induced hypoglycemia, was associated with higherhospital mortality (152). Similarly, another study among 31,970 patients also reported that hypoglycemia is associatedwith increased in-hospital mortality. Still, the risk was limited to patients with spontaneous hypoglycemia and not topatients with drug-associated hypoglycemia (165). These studies raise the possibility that hypoglycemia, like hyperglycemia, despite the biochemical and other changes described, is a marker of disease burden rather than a directcause of death.

 

RECOMMENDATIONS FOR MANAGING HYPERGLYCEMIA IN THE  HOSPITAL ENVIRONMENT

 

Knowledge of Diabetes Management Amongst Medical Staff

 

The burden on inpatient diabetes falls most frequently on junior medical staff, who often have little or no specialist diabetes training. As such, it is perhaps unsurprising that errors occur. In the UK, a survey of junior doctors showed that unlike other commonly encountered medical conditions, such as acute asthma or angina, their knowledge about and confidence in managing diabetes was significantly lower (166). In 2019, this was also shown in a multicenter study from the US – with the major difference being that whilst most staff felt confident and comfortable managing diabetes, when challenged on how to manage certain situations, and in particular identifying glucose targets for those who were critically ill or the threshold for defining hypoglycemia, their confidence was far higher than their knowledge – a potentially devastating combination (167). Given the high prevalence of diabetes amongst hospital inpatients, essential diabetes management should be part of mandatory training. However, studies have found that despite the implementation of training programs, structured staff education has not shown to be of significant benefit in terms of improved patient outcomes (168; 169)

 

Management of Inpatient Hyperglycemia in the ICU

 

Insulin is the best way to control hyperglycemia in the inpatient setting, especially in critically ill patients. A variable-rate intravenous insulin infusion is the preferred method to achieve the recommended glycemic target (ADA Standards of Care 2025). The short half-life of intravenous insulin makes it ideal in this setting because it allows flexibility in the eventof unpredicted changes in an individual’s health, medications, and nutrition.

 

When someone is identified as having hyperglycemia (blood glucose ≥180 mg/dl [≥10.0 mmol/l]), a variable rate intravenous insulin infusion should be started to maintain blood glucose levels <180 mg/dl (<10.0 mmol/l). A variety ofintravenous infusion protocols are effective in achieving glycemic control with a low rate of hypoglycemic events and in improving hospital outcomes (73; 86; 113; 121; 170-174). A proper protocol should allow flexible blood glucose targets to be modified based on the individual’s clinical situation. Further, it should have clear instructions about the blood glucosethreshold for initiating an insulin infusion and the initial rate. The appropriate fluids should also be prescribed. It should bevalidated to avoid hyperglycemia if adjusted too slowly and hypoglycemia if adjusted too fast. Accurate insulin administration requires a reliable infusion pump that can deliver the insulin dose in increments of 0.1 units per hour (138; 172).

 

There is no ideal insulin protocol for managing hyperglycemia in the critically ill patient. In addition, no clear evidence demonstrates the benefit of one protocol/algorithm versus any other (138). Implementing any of these algorithms requires close follow-up by the nursing staff and is prone to human errors. Some institutions have developedcomputerized protocols that can be implemented to avoid errors in dosing (138; 175-179). Essential elements thatincrease protocol success of continuous insulin infusion are: 1) rate adjustment considers the current and previous glucose value and the current rate of insulin infusion, 2) rate adjustment considers the rate of change (or lack of change) from the previous reading, and 3) frequent glucose monitoring (hourly until stable glycemia is established, and then at least every 2 – 3 hours) (138; 171; 180-182).

 

Several computer-based algorithms aiming to direct the nursing staff in adjusting the insulin infusion rate have become commercially available (175-177; 179; 183). Retrospective cohorts and controlled trials have reported a more rapid and tighter glycemic control with computer-guided algorithms than standard paper form protocols in ICU patients (176; 184), as well as lower glycemic variability than patients treated with the standard insulin infusion regimens. Despite differences in glycemic control between insulin algorithms, another study showed no difference between computerized protocolsversus conventional glucose control (128). Thus, most insulin algorithms appear to be appropriate alternatives for managing hyperglycemia in critically ill patients, and the choice depends upon the physician’s preferences, staffing availability, and cost considerations. As mentioned, the increasing implementation of available technology, in particular the use of closed loops should improve the management of dysglycemia over the coming years (185-187).

 

Managing Hyperglycemia in the Non-ICU Setting

 

Subcutaneous insulin is the preferred therapeutic agent for glucose control in those admitted to non-ICU settings under general medicine and surgery. A recent study suggested that the use of bolus correction doses of subcutaneous insulin (“subcutaneous sliding scale insulin” (SSI)) is an acceptable way of controlling dysglycemia, particularly in those whose admission glucose levels were <180 mg/dl (10 mmol/l) (188; 189). However, many studies do not agree and advocate against using this method as the only way to control glucose levels because it results in undesirable hypoglycemia andhyperglycemia or inadequately controls dysglycemia (109; 190-193). It has become evident in recent years that the useof scheduled subcutaneous insulin therapy with basal (e.g. glargine, detemir or degludec) once daily or with intermediate-acting insulin (NPH) given twice daily alone or in combination with short (regular) or rapid-acting insulin (lispro, aspart, glulisine) prior to meals is effective and safe for the management of most patients with hyperglycemia and diabetes (20; 108; 194).

 

The basal-bolus (prandial) insulin regimen is considered the physiologic approach as it addresses the three componentsof insulin requirement: basal (what is required in the fasting state), nutritional (what is needed for peripheral glucosedisposal following a meal), and supplemental (what is necessary for unexpected glucose elevations, or to dispose ofglucose in hyperglycemia (195).

 

A prospective, randomized multi-center trial compared the efficacy and safety of a basal/bolus insulin regimen with basal-bolus regimen and SSI in people with type 2 diabetes admitted to a general medicine service (139). The use of a basal-bolus insulin regimen improved blood glucose control more than the subcutaneous sliding scale alone. A blood glucose target <140 mg/dl (<7.8 mmol/l) was achieved in 66% of those in the glargine plus glulisine group and 38% in the slidingscale group (139). The incidence of hypoglycemia, defined as a BG <60 mg/dl (<3.3 mmol/l), was less than 5% in thosetreated with basal-bolus or SSI. A different study on general surgery inpatients also compared the efficacy and safety of a basal-bolus regimen to SSI in those with type 2 diabetes (71). The basal-bolus regimen resulted in a significant improvement in glucose control and a reduction in the frequency of the composite of postoperative complications, including wound infection, pneumonia, respiratory failure, acute renal failure, and  bacteremia.

 

Multi-dose human NPH and regular insulin have been compared to basal-bolus treatment with insulin analogs in an open-label, controlled, multicenter trial in 130 medical admissions with type 2 diabetes (196). This study found that both treatment regimens significantly improved inpatient glycemic control with a glucose target of <140 mg/dl (<7.8 mmol/l) before meals and no difference in the rate of hypoglycemic events. Thus, a similar improvement in glycemic control can be achieved with either basal-bolus therapy with insulin analogs or with NPH/regular human insulin in people with type 2diabetes.

 

Most people in the hospital have reduced caloric intake due to a lack of appetite, medical procedures, or surgical intervention. In the Basal Plus trial, people with type 2 diabetes who were treated with diet, oral antidiabetic agents, orlow-dose insulin (≤ 0.4 unit/kg/day) prior to admission were randomized to receive a standard basal-bolus regimen withglargine once daily and glulisine before meals or a single daily dose of glargine. In addition, supplemental doses ofglulisine were administered for correction of hyperglycemia (>140 mg/dl [>7.8 mmol/l]) per sliding scale (197). This study reported that the basal approach resulted in similar improvement in glycemic control and the frequency of hypoglycemia compared to a standard basal-bolus regimen (197). Thus, in insulin-naive individuals or those receiving low-dose insulin on admission, as well as those with reduced oral intake, the use of a basal plus regimen is an effective alternative to basal-bolus (108).

 

The recommended total daily insulin dose should start between 0.3 to 0.5 units per Kg (139; 147; 198; 199) for mostpeople with diabetes. Starting doses greater than 0.6 – 0.8 units/kg/day have been associated with 3-fold higher odds of hypoglycemia than doses lower than 0.2 U/kg/day. In elderly individuals or those with impaired renal function, lower initial daily doses (≤ 0.3 units/kg) may lower the risk of hypoglycemia (200).

 

Hospital Use of Non-Insulin Therapy in Non-Critical Care Settings

 

Several other classes of non-insulin glucose-lowering agents have been tried in the hospital setting. However, most are not suitable for use. Metformin, while the first line for type 2 diabetes in the outpatient setting, may not be appropriate where there is any evidence of dehydration, renal impairment, or if intravenous contrast is due to be administered due to the risk of lactic acidosis or worsening of renal function (195). Despite the lack of robust evidence of benefit, it remains in everyday use in many countries (201). Thiazolidinediones are excellent at lowering glucose but are used rarely, and possibly inappropriately, in hospitalized patients because they take several weeks to reach their maximum effect, may precipitate heart failure, and may cause peripheral edema due to fluid retention (202-204).

 

Sulfonylureas work rapidly and are often the drugs of choice for worsening diabetes in an outpatient setting (205). They remain in everyday use in many countries, with up to 20% of inpatients with diabetes in the USA and UK remaining on them (140; 203). However, they increase the risk of hypoglycemia. There is data to show that they remain one of the most frequent causes of inpatient hypoglycemia, thus extending the length of hospital stay and increasing the risk of inpatient mortality (141; 206-208).

 

Oral glucose-lowering medication use is limited by the delay and unpredictability of onset of action, and there is also concern regarding the cardiovascular effects of sulfonylureas and the contraindication of metformin use in patients with renal or liver dysfunction (19; 209). Recent work using the sodium-glucose co-transporter 2 inhibitors for corticosteroid-induced hyperglycemia in acute exacerbation of chronic obstructive pulmonary disease (COPD) or used in COVID infections failed to demonstrate an improvement in outcomes (210; 211). Indeed, despite their clear benefits in the outpatient population with and without diabetes, robust evidence for the benefit of SGLT2i use in the inpatient population (in people with diabetes) is lacking (212; 213).

 

The use of oral antidiabetic agents was not recommended in previous guidelines because of the need for more safety and efficacy studies in the inpatient setting (20). However, increasing evidence indicates that treatment with dipeptidyl peptidase-4 (DPP4) inhibitors, alone or in combination with basal insulin, is safe and effective in general medicine and surgery with mild to moderate hyperglycemia (48). In a pilot study, general medicine and surgical inpatients with bloodglucose between 140 and 400 mg/dl (7.8 – 22.2 mmol/l) treated with diet, oral antidiabetic drugs, or low-dose insulin (≤0.4 U/kg/day) were randomized to sitagliptin once daily, sitagliptin and basal insulin, or basal-bolus insulin (214). All groups received correction doses of lispro before meals and bedtime for blood glucose >140 mg/dl (>7.8 mmol/l). In those with mild-moderate hyperglycemia (<180 mg/dl [<10 mmol/l]), the use of sitagliptin plus supplemental (correctiondoses) or in combination with basal insulin resulted in no significant differences in mean daily blood glucose, frequency of hypoglycemia or the number of treatment failures compared to the basal-bolus regimen (214). The SITA-HOSPITAL trial, a multicenter, randomized controlled study in 279 general medicine and surgery individuals with type 2 diabetes previously treated with oral anti-diabetic agents or low-dose insulin (<0.6 U/kg/d), also reported similar glycemic control,hypoglycemia rate, hospital length-of-stay, treatment failures or hospital complications (including acute kidney injury orpancreatitis) between the combination of oral sitagliptin plus basal insulin to the more labor-intensive basal-bolus insulin regimen (215).

 

Analysis from prospective studies using DPP4-i in various inpatient situations with type 2 diabetes (T2D) reported that treatment with DPP4-i alone or with basal insulin suggested they were safe and lowered glucose concentrations without increasing the risk of hypoglycemia (216; 217).

 

For people with type 2 diabetes hospitalized with heart failure, the ADA has recommended that the use of a sodium-glucose cotransporter 2 (SGLT2) inhibitor be initiated or continued during hospitalization and upon discharge if there are no contraindications and after recovery from the acute illness (218-221). In patients with acute heart failure, empagliflozin was well tolerated, resulting in significant clinical benefits, including heart failure readmissions and quality of life (221). However, SGLT2 inhibitors should be avoided in cases of severe illness, in people with type 1 diabetes, ketonemia, or ketonuria, and during prolonged fasting and surgical procedures. Proactive adjustment of diuretic dosing is recommended during hospitalization and/or discharge, especially in collaboration with a cardiology/heart failure consult team. The FDA has warned that SGLT2 inhibitors should be stopped three days before scheduled surgeries (4 days in the case of ertugliflozin) (222). This differs from the UK guideline, which states that these drugs should be omitted from the day before a procedure (223).

 

Staffing Levels in the Hospital

 

Inadequate levels of appropriately knowledgeable staff are a concern for patients with diabetes (224). An insufficient level of specialist diabetes staff is a factor that inhibits safe and optimal care (34). Recently, the UK JBDS group developed a simple calculator into which individual teams could enter data to calculate their staffing needs (49). That data showed the discrepancy between the number of people delivering care and the number of people that specialist teams felt was necessary to provide safe and effective care for five days or seven days per week. This was for senior medical staff, specialist nursing staff, dieticians, podiatrists, pharmacists and psychologists (49).

 

Glucose Monitoring in the Hospital

 

All patients admitted to the hospital with a diagnosis of diabetes and those with newly discovered hyperglycemia should be monitored closely (21). The frequency and method of monitoring and the schedule of the blood glucose checks will depend on the nutritional intake, patient treatment, and insulin schedule, as well as the ability of the individual to self-manage their diabetes (225). There is some controversy regarding the best method to monitor blood glucose. However,considering the convenience and wide availability of capillary point of care (POC) testing, we suggest this as the bestapproach if done with a monitoring device that has demonstrated accuracy (226-228). When using POC blood glucose meters, it is important to keep several things in mind. In particular, overall clinical conditions that might affect the POC value, such as hemoglobin level, perfusion, and medications, as well as the policy of the health care organization in guiding the patient and the staff on the use of POC devices or newer technologies.

 

Bedside point-of-care (POC) capillary glucose testing is usually ordered before meals and bedtime to assess glycemic control and adjust insulin therapy in the hospital (19; 228). However, this approach has been shown to fail to detect hypoglycemia, particularly nocturnal and asymptomatic hypoglycemia, which is a common scenario in the hospital setting (229; 230).

 

Continuous glucose monitoring (CGM) has increased over the last few years, helping to improve glycemic management in the ICU. The use of this technology was accelerated during the COVID pandemic, where the use of CGM meant that close contact with sick individuals was avoided using remote sensing (231-234). The use of CGM is questioned, with the accuracy of readings when dealing with hypoglycemia or in the operating room (235; 236). However, in general, most studies have been associated with overall benefit (234; 237; 238).

 

CGM is reliable compared to point-of-care testing and laboratory values in the inpatient setting. It is currently being evaluated for managing ICU and general ward patients (159; 185; 235; 239-242). Studies have shown that CGM offers advantages over intermittent capillary monitoring in the ICU. CGM can help identify and prevent severe hyperglycemia and hypoglycemia by allowing for more rapid and accurate adjustments to insulin infusions compared to capillary blood glucose testing. Research has also demonstrated that CGM is better at detecting hypoglycemia, predominantly asymptomatic and nocturnal hypoglycemia, than capillary glucose testing (243; 244). Additionally, CGM is as safe and effective as standard care in hospitalized patients and can lead to a significant decrease in recurrent hypoglycemia events compared with standard point-of-care testing (243; 245). Regulatory approval for CGM use in hospitals is still pending, but consensus guidelines suggest that the use of CGM in the hospital setting has the potential to provide a better glycemic assessment than capillary glucose testing (Walia et al.; other, Endo Soc Guidelines).  Furthermore, advanced technology in guiding insulin therapy using machine learning and artificial intelligence is being integrated more frequently into diabetes care (246). A proof-of-concept trial in patients with type 2 diabetes evaluated the efficacy and safety of a model-based reinforcement learning framework in titrating insulin dosing. After applying the intervention, the mean daily BG was lower by approximately 56 mg/dl (3 mmol/l) with no severe hypo- or hyperglycemia (247).

 

The American Diabetes Association (ADA) and UK JBDS recently recommended that people with diabetes who use a personal continuous glucose monitoring (CGM) device should be allowed to continue during hospitalization (48; 159; 248). Both organizations also recommend that confirmatory point-of-care (POC) glucose measurements be used for insulin dosing decisions, hypoglycemia assessment, and treatment.

 

A recent survey of inpatient teams across the UK showed significant variations in accessing and using technologies (249). These included networked glucose and ketone meters, and wearable diabetes technologies such as CGM, pumps, or closed loop systems. While almost two-thirds of respondents agreed that technology would help prevent hypoglycemia, there was a wide variety of specialist diabetes nursing or medical staff support available to help non-specialists, particularly on weekends or outside of regular working hours (249).

 

Medical Nutrition Therapy (MNT) in Hospitalized Patients with Diabetes

 

Medical nutrition therapy (MNT) is a key component of the comprehensive management of diabetes and hyperglycemia in the inpatient setting. Maintaining adequate nutrition is essential for glycemic control and to meet adequate caloric demands. Caloric demand in acute illness will differ from that in the outpatient setting. Achieving the proper nutritionalbalance in the inpatient setting is challenging. Anyone admitted to the hospital with diabetes or hyperglycemia should beassessed to determine the need for a modified diet to meet caloric demands.

 

The general approach to addressing MNT in the inpatient setting is usually based on expert opinions and patients' needs. Limited data exist regarding the best approach or method to achieve the ideal caloric supply. To determine the best approach, method, and caloric needs of their patients, providers should work closely with the nutrition professional.

 

All patients with diabetes or hyperglycemia should receive an individualized assessment. Most patients will generallyreceive adequate caloric needs with 3 discrete meals daily. Further, the metabolic need for patients with diabetes isusually provided by 25 to 35 calories/kg, whereas some critically ill patients might require less than 15 to 25 calories/kg per day (250; 251). A consistent carbohydrate meal-planning system might help to facilitate glycemic control and insulin dosing in the inpatient setting. Most patients require 1,500-2000 calories daily with 12-15 grams of carbohydrates per meal (19). Ideally, the carbohydrates should come from low glycemic index foods such as whole grains and vegetables.

 

Those individuals unable to achieve these goals should be evaluated to determine the need for enteral or parenteral nutrition. Enteral nutrition is the second-best option after oral nutrition and should be preferred over parenteral nutrition in hospitalized individuals (252-254). There are several advantages of enteral feeding versus parenteral feeding, including low cost, low risk of complications, a physiologic route, less risk for gastric mucosa atrophy, and lower risk of infectiousand thrombotic complications compared with the latter form of therapy (252-254). The benefit of parental nutrition has been documented in critically ill patients. However, some research has shown a detrimental effect on patients withdiabetes and hyperglycemia. Parental nutrition should be considered only in patients who cannot receive enteral nutritionand should be coordinated with the institution’s parenteral nutrition team. There has been guidance published in the surgical population on peri-operative nutrition, but the recommendations for people with diabetes is lacking because the literature remains scanty (251). A recent UK survey of diabetes teams showed no consensus on enteral feeding regimens (253). For those tube-fed, there were 3 main regimens:  continuous 24-hour feeding, a single feed with one break in 24 hours, or multiple feeds in 24 hours. In addition, there were multiple insulin regimens used: premixed insulin, isophane insulin, analog basal insulin, variable rate intravenous insulin, or basal-bolus insulin. None of these provided adequate glycemic control (253).

 

Enteral and parenteral nutrition  can prevent the effects of starvation and malnutrition (252). Enteral nutrition over parenteral nutrition is preferred whenever possible due to a lower risk of infectious and thrombotic complications (254-256). Standard enteral formulas reflect the reference values for macro- and micronutrients for a healthy population and contain 1-2 cal/ml. Most standard formulas contain whole protein, lipids in the form of long-chain triglycerides, andcarbohydrates. Standard diabetes-specific formulas provide low amounts of lipids (30% of total calories) combined with a high carbohydrate (257) content (55–60% of total calories); however, newer “diabetic” formulas have replaced part of carbohydrates with monounsaturated fatty acids (up to 35% of total calories) and also include 10-15 g/l dietary fiber andup to 30% fructose (257; 258).

 

“Diabetic” enteral formulas containing low-carbohydrate high–monounsaturated fatty acid (LCHM) are preferable to standard high-carbohydrate formulas in hospitalized patients with type 1 and type 2 diabetes (257; 258). In a meta-analysis of studies comparing relatively newer enteral LCHM formulas with older formulations, the postprandial rise inblood glucose was reduced by 18- 29 mg/dl [1.0-1.6 mmol/l] with the newer formulations (258). Table 4 depicts the composition of standard and diabetic-specific enteral formulas commonly used in hospitalized patients.

 

Table 4. Composition of Standard and Diabetic Specific Enteral Formulas Commonly Usedin Hospitalized Patients in the USA

 

Calories(kcal/mL)

Carbohydrate(g/l)

Fat (g/l)

Protein (g/l)

Manufacture

Standard formula

 

Jevity® 1.0 Cal

1.0

140

35

40

Abbott Nutrition

Nutren® 1.0

1.0

109

27

70

Nestle Nutrition

Osmolite® 1.2 Cal

1.2

158

39

56

Abbott Nutrition

Jevity® 1.2

1.2

169

39

56

Nestle Nutrition

Fibersource® HN

1.2

164

40

54

Nestle Nutrition

Isosource® 1.5 Cal

1.5

176

60

68

Nestle Nutrition

Jevity® 1.5

1.5

216

50

64

Nestle Nutrition

Diabetes specific formula

Glucerna® 1.0 Cal

1.0

75

54

50

Abbott Nutrition

Nutren® Glytrol®

1.0

100

48

45

Nestle Nutrition

Glucerna® 1.2 Cal

1.2

114

60

60

Abbott Nutrition

Diabetisource® AC

1.2

100

59

60

Nestle Nutrition

Glucerna® 1.5 Cal

1.5

133

75

83

Abbott Nutrition

             

The UK Joint British Diabetes Societies has updated its guidelines for the management of diabetes in enterally fed people (259).

 

Corticosteroid Therapy – Impact on Blood Glucose

 

Steroid use in hospitalized patients is common. A single-center cross-sectional study showed that 12.8% of all the people in the hospital were on glucocorticoids (260). Steroids may be administered by various regimes and at variable doses. A single daily dose of steroid (e.g., prednisolone/prednisone) in the morning may be the most standard mode ofadministration (205; 260-262). Limbachia et al. showed that, in susceptible individuals, steroid use will often result in arise in blood glucose by late morning that continues through to the evening (263). Overnight, the blood glucose generallyfalls back to baseline levels by the following day. They also showed the differential effects between different steroid types, with oral or IV dexamethasone or methylprednisolone leading to higher glucose excursions than prednisolone or hydrocortisone (262). Thus, treatment should be tailored to treating the hyperglycemia while avoiding nocturnal and earlymorning hypoglycemia. Multiple daily doses of steroid, be it intravenous hydrocortisone or oral dexamethasone, can cause a hyperglycemic effect throughout the 24-hour period. It may be, however, that a twice-daily premixed or basal-bolus regimen may need to be started if oral medication or once-daily insulin proves insufficient to control hyperglycemia (205). Close attention will therefore need to be paid to blood glucose monitoring, and early intervention may benecessary.

 

Glucose levels in most individuals can be predicted to rise approximately 4 to 8 hours following the administration of once-daily oral steroids and sooner following the administration of intravenous steroids. Again, capillary blood glucose monitoring is paramount to guide appropriate therapeutic interventions. Conversely, glucose levels may improve to pre-steroid levels 24 hours after intravenous steroids are discontinued. When oral steroids are weaned down, the glucose levels may decline in a dose-dependent fashion, but this may not occur, particularly in those with pre-existing undiagnosed diabetes.

 

At the commencement of steroid therapy, or for those already on a supraphysiological dose of corticosteroid, capillary blood glucose testing should occur before meals and at bedtime, in particular before lunch or evening meal, when thehyperglycemic effects of a morning dose of steroid are likely to be greatest (205; 262).

 

Subcutaneous insulin using a basal or multiple daily injection regimen will likely be the most appropriate choice for most patients to achieve glycemic control in the event of hyperglycemia. While the UK has advocated for short-acting sulfonylureas (205), the morning administration of basal human insulin may closely fit the glucose excursion induced by a single morning dose of oral steroid. Basal analog insulin may be appropriate if hyperglycemia is present for more prolonged periods. However, if long-acting insulin analogs are used in this context, care should be taken to identify and protect against hypoglycemia overnight and in the early morning. Subsequent titration of the insulin dose may be required to maintain glucose control in the face of increasing or decreasing the steroid dose.

 

When a patient is discharged from the hospital on steroid therapy, a clear strategy for managing hyperglycemia or potential hyperglycemia and the titration of treatment to address the hyperglycemia should be communicated to the community diabetes team and primary care team. Patients who commenced on steroids as inpatients and were discharged after a short stay with the intention of continuing high-dose steroids should receive standard diabetes education, encompassing the risks associated with hyperglycemia and hypoglycemia.

 

Closed Loop Technology

 

Several organizations have recommended that people who are well enough to do so should continue to use their insulin pumps in hospitals (108; 109; 240; 264). However, only a few recent studies have reported using closed-loop systems, also referred to as the artificial pancreas or automated insulin delivery systems, in hospitalized inpatients. Small randomized trials have reported good efficacy with improved time in target and lower mean daily blood glucose without an increased rate of hypoglycemia in the ICU (265-267) and non-ICU settings (268-271). However, some of these studies were done in those with type 2 diabetes (268; 270). In one non-ICU study, the time in the target range between 100-180 mg/dl (5.6-10.0 mmol/l) was reported as 59.8% in patients using the closed-loop technology compared to 38.1% with standard subcutaneous insulin regimen (269).

 

Similarly, a closed-loop study in patients receiving nutritional support also reported higher time in target glucose (68% vs 36.4%) and lower mean glucose values (153 vs 205 mg/dl [8.5-11.4 mmol/l]) compared to a standard insulin regimen (270). As with the use of CGM in the hospital, treatment with artificial pancreas is still experimental, and larger studies are needed to prove its safety and efficacy in ICU and non-ICU settings. Further challenges lie ahead because of the unfamiliarity of these systems, with non-specialist staff the primary carers for people with diabetes.

 

The ADA has recommended that insulin pumps or automated insulin delivery (closed-loop) systems be continued for hospitalized individuals with diabetes when clinically appropriate. Confirmatory POC blood glucose measurements should be used for insulin-dosing decisions and for assessing and treating hypoglycemia. However, this depends on the availability of required supplies and resources, proper training, ongoing competency assessments, and the implementation of institutional diabetes technology protocols (48).

 

As with the CGM, those who are well and can self-manage can look after their devices and diabetes. However, in those who are unwell or incapacitated, the systems must be disengaged from automatic and set to ‘manual’ mode to allow the diabetes teams to help manage the diabetes. The systems may not also be able to cope with the acute changes that occur in the hospital, including (but not limited to) change in oral carbohydrate intake, the use of glucocorticoids or other medications inducing insulin resistance; peri-operative use, nausea and vomiting; enteral or parenteral nutrition. Once again, the use of ‘manual’ mode is recommended in these situations, and the diabetes is managed in conjunction with the specialist diabetes team.

 

REFERENCES

 

  1. IDF Diabetes Atlas 2021 [article online], 2021. Available from https://diabetesatlas.org/.
  2. National Diabetes Statistics Report - 2024 [article online], 2024. Available from https://www.cdc.gov/diabetes/php/data-research/index.html.
  3. Koyama AK, Cheng YJ, Brinks R, Xie, Gregg EW, Hoyer A, Pavkov ME, Imperatore G. Trends in lifetime risk and years of potential life lost from diabetes in the United States, 1997–2018. PLOS ONE 2022;17:e0268805
  4. Pournaras DJ, Photi ES, Barnett N, Challand CP, Chatzizacharias NA, Dlamini NP, Doulias T, Foley A, Hernon J, Kumar B, Martin J, Nunney I, Panagiotopoulou I, Sengupta N, Shivakumar O, Sinclair P, Stather P, Than MM, Wells AC, Xanthis A, Dhatariya K. Assessing the quality of primary care referrals to surgery of patients with diabetes in the East of England: A multi-centre cross-sectional cohort study. International Journal of Clinical Practice 2017;71:e12971
  5. Jiang HJ, Stryer D, Friedman B, Andrews R. Multiple hospitalizations for patients with diabetes. Diabetes Care 2003;26:1421-1426
  6. Donnan PT, Leese GP, Morris AD. Hospitalizations for people with type 1 and type 2 diabetes compared with the nondiabetic population of Tayside, Scotland: a retrospective cohort study of resource use. Diabetes Care 2000;23:1774-1779
  7. Harding JL, Benoit SR, Gregg EW, Pavkov ME, Perreault L. Trends in rates of infections requiring hospitalization among adults with versus without diabetes in the U.S., 2000 - 2015. Diabetes Care 2020;43:106-116
  8. Umpierrez GE, Isaacs SD, Bazargan N, You X, Thaler LM, Kitabchi AE. Hyperglycemia: An independent marker of in-hospital mortality in patients with undiagnosed diabetes. Journal of Clinical Endocrinology Metabolism 2002;87:978-982
  9. National Diabetes Statistics Report 2022 [article online], 2022. Available from https://www.cdc.gov/diabetes/data/statistics-report/index.html.
  10. Zhang Y, McKeever Bullard K, Imperatore G, Holliday CS, Benoit SR. Proportions and trends of adult hospitalizations with diabetes, United States, 2000–2018. Diabetes Research and Clinical Practice 2022;187:109862
  11. National Diabetes Inpatient Audit (NaDIA) - 2019 [article online], 2020. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit/2019.
  12. Sampson MJ, Dozio N, Ferguson B, Dhatariya K. Total and excess bed occupancy by age, speciality and insulin use for nearly one million diabetes patients discharged from all English Acute Hospitals. Diabetes Research & Clinical Practice 2007;77:92-98
  13. American Diabetes Association. Economic costs of diabetes in the U.S. in 2017. Diabetes Care 2018;41:917-928
  14. Parker ED, Lin J, Mahoney T, Ume N, Yang G, Gabbay RA, ElSayed NA, Bannuru RR. Economic costs of diabetes in the U.S. in 2022. Diabetes Care 2023;47:26-43
  15. Friel KM, Gillespie P, Coates V, McCauley C, McCann M, O’Kane M, McGuigan K, Khamis A, Manktelow M. Estimating and examining the costs of inpatient diabetes care in an Irish Public Hospital. Diabetic Medicine 2022;39:e14753
  16. Bommer C, Heesemann E, Sagalova V, Manne-Goehler J, Atun R, Barnighausen T, Vollmer S. The global economic burden of diabetes in adults aged 20-79 years: a cost-of-illness study. Lancet Diabetes and Endocrinology 2017;5:423-430
  17. Bommer C, Sagalova V, Heesemann E, Manne-Goehler J, Atun R, Barnighausen T, Davies J, Vollmer S. Global economic burden of diabetes in adults: Projections from 2015 to 2030. Diabetes Care 2018;41:963-970
  18. Nolan JJ, O'Halloran D, McKenna TJ, Firth R, Redmond S. The cost of treating type 2 diabetes (CODEIRE). Irish Medical Journal 2006;99:307-310
  19. Umpierrez GE, Hellman R, Korytkowski MT, Kosiborod M, Maynard GA, Montori VM, Seley JJ, Van den Berghe G. Management of hyperglycemia in hospitalized patients in non-critical care setting: An Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology Metabolism 2012;97:16-38
  20. Moghissi ES, Korytkowski MT, Dinardo MM, Hellman R, Hirsch IB, Inzucchi S, Ismail-Beigi F, Kirkman MS, Umpierrez GE. American Association of Clinical Endocrinologists and American Diabetes Association consensus statement on inpatient glycemic control. Diabetes Care 2009;32:1119-1131
  21. Dhatariya K, James J, Kong M-F, Berrington R. Diabetes at the front door. A guideline for dealing with glucose related emergencies at the time of acute hospital admission from the Joint British Diabetes Society (JBDS) for Inpatient Care Group. Diabetic Medicine 2020;37:1578-1589
  22. Dhatariya K, Mustafa OG, Rayman G. Safe care for people with diabetes in hospital. Clinical Medicine 2020;20:21-27
  23. A good inpatient diabetes service [article online], 2019. Available from https://abcd.care/resource/jbds-14-good-inpatient-diabetes-service.
  24. NICE SUGAR investigators. Intensive versus conventional glucose control in critically ill patients. New England Journal of Medicine 2009;360:1283-1297
  25. Hulkower RD, Pollack RM, Zonszein J. Understanding hypoglycemia in hospitalized patients. Diabetes Management (Lond) 2014;4:165-176
  26. McDonnell ME, Umpierrez GE. Insulin therapy for the management of hyperglycemia in hospitalized patients. Endocrinology & Metabolism Clinics of North America 2012;41:175-201
  27. Qaseem A, Humphrey LL, Chou R, Snow V, Shekelle P, Physicians for the Clinical Guidelines Committee of the American College of Physicians. Use of intensive insulin therapy for the management of glycemic control in hospitalized patients: A clinical practice guideline from the American College of Physicians. Annals of Internal Medicine 2011;154:260-267
  28. Cook CB, Kongable GL, Potter DJ, Abad VJ, Leija DE, Anderson M. Inpatient glucose control: a glycemic survey of 126 U.S. hospitals. Journal of Hospital Medicine 2009;4:E7-E14
  29. NICE SUGAR investigators. Hypoglycemia and risk of death in critically ill patients. New England Journal of Medicine 2012;367:1108-1118
  30. Turchin A, Matheny ME, Shubina M, Scanlon JV, Greenwood B, Pendergrass ML. Hypoglycemia and clinical outcomes in patients with diabetes hospitalized in the general ward. Diabetes Care 2009;32:1153-1157
  31. Yamada T, Shojima N, Noma H, Yamauchi T, Kadowaki T. Glycemic control, mortality, and hypoglycemia in critically ill patients: a systematic review and network meta-analysis of randomized controlled trials. Intensive Care Medicine 2017;43:1-15
  32. Levy N, Hall GM. National guidance contributes to the high incidence of inpatient hypoglycaemia. Diabetic Medicine 2019;36:120-121
  33. National Diabetes Inpatient Audit - Harms 2020 England [article online], 2020. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit---harms/national-diabetes-inpatient-audit---harms-2020.
  34. National Diabetes Inpatient Safety Audit (NDISA) 2018-2021 [article online], 2022. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-safety-audit-ndisa/ndisa-2018-2021.
  35. Handelsman Y, Bloomgarden ZT, Grunberger G, Umpierrez G, Zimmerman RS, Bailey TS, Blonde L, Bray GA, Cohen AJ, Dagogo-Jack S, Davidson JA, Einhorn D, Ganda AP, Garber AJ, Garvey WT, Henry RR, Hirsch IB, Horton ES, Hurley DL, Jellinger PS, Jovanovic L, Lebovitz HE, LeRoith D, Levy P, McGill JB, Mechanick JI, Mestman JH, Moghissi ES, Orzeck EA, Pessah-Pollack R, Rosenblit PD, Vinik AI, Wyne K, Zangeneh F. American Association of Clinical Endocrinologists and American College of Endocrinology - clinical practice guidelines for developing a diabetes mellitus comprehensive care plan - 2015. Endocrine Practice 2015;21:1-87
  36. Jacobi J, Bircher N, Krinsley J, Agus M, Braithwaite SS, Duutschman C, Freire AX, Geehan D, Kohl B, Nasraway SA, Rigby M, Sands K, Schallom L, Taylor B, Umpierrez G, Mazuski J, Schunemann H. Guidelines for the use of an insulin infusion for the management of hyperglycemia in critically ill patients. Critical Care Medicine 2012;40:3251-3276
  37. Kitabchi AE, Umpierrez GE, Miles JM, Fisher JN. Hyperglycemic crises in adult patients with diabetes. Diabetes Care 2009;32:1335-1343
  38. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes Association. Diabetes care in the hospital: Standards of care in diabetes—2023. Diabetes Care 2023;46:S267-S278
  39. Swanson C, Potter D, Kongable G, Cook C. Update on inpatient glycemic control in hospitals in the United States. Endocrine Practice 2017;17:853-861
  40. Schmeltz LR, DeSantis AJ, Thiyagarajan V, Schmidt K, Shea-Mahler E, Johnson D, Henske J, McCarthy PM, Gleason TG, McGee EC, Molitch ME. Reduction of surgical mortality and morbidity in diabetic patients undergoing cardiac surgery with a combined intravenous and subcutaneous insulin glucose management strategy. Diabetes Care 2007;30:823-828
  41. Carpenter DL, Gregg SR, Xu K, Buchman TG, Coopersmith CM. Prevalence and impact of unknown diabetes in the ICU. Critical Care Medicine 2015;43:e541-e550
  42. Desgrouas M, Demiselle J, Stiel L, Brunot V, Marnai R, Sarfati S, Fiancette M, Lambiotte F, Thille AW, Leloup M, Clerc S, Beuret P, Bourion A-A, Daum J, Malhomme R, Ravan R, Sauneuf B, Rigaud J-P, Dequin P-F, Boulain T. Insulin therapy and blood glucose management in critically ill patients: a 1-day cross-sectional observational study in 69 French intensive care units. Annals of Intensive Care 2023;13:53
  43. Van Ackerbroeck S, Schepens T, Janssens K, Jorens PG, Verbrugge W, Collet S, Van Hoof V, Van Gaal L, De Block C. Incidence and predisposing factors for the development of disturbed glucose metabolism and DIabetes mellitus AFter Intensive Care admission: the DIAFIC study. Critical Care 2015;19:355
  44. Kar P, Plummer MP, Ali Abdelhamid Y, Giersch EJ, Summers MJ, Weinel LM, Finnis ME, Phillips LK, Jones KL, Horowitz M, Deane AM. Incident diabetes in survivors of critical illness and mechanisms underlying persistent glucose intolerance: A prospective cohort study. Critical Care Medicine 2019;47:e103-e111
  45. Gomez AM, Imitola Madero A, Henao Carrillo DC, Rondon M, Munoz OM, Robledo MA, Rebolledo M, Garcia Jaramillo M, Leon Vargas F, Umpierrez G. Hypoglycemia incidence and factors associated in a cohort of patients with type 2 diabetes hospitalized in general ward treated with basal bolus insulin regimen assessed by continuous glucose monitoring. Journal of Diabetes Science and Technology 2020;14:233-239
  46. Greci LS, Kailasam M, Malkani S, Katz DL, Hulinsky I, Ahmadi R, Nawaz H. Utility of HbA1c levels for diabetes case finding in hospitalized patients with hyperglycemia. Diabetes Care 2003;26:1064-1068
  47. Mazurek JA, Hailpern SM, Goring T, Nordin C. Prevalence of hemoglobin A1c greater than 6.5% and 7.0% among hospitalized patients without known diagnosis of diabetes at an urban inner city hospital. Journal of Clinical Endocrinology & Metabolism 2010;95:1344-1348
  48. American Diabetes Association. 16. Diabetes care in the hospital: Standards of Care in Diabetes–2025. Diabetes Care 2025;48 (Suppl 1).
  49. Dashora U, Flanagan D, Rayman G, Mustafa OG, Walden E, Dhatariya K, The Joint British Diabetes Societies for Inpatient Care. Optimal staffing for a good quality inpatient diabetes service. Diabetic Medicine 2023;40:e15151
  50. Cryer PE. Hypoglycemia, functional brain failure, and brain death. Journal of Clinical Investigation 2014;117:868-870
  51. Amiel SA, Aschner P, Childs B, Cryer PE, de Galan BE, Frier BM, Gonder-Frederick L, Heller SR, Jones T, Khunti K, Leiter LA, Luo Y, McCrimmon RJ, Pedersen-bjergaard U, Seaquist ER, Zoungas S. Hypoglycaemia, cardiovascular disease, and mortality in diabetes: epidemiology, pathogenesis, and management. Lancet Diabetes & Endocrinology 2019;7:385-396
  52. Corssmit EP, Romijn JA, Sauerwein HP. Regulation of glucose production with special attention to nonclassical regulatory mechanisms: A review. Metabolism 2014;50:742-755
  53. Boden G. Gluconeogenesis and glycogenolysis in health and diabetes. Journal of Investigative Medicine 2004;52:375-378
  54. Gerich JE, Meyer C, Woerle HJ, Stumvoll M. Renal gluconeogenesis. Diabetes Care 2001;24:382-391
  55. van de Werve G, Jeanrenaud B. Liver glycogen metabolism: An overview. Diabetes/Metabolism Reviews 1987;3:47-78
  56. Gerich JE, Lorenzi M, Bier DM, Tsalikian E, Schneider V, Karam JH, Forsham PH. Effects of physiologic levels of glucagon and growth hormone on human carbohydrate and lipid metabolism. Studies involving administration of exogenous hormone during suppression of endogenous hormone secretion with somatostatin. Journal of Clinical Investigation 1976;57:875-884
  57. Rizza RA, Mandarino LJ, Gerich JE. Dose-response characteristics for effects of insulin on production and utilization of glucose in man. American Journal of Physiology - Endocrinology & Metabolism 1981;240:E630-E639
  58. Dhatariya KK, Glaser NS, Codner E, Umpierrez GE. Diabetic ketoacidosis. Nature Reviews Disease Primers 2020;6:40
  59. Dungan KM, Braithwaite SS, Preiser J-C. Stress hyperglycaemia. Lancet 2009;373:1798-1807
  60. McCowen KC, Malhotra A, Bistrian BR. Stress-induced hyperglycemia. Critical Care Clinics 2001;17:107-124
  61. Stentz FB, Umpierrez GE, Cuervo R, Kitabchi AE. Proinflammatory cytokines, markers of cardiovascular risks, oxidative stress, and lipid peroxidation in patients with hyperglycemic crises. Diabetes 2004;53:2079-2086
  62. Chaudhuri A, Umpierrez GE. Oxidative stress and inflammation in hyperglycemic crises and resolution with insulin: implications for the acute and chronic complications of hyperglycemia. Journal of Diabetes and its Complications 2012;26:257-258
  63. Reyes-Umpierrez D, Davis G, Cardona S, Pasquel FJ, Peng L, Jacobs S, Vellanki P, Fayfman M, Haw S, Halkos M, Guyton RA, Thourani VH, Umpierrez GE. Inflammation and oxidative stress in cardiac surgery patients treated to intensive versus conservative glucose targets. Journal of Clinical Endocrinology & Metabolism 2016;102:309-315
  64. Esposito K, Nappo F, Marfella R, Giugliano G, Giugliano F, Ciotola M, Quagliaro L, Ceriello A, Giugliano D. Inflammatory cytokine concentrations are acutely increased by hyperglycemia in humans: Role of oxidative stress. Circulation 2002;106:2067-2072
  65. Fan J, Li YH, Wojnar MM, Lang CH. Endotoxin-induced alterations in insulin-stimulated phosphorylation of insulin receptor, IRS-1, and MAP kinase in skeletal muscle. Shock 1996;6:164-170
  66. Evans NR, Dhatariya KK. Assessing the relationship between admission glucose levels, subsequent length of hospital stay, readmission and mortality. Clinical Medicine 2012;12:137-139
  67. Haddadin F, Clark A, Evans N, Dhatariya K. Admission blood glucose helps predict 1 year, but not 2 years, mortality in an unselected cohort of acute general medical admissions. International Journal of Clinical Practice 2014;69:643-648
  68. Frisch A, Chandra P, Smiley D, Peng L, Rizzo M, Gatcliffe C, Hudson M, Mendoza J, Johnson R, Lin E, Umpierrez GE. Prevalence and clinical outcome of hyperglycemia in the perioperative period in noncardiac surgery. Diabetes Care 2010;33:1783-1788
  69. Kotagal M, Symons RG, Hirsch IB, Umpierrez GE, Dellinger EP, Farrokhi ET, Flum DR. Perioperative hyperglycemia and risk of adverse events among patients with and without diabetes. Annals of Surgery 2016;261:97-103
  70. Umpierrez G, Cardona S, Pasquel F, Jacobs S, Peng L, Unigwe M, Newton CA, Smiley-Byrd D, Vellanki P, Halkos M, Puskas JD, Guyton RA, Thourani VH. Randomized controlled trial of intensive versus conservative glucose control in patients undergoing coronary artery bypass graft surgery: GLUCO-CABG trial. Diabetes Care 2015;38:1665-1672
  71. Umpierrez GE, Smiley D, Jacobs S, Peng L, Temponi A, Mulligan P, Umpierrez D, Newton C, Olson D, Rizzo M. Randomized study of basal-bolus insulin therapy in the inpatient management of patients with type 2 diabetes undergoing general surgery (RABBIT 2 Surgery). Diabetes Care 2011;34:256-261
  72. Krinsley JS. Association between hyperglycemia and increased hospital mortality in a heterogeneous population of critically ill patients. Mayo Clinic Proceedings 2003;78:1471-1478
  73. Furnary AP, Zerr KJ, Grunkemeier GL, Starr A. Continuous intravenous insulin infusion reduces the incidence of deep sternal wound infection in diabetic patients after cardiac surgical procedures. Annals of Thoracic Surgery 1999;67:352-362
  74. Montori VM, Bistrian BR, McMahon MM. Hyperglycemia in acutely ill patients. JAMA 2002;288:2167-2169
  75. Adie SK, Ketcham SW, Marshall VD, Farina N, Sukul D. The association of glucose control on in-hospital mortality in the cardiac intensive care unit. Journal of Diabetes and its Complications 2023;37:108453
  76. Levy N, Dhatariya K. Pre-operative optimisation of the surgical patient with diagnosed and undiagnosed diabetes: a practical review. Anaesthesia 2019;74:58-66
  77. Falciglia M, Freyberg RW, Almenoff PL, D'Alessio DA, Render ML. Hyperglycemia-related mortality in critically ill patients varies with admission diagnosis. Critical Care Medicine 2009;37:3001-3009
  78. Bruno A, Gregori D, Caropreso A, Lazzarato F, Petrinco M, Pagano E. Normal glucose values are associated with a lower risk of mortality in hospitalized patients. Diabetes Care 2008;31:2209-2210
  79. Critchley JA, Carey IM, Harris T, DeWilde S, Hosking FJ, Cook DG. Glycemic control and risk of infections among people with type 1 or type 2 diabetes in a large primary care cohort study. Diabetes Care 2018;41:2127-2135
  80. Blaha J, Mraz M, Kopecky P, Stritesky M, Lips M, Matias M, Kunstyr J, Porizka M, Kotulak T, Kolnikova I, Simanovska B, Zakharchenko M, Rulisek J, Sachl R, Anyz J, Novak D, Linder J, Hovorka R, Svacina S, Haluzik M. Perioperative tight glucose control reduces postoperative adverse events in nondiabetic cardiac surgery patients. Journal of Clinical Endocrinology & Metabolism 2015;100:3081-3089
  81. Barmanray RD, Cheuk N, Fourlanos S, Greenberg PB, Colman PG, Worth LJ. In-hospital hyperglycemia but not diabetes mellitus alone is associated with increased in-hospital mortality in community-acquired pneumonia (CAP): a systematic review and meta-analysis of observational studies prior to COVID-19. BMJ Open Diabetes Research & Care 2022;10:e002880
  82. Shiffermiller J, Anderson M, Thompson R. Postoperative length of stay in patients with stress hyperglycemia compared to patients with diabetic hyperglycemia: A retrospective cohort study. Journal of Diabetes Science and Technology 2024;18:556-561
  83. Jones CE, Graham LA, Morris MS, Richman JS, Hollis RH, Wahl TS, Copeland LA, Burns EA, Itani KM, Hawn MT. Association between preoperative hemoglobin A1c levels, postoperative hyperglycemia, and readmissions following gastrointestinal surgery. JAMA Surgery 2017;152:1031-1038
  84. Rains JL, Jain SK. Oxidative stress, insulin signaling, and diabetes. Free Radical Biology and Medicine 2011;50:567-575
  85. Meza CA, La Favor JD, Kim DH, Hickner RC. Endothelial dysfunction: Is there a hyperglycemia-induced imbalance of NOX and NOS? International  Journal of Molecular Science 2019;20
  86. Furnary AP, Gao G, Grunkemeier GL, Wu Y, Zerr KJ, Bookin SO, Floten HS, Starr A. Continuous insulin infusion reduces mortality in patients with diabetes undergoing coronary artery bypass grafting. Journal of Thoracic & Cardiovascular Surgery 2003;125:1007-1021
  87. Krinsley JS, Maurer P, Holewinski S, Hayes R, McComsey D, Umpierrez GE, Nasraway SA. Glucose control, diabetes status, and mortality in critically ill patients. Mayo Clinic Proceedings 2017;92:1019-1029
  88. van Vught LA, Holman R, de Jong E, de Keizer NF, van der Poll T. Diabetes is not associated with increased 90-day mortality risk in critically ill patients with sepsis. Critical Care Medicine 2017;45:e1026-e1035
  89. Gunst J, Debaveye Y, Güiza F, Dubois J, De Bruyn A, Dauwe D, De Troy E, Casaer MP, De Vlieger G, Haghedooren R, Jacobs B, Meyfroidt G, Ingels C, Muller J, Vlasselaers D, Desmet L, Mebis L, Wouters PJ, Stessel B, Geebelen L, Vandenbrande J, Brands M, Gruyters I, Geerts E, De Pauw I, Vermassen J, Peperstraete H, Hoste E, De Waele JJ, Herck I, Depuydt P, Wilmer A, Hermans G, Benoit DD, Van den Berghe G. Tight blood-glucose control without early parenteral nutrition in the ICU. New England Journal of Medicine 2023;389:1180-1190
  90. Mehta G, Lo B, Memarpour M, Chan A. Impact of diabetes on inpatient length of stay in adult mental health services in a community hospital setting: A retrospective cohort study. Canadian Journal of Diabetes 2022;46:678-682
  91. McAlister FA, Majumdar SR, Blitz S, Rowe BH, Romney J, Marrie TJ. The relation between hyperglycemia and outcomes in 2,471 patients admitted to the hospital with community-acquired pneumonia. Diabetes Care 2005;28:810-815
  92. Baker EH, Janaway CH, Philips BJ, Brennan AL, Baines DL, Wood DM, Jones PW. Hyperglycaemia is associated with poor outcomes in patients admitted to hospital with acute exacerbations of chronic obstructive pulmonary disease. Thorax 2006;61:284-289
  93. Noordzij PG, Boersma E, Schreiner F, Kertai MD, Feringa HH, Dunkelgrun M, Bax JJ, Klein J, Poldermans D. Increased preoperative glucose levels are associated with perioperative mortality in patients undergoing noncardiac, nonvascular surgery. European Journal of Endocrinology 2007;156:137-142
  94. Martin ET, Kaye KS, Knott C, Nguyen H, Santarossa M, Evans R, Bertran E, Jaber L. Diabetes and risk of surgical site infection: A systematic review and meta-analysis. Infection Control & Hospital Epidemiology 2016;37:88-99
  95. Pomposelli JJ, Baxter JK, Babineau TJ, Pomfret EA, Driscoll DF, Forse RA, Bistrian BR. Early postoperative glucose control predicts nosocomial infection rate in diabetic patients. Journal of Parenteral and Enteral Nutrition 1998;22:77-81
  96. Ramos M, Khalpey Z, Lipsitz S, Steinberg J, Panizales MT, Zinner M, Rogers SO. Relationship of perioperative hyperglycemia and postoperative infections in patients who undergo general and vascular surgery. Annals of Surgery 2015;248:585-591
  97. Sadoskas D, Suder NC, Wukich DK. Perioperative glycemic control and the effect on surgical site infections in diabetic patients undergoing foot and ankle surgery. Foot and Ankle Specialist 2016;9:24-30
  98. Wallia A, Schmidt K, Oakes DJ, Pollack T, Welsh N, Kling-Colson S, Gupta S, Fulkerson C, Aleppo G, Parikh N, Levitsky J, Norvell JP, Rademaker A, Molitch ME. Glycemic control reduces infections in post-liver transplant patients: Results of a prospective, randomized study. Journal of Clinical Endocrinology & Metabolism 2017;102:451-459
  99. Sheen YJ, Huang CC, Huang SC, Hung MD, Lin CH, Lee IT, Lin SY, Sheu WH. Implimentation of an electronic dashboard with a remote management system to improve glycemic management among hospitalized adults. Endocrine Practice 2020;26:179-191
  100. Garg R, Schuman B, Bader A, Hurwitz S, Turchin A, Underwood P, Metzger C, Rein R, Lortie M. Effect of preoperative diabetes management on glycemic control and clinical outcomes after elective surgery. Annals of Surgery 2018;267:858-862
  101. Gregory NS, Seley JJ, Ukena J, Shah S, Fred MR, Dargar SK, Mauer E, Kim RJ. Decreased rates of inpatient hypoglycemia following implementation of an automated tool in the electronic medical record for identifying root causes. Journal of Diabetes Science and Technology 2018;12:63-68
  102. Pichardo-Lowden A, Umpierrez G, Lehman EB, Bolton MD, DeFlitch CJ, Chinchilli VM, Haidet PM. Clinical decision support to improve management of diabetes and dysglycemia in the hospital: a path to optimizing practice and outcomes. BMJ Open Diabetes Research & Care 2021;9:e001557
  103. Wang YY, Hu SF, Ying HM, Chen L, Li HL, Tian F, Zhou ZF. Postoperative tight glycemic control significantly reduces postoperative infection rates in patients undergoing surgery: A meta-analysis. BMC Endocrine Disorders 2018;18:42
  104. Vanhorebeek I, Gunst J, Van den Berghe G. Critical care management of stress-induced hyperglycemia. Current Diabetes Reports 2018;18:17
  105. Gunst J, De Bruyn A, Van den Berghe G. Glucose control in the ICU. Current Opinion in Anesthesiology 2019;32:156-162
  106. Dashora U, George S, Sampson M, Walden E. National guidelines have contributed to safer care for inpatients with diabetes. Diabetic Medicine 2019;36:124-126
  107. Honarmand K, Sirimaturos M, Hirshberg EL, Bircher NG, Agus MS, Carpenter DL, Downs CR, Farrington EA, Freire AX, Grow A, Irving SY, Krinsley JS, Lanspa MJ, Long MT, Nagpal D, Preiser JC, Srinivasan V, Umpierrez GE, Jacobi J. Society of Critical Care Medicine guidelines on glycemic control for critically ill children and adults 2024. Critical Care Medicine 2024;52:e161-e181
  108. Association AD. Diabetes care in the hospital: Standards of care in diabetes—2024. Diabetes Care 2023;47:S295-S306
  109. Korytkowski MT, Muniyappa R, Antinori-Lent K, Donihi AC, Drincic AT, Hirsch IB, Luger A, McDonnell ME, Murad MH, Nielsen C, Pegg C, Rushakoff RJ, Santesso N, Umpierrez GE. Management of hyperglycemia in hospitalized adult patients in non-critical care settings: An Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology & Metabolism 2022;107:2101–2128
  110. Lazar HL, McDonnell M, Chipkin SR, Furnary AP, Engelman RM, Sadhu AR, Bridges CR, Haan CK, Svedjeholm R, Taegtmeyer H, Shemin RJ. The Society of Thoracic Surgeons Practice Guideline Series: Blood glucose management during adult cardiac surgery. Annals of Thoracic Surgery 2009;87:663-669
  111. Guidelines for the management of inpatient diabetes [article online], 2022. Available from https://abcd.care/jbds-ip.
  112. End of life diabetes care [article online], 2021. Available from https://www.diabetes.org.uk/professionals/position-statements-reports/diagnosis-ongoing-management-monitoring/end-of-life-care.
  113. Van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, Vlasselaers D, Ferdinande P, Bouillon R, Lauwers P. Intensive insulin therapy in critically ill patients. New England Journal of Medicine 2001;345:1359-1367
  114. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, Van Wijngaerden E, Bobbaers H, Bouillon R. Intensive insulin therapy in the medical ICU. New England Journal of Medicine 2006;354:449-461
  115. Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U, John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M, Hartog C, Natanson C, Loeffler M, Reinhart K. Intensive insulin therapy and pentastarch resuscitation in severe sepsis. New England Journal of Medicine 2008;358:125-139
  116. De La Rosa G, Donado JH, Restrepo AH, Quintero AM, Gonzalez LG, Saldarriaga NE, Bedoya M, Toro JM, Velasquez JB, Valencia JC, Arango CM, Aleman PH, VAsquez EM, Chavarriaga JC, Yepes A, Pulido W, Cadavid CA. Strict glycaemic control in patients hospitalised in a mixed medical and surgical intensive care unit: a randomised clinical trial. Critical Care 2008;12:R120
  117. Preiser J-C, Brunkhorst F. Tight glucose control and hypoglycemia. Critical Care Medicine 2008;36:1391-1392
  118. Marik PE. Tight glycemic control in acutely ill patients: low evidence of benefit, high evidence of harm! Intensive Care Medicine 2016;42:1475-1477
  119. Umpierrez GE. Glucose control in the ICU. New England Journal of Medicine 2023;389:1234-1237
  120. Krinsley JS, Rule P, Pappy L, Ahmed A, Huley-Rodrigues C, Prevedello D, Preiser J-C. The interaction of acute and chronic glycemia on the relationship of hyperglycemia, hypoglycemia, and glucose variability to mortality in the critically ill. Critical Care Medicine 2020;48:1744-1751
  121. Malmberg KA, Efendic S, Ryden LE. Feasibility of insulin-glucose infusion in diabetic patients with acute myocardial infarction: A report from the multicenter trial: DIGAMI. Diabetes Care 1994;17:1007-1014
  122. Lazar HL, Chipkin SR, Fitzgerald CA, Bao Y, Cabral H, Apstein CS. Tight glycemic control in diabetic coronary artery bypass graft patients improves perioperative outcomes and decreases recurrent ischemic events. Circulation 2004;109:1497-1502
  123. Gandhi GY, Nuttall GA, Abel MD, Mullany CJ, Schaff HV, O'Brien PC, Johnson MG, Williams AR, Cutshall SM, Mundy LM, Rizza RA, McMahon MM. Intensive intraoperative insulin therapy versus conventional glucose management during cardiac surgery: A randomized trial. Annals of Internal Medicine 2007;146:233-243
  124. Preiser J-C, Devos P, Ruiz-Santana S, Melot C, Annane D, Groeneveld J, Iapichino G, Leverve XM, Nitenberg G, Singer P, Wernerman J, Joannidis M, Stecher A, Chiolero R. A prospective randomised multi-centre controlled trial on tight glucose control by intensive insulin therapy in adult intensive care units: the Glucontrol study. Intensive Care Medicine 2009;35:1738-1748
  125. Agus MS, Steil GM, Wypij D, Costello JM, Laussen PC, Langer M, Alexander JL, Scoppettuolo LA, Pigula FA, Charpie JR, Ohye RG, Gaies MG. Tight glycemic control versus standard care after pediatric cardiac surgery. New England Journal of Medicine 2012;367:1208-1219
  126. Agus MS, Wypij D, Hirshberg EL, Srinivasan V, Faustino EV, Luckett PM, Alexander JL, Asaro LA, Curley MA, Steil GM, Nadkarni VM. Tight glycemic control in critically ill children. New England Journal of Medicine 2017;376:729-741
  127. Macrae D, Grieve R, Allen E, Sadique Z, Morris K, Pappachan J, Parslow R, Tasker RC, Elbourne D. A randomized trial of hyperglycemic control in pediatric intensive care. New England Journal of Medicine 2014;370:107-118
  128. Kalfon P, Giraudeau B, Ichai C, Guerrini A, Brechot N, Cinotti R, Dequin P-F, Riu-Poulenc B, Montravers P, Annane D, Dupont H, Sorine M, Riou B. Tight computerized versus conventional glucose control in the ICU: a randomized controlled trial. Intensive Care Medicine 2014;40:171-181
  129. Cinotti R, Ichai C, Orban JC, Kalfon P, Feuillet F, Roquilly A, Riou B, Blanloeil Y, Asehnoune K, Rozec B. Effects of tight computerized glucose control on neurological outcome in severely brain injured patients: A multicenter sub-group analysis of the randomized-controlled open-label CGAO-REA study. Critical Care 2014;18:498
  130. Okabayashi T, Shima Y, Sumiyoshi T, Kozuki A, Tokumaru T, Iiyama T, Sugimoto T, Kobayashi M, Yokoyama M, Hanazaki K. Intensive versus intermediate glucose control in surgical intensive care unit patients. Diabetes Care 2014;37:1516
  131. Cardona S, Pasquel FJ, Fayfman M, Peng L, Jacobs S, Vellanki P, Weaver J, Halkos M, Guyton RA, Thourani VH, Umpierrez GE. Hospitalization costs and clinical outcomes in CABG patients treated with intensive insulin therapy. Journal of Diabetes and its Complications 2017;31:742-747
  132. Dhatariya K. Should inpatient hyperglycaemia be treated? British Medical Journal 2013;346:f134
  133. Phillips VL, Byrd AL, Adeel S, Peng L, Smiley DD, Umpierrez GE. A comparison of inpatient cost per day in general surgery patients with type 2 diabetes treated with basal-bolus versus sliding scale insulin regimens. PharmacoEconomics - Open 2017;1:109-115
  134. Barmanray RD, Kyi M, Colman PG, Rowan L, Raviskanthan M, Collins L, Donaldson L, Montalto S, Tsan J, Sun E, Le M, Worth LJ, Thomson B, Fourlanos S. The specialist treatment of inpatients: Caring for diabetes in surgery (STOIC-D Surgery) trial: A randomized controlled trial of early intervention with an electronic specialist-led model of diabetes care. Diabetes Care 2024;47:948-955
  135. Cryer PE, Axelrod L, Grossman AB, Heller SR, Montori VM, Seaquist ER, Service FJ. Evaluation and management of adult hypoglycemic disorders: An Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology & Metabolism 2009;94:709-728
  136. Seaquist ER, Anderson J, Childs B, Cryer P, Dagogo-Jack S, Fish L, Heller SR, Rodriguez H, Rosenzweig J, Vigersky R. Hypoglycemia and diabetes: A report of a workgroup of the American Diabetes Association and The Endocrine Society. Diabetes Care 2013;36:1384-1395
  137. The hospital management of hypoglycaemia in adults with diabetes mellitus. Revised April 2021 [article online], 2021. Available from https://abcd.care/resource/current/jbds-01-hospital-management-hypoglycaemia-adults-diabetes-mellitus.
  138. Krikorian A, Ismail-Beigi F, Moghissi ES. Comparisons of different insulin infusion protocols: a review of recent literature. Current Opinion in Clinical Nutrition & Metabolic Care 2010;13:198-204
  139. Umpierrez GE, Smiley D, Zisman A, Prieto LM, Palacio A, Ceron M, Puig A, Mejia R. Randomized study of basal-bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial). Diabetes Care 2007;30:2181-2186
  140. Rajendran R, Kerry C, Rayman G, on behalf of the MAGIC group. Temporal patterns of hypoglycaemia and burden of sulfonylurea-related hypoglycaemia in UK hospitals: a retrospective multicentre audit of hospitalised patients with diabetes. BMJ Open 2014;4
  141. National Diabetes Inpatient Audit (NaDIA) - 2017 [article online], 2018. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit/national-diabetes-inpatient-audit-nadia-2017.
  142. Krinsley JS, Grover A. Severe hypoglycemia in critically ill patients: Risk factors and outcomes. Critical Care Medicine 2007;35:2262-2267
  143. Kagansky N, Levy S, Rimon E. Hypoglycemia as a predictor of mortality in hospitalized elderly patients. Archives of Internal Medicine 2003;163:1825-1829
  144. Lake A, Arthur A, Byrne C, Davenport K, Yamamoto JM, Murphy HR. The effect of hypoglycaemia during hospital admission on health-related outcomes for people with diabetes: a systematic review and meta-analysis. Diabetic Medicine 2019;36:1349-1359
  145. Dendy J, Chockalingam V, Tirumalasetty N, Dornelles A, Blonde L, Bolton P, Meadows R, Andrews S. Identifying risk factors for severe hypoglycemia in hospitalized patients with diabetes. Endocrine Practice 2014;20:1051-1056
  146. Smith WD, Winterstein AG, Johns T, Rosenberg E, Sauer BC. Causes of hyperglycemia and hypoglycemia in adult inpatients. American Journal of Health-System Pharmacy 2005;62:714-719
  147. Maynard G, Lee J, Phillips G, Fink E, Renvall M. Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: Effect of structured subcutaneous insulin orders and an insulin management algorithm. Journal of Hospital Medicine 2009;4:3-15
  148. Insulin safety. Reducing harm associated with the unsafe use of insulin products [article online], 2010. Available from https://www.centreformedicinesoptimisation.co.uk/new-insulin-safety-guidance-issued-to-reduce-wrong-dosages/#:~:text=Learning%20from%20previous%20incidents%20like,insulin%20administration%20times%3B%20and%20putting.
  149. Griesdale DE, de Souza RJ, van Dam RM, Heyland DK, Cook DJ, Malhotra A, Dhaliwal R, Henderson WR, Chittock DR, Finfer S, Talmor D. Intensive insulin therapy and mortality among critically ill patients: a meta-analysis including NICE-SUGAR study data. Canadian Medical Association Journal 2009;180:821-827
  150. Zoungas S, Patel A, Chalmers J, de Galan BE, Li Q, Billot L, Woodward M, Ninomiya T, Neal B, MacMahon S, Grobbee DE, Kengne AP, Marre M, Heller S. Severe hypoglycemia and risks of vascular events and death. New England Journal of Medicine 2010;363:1410-1418
  151. Wiener RS, Wiener DC, Larson RJ. Benefits and risks of tight glucose control in critically ill adults: A meta-analysis. JAMA 2008;300:933-944
  152. Kosiborod M, Inzucchi S, Goyal A, Krumholz HM, Masoudi FA, Xiao L, Spertus JA. Relationship between spontaneous and iatrogenic hypoglycemia and mortality in patients hospitalized with acute myocardial infarction. JAMA 2009;301:1556-1564
  153. Arabi YM, Tamin HM, Rishu AH. Hypoglycemia with intensive insulin therapy in critically ill patients: Predisposing factors and association with mortality. Critical Care Medicine 2009;37:2536-2544
  154. Vriesendorp TM, DeVries JH, van Santen S, Moeniralam HS, de Jong E, Roos YB, Schultz MJ, Rosendaal FR, Hoekstra JB. Evaluation of short-term consequences of hypoglycemia in an intensive care unit. Critical Care Medicine 2006;34:2714-2718
  155. Krinsley JS, Schultz MJ, Spronk PE, Harmsen RE, van Braam Houckgeest F, van der Sluijs JP, Melot C, Preiser JC. Mild hypoglycemia is independently associated with increased mortality in the critically ill. Critical Care 2011;15:R173
  156. National Diabetes Inpatient Audit (NaDIA) - 2016 [article online], 2017. Available from http://www.content.digital.nhs.uk/catalogue/PUB23539.
  157. National Diabetes Inpatient Audit - Harms, 2019 [article online], 2020. Available from https://digital.nhs.uk/data-and-information/publications/statistical/national-diabetes-inpatient-audit---harms/national-diabetes-inpatient-audit---harms-2019/2019.
  158. Dashora U, Sampson M, Castro E, Stanisstreet D, Hillson R. The best hypoglycaemia avoidance initiative in the UK. British Journal of Diabetes 2017;17:74-77
  159. Flanagan D, Avari P, Choudhary P, Lumb A, Misra S, Rayman G, Dhatariya K. Using technology to improve diabetes care in hospital: The challenge and the opportunity. Journal of Diabetes Science and Technology 2023;17:503-508
  160. Gill GV, Woodward A, Casson IF, Weston PJ. Cardiac arrhythmia and nocturnal hypoglycaemia in type 1 diabetes—the 'dead in bed' syndrome revisited. Diabetologia 2009;52:42-45
  161. Desouza C, Salazar H, Cheong B, Murgo J, Fonseca V. Association of hypoglycemia and cardiac ischemia. Diabetes Care 2003;26:1485-1489
  162. Petersen KG, Schluter KJ, Kerp L. Regulation of serum potassium during insulin-induced hypoglycemia. Diabetes 1982;31:615-617
  163. Razavi Nematollahi L, Kitabchi AE, Stentz FB, Wan JY, Larijani BA, Tehrani MM, Gozashti MH, Omidfar K, Taheri E. Proinflammatory cytokines in response to insulin-induced hypoglycemic stress in healthy subjects. Metabolism - Clinical and Experimental 2009;58:443-448
  164. Desouza CV, Bolli GB, Fonseca V. Hypoglycemia, diabetes, and cardiovascular events. Diabetes Care 2010;33:1389-1394
  165. Boucai L, Southern WN, Zonszein J. Hypoglycemia-associated mortality is not drug-associated but linked to comorbidities. American Journal of Medicine 2011;124:1028-1035
  166. George JT, Warriner D, McGrane DJ, Rozario KS, Price HC, Wilmot EG, Kar P, Stratton IM, Jude EB, McKay GA. Lack of confidence among trainee doctors in the management of diabetes: the Trainees Own Perception of Delivery of Care (TOPDOC) Diabetes Study. QJM 2011;104:761-766
  167. Horton WB, Law S, Darji M, Conaway MR, Akbashev MY, Kubiak NT, Kirby JL, Thigpen SC. A multicenter study evaluating perceptions and knowledge of inpatient glycemic control among resident physicians: analyzing themes to inform and improve care. Endocrine Practice 2019;25:1295-1303
  168. Dei Cas A, Aldigeri R, Ridolfi V, Vazzana A, Ciardullo AV, Manicardi V, Sforza A, Tomasi F, Zavaroni D, Zavaroni I, Bonadonna RC. Efficacy of a training programme for the management of diabetes mellitus in the hospital: A randomized study (stage 2 of GOVEPAZ healthcare). Diabetes/Metabolism Research and Reviews 2023;39:e3708
  169. Hommel I, Wollersheim H, Tack CJ, Mulder J, van Gurp PJ, Hulscher ME. Impact of a multifaceted strategy to improve perioperative diabetes care. Diabetic Medicine 2017;34:278-285
  170. Brown G, Dodek P. Intravenous insulin nomogram improves blood glucose control in the critically ill. Critical Care Medicine 2001;29:1714-1719
  171. Rea RS, Donihi AC, Bobeck M, Herout P, McKaveney TP, Kane-Gill SL, Korytkowski MT. Implementing an intravenous insulin infusion protocol in the intensive care unit. American Journal of Health-System Pharmacy 2007;64:385-395
  172. George S, Dale J, Stanisstreet D. A guideline for the use of variable rate intravenous insulin infusion in medical inpatients. Diabetic Medicine 2015;32:706-713
  173. Miller EE, Lalla M, Zaidi A, Elgash M, Zhao H, Rubin DJ. Eating and glycemic control among critically ill patients receiving continuous intravenous insulin. Endocrine Practice 2020;26:43-50
  174. Salinas PD, Mendez CE. Glucose management technologies for the critically ill. Journal of Diabetes Science and Technology 2019;13:682-690
  175. Ullal J, Aloi JA. Subcutaneous insulin dosing calculators for inpatient glucose control. Current Diabetes Reports 2019;19:120
  176. Bouw JW, Campbell N, Hull MA, Juneja R, Guzman O, Overholser BR. A retrospective cohort study of a nurse-driven computerized insulin infusion program versus a paper-based protocol in critically ill patients. Diabetes Technology & Therapeutics 2012;14:125-130
  177. Juneja R, Roudebush C, Kumar N, Macy A, Golas A, Wall D, Wolverton C, Nelson D, Carroll J, Flanders SJ. Utilization of a computerized intravenous insulin infusion program to control blood glucose in the intensive care unit. Diabetes Technology & Therapeutics 2007;9:232-240
  178. Marvin MR, Inzucchi SE, Besterman BJ. Computerization of the Yale insulin infusion protocol and potential insights into causes of hypoglycemia with intravenous insulin. Diabetes Technology & Therapeutics 2013;15:246-252
  179. Bouldin MG, Hong B, Setji T, Greenlee J, Cooper A, Thompson J, Capes K. Evaluation of the efficacy and safety of an eglycemic management system in a community hospital setting. Journal of Diabetes Science and Technology 2021;15:236-241
  180. Goldberg PA, Siegel MD, Sherwin RS, Halickman JI, Lee M, Bailey VA, Lee SL, Dziura JD, Inzucchi SE. Implementation of a safe and effective insulin infusion protocol in a medical intensive care unit. Diabetes Care 2004;27:461-467
  181. Boutin JM, Gauthier L. Insulin infusion therapy in critically ill patients. Canadian Journal of Diabetes 2014;38:144-150
  182. DeSantis AJ, Schmeltz LR, Schmidt K, O'Shea-Mahler E, Rhee C, Wells A, Brandt S, Peterson S, Molitch ME. Inpatient management of hyperglycemia: The Northwestern experience. Endocrine Practice 2006;12:491-505
  183. Davidson PC, Steed RD, Bode BW. Glucommander. A computer-directed intravenous insulin system shown to be safe, simple, and effective in 120,618 h of operation. Diabetes Care 2005;28:2418-2423
  184. Newton CA, Smiley D, Bode BW, Kitabchi AE, Davidson PC, Jacobs S, Steed RD, Stentz F, Peng L, Mulligan P, Freire AX, Temponi A, Umpierrez GE. A comparison study of continuous insulin infusion protocols in the medical intensive care unit: Computer-guided vs. standard column-based algorithms. Journal of Hospital Medicine 2010;5:432-437
  185. Leelarathna L, English SW, Thabit H, Caldwell K, Allen JM, Kumareswaran K, Wilinska ME, Nodale M, Mangat J, Evans ML, Burnstein R, Hovorka R. Feasibility of fully automated closed-loop glucose control using continuous subcutaneous glucose measurements in critical illness: a randomized controlled trial. Critical Care 2013;17:R159
  186. Gómez Medina AM, Henao-Carrillo DC, Yepes C, Silva J, Gómez González JA, Cortes D, Robledo S, Mejía G, Rondon M. Glycemic control metrics in a cohort of hospitalized patients with type 1 diabetes using hybrid closed-loop and advanced hybrid closed-loop systems. Diabetes Research and Clinical Practice 2023;204:110897
  187. Avari P, Lumb A, Flanagan D, Rayman G, Misra S, Choudhary P, Dhatariya K. Insulin pumps and hybrid close loop systems within hospital: A scoping review and practical guidance From the Joint British Diabetes Societies for Inpatient Care. Journal of Diabetes Science and Technology 2023;17:625-634
  188. Migdal AL, Fortin-Leung C, Pasquel F, Wang H, Peng L, Umpierrez GE. Inpatient glycemic control with sliding scale insulin in noncritical patients with type 2 diabetes: Who can slide? Journal of Hospital Medicine 2021;16:462-468
  189. Migdal AL, Idrees T, Umpierrez GE. Selecting insulin regimens for the management of non-ICU patients with type 2 diabetes. Journal of the Endocrine Society 2021;5
  190. Hirsch IB. Sliding scale insulin--time to stop sliding. JAMA 2009;301:213-214
  191. Umpierrez GE, Maynard G. Glycemic chaos (not glycemic control) still the rule for inpatient care. How do we stop the insanity? Journal of Hospital Medicine 2006;1:141-144
  192. Swan E, Dhatariya K. Differential effects of intravenous and subcutaneous sliding scale insulin regimes used to improve blood glucose levels in a tertiary care setting. Practical Diabetes International 2009;26:2i
  193. Vellanki P, Cardona S, Galindo RJ, Urrutia MA, Pasquel FJ, Davis GM, Fayfman M, Migdal A, Peng L, Umpierrez GE. Efficacy and safety of intensive versus nonintensive supplemental insulin with a basal-bolus insulin regimen in hospitalized patients with type 2 diabetes: A randomized clinical study. Diabetes Care 2022;45:2217-2223
  194. King AB, Armstrong DU. Basal bolus dosing: A clinical experience. Current Diabetes Reviews 2005;1:215-220
  195. Galindo RJ, Dhatariya K, Gomez-Peralta F, Umpierrez GE. Safety and efficacy of inpatient diabetes management with non‑insulin agents: an overview of international practices. Current Diabetes Reports 2022;22:237-246
  196. Umpierrez GE, Hor T, Smiley D, Temponi A, Umpierrez D, Ceron M, Munoz C, Newton C, Peng L, Baldwin D. Comparison of inpatient insulin regimens with detemir plus aspart versus neutral protamine hagedorn plus regular in medical patients with type 2 diabetes. Journal of Clinical Endocrinology Metabolism 2009;94:564-569
  197. Umpierrez GE, Smiley D, Hermayer K, Khan A, Olson DE, Newton C, Jacobs S, Rizzo M, Peng L, Reyes D, Pinzon I, Fereira ME, Hunt V, Gore A, Toyoshima MT, Fonseca VA. Randomized study comparing a basal-bolus with a basal plus correction insulin regimen for the hospital management of medical and surgical patients with type 2 diabetes: Basal plus trial. Diabetes Care 2013;36:2169-2174
  198. Pietras S, Hanrahan P, Arnold L, Sternthal E, McDonnell M. State-of-the-art inpatient diabetes care: The evolution of an academic hospital. Endocrine Practice 2010;16:512-521
  199. Clement S, Bowen-Wright H. Twenty-four hour action of insulin glargine (Lantus) may be too short for once-daily dosing: A case report. Diabetes Care 2002;25:1479-1147a
  200. Rubin DJ, Rybin D, Doros G, McDonnell ME. Weight-based, insulin dose-related hypoglycemia in hospitalized patients with diabetes. Diabetes Care 2011;34:1723-1728
  201. Koufakis T, Mustafa OG, Zebekakis P, Kotsa K. Oral antidiabetes agents for the management of inpatient hyperglycaemia: so far, yet so close. Diabetic Medicine 2020;37:1418-1426
  202. Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn R. Thiazolidinedione use, fluid retention, and congestive heart failure. Diabetes Care 2004;27:256-263
  203. Khalam A, Dilip C, Shinu C. Drug use evaluation of diabetes mellitus in hospitalized patients of a tertiary care referral hospital. Journal of Basic and Clinical Physiology and Pharmacology 2012;23:173-177
  204. Willi SM, Kennedy A, Brant BP, Wallace P, Rogers NL, Garvey WT. Effective use of thiazolidinediones for the treatment of glucocorticoid-induced diabetes. Diabetes Research & Clinical Practice 2002;58:87-96
  205. Roberts A, James J, Dhatariya K, on behalf of the Joint british Diabetes Societies for Inpatient Care Group. Management of hyperglycaemia and steroid (glucocorticoid) therapy: a guideline from the Joint British Diabetes Societies (JBDS) for Inpatient Care group. Diabetic Medicine 2018;35:1011-1017
  206. Nirantharakumar K, Marshall T, Kennedy A, Hemming K, Coleman JJ. Hypoglycaemia is associated with increased length of stay and mortality in people with diabetes who are hospitalized. Diabetic Medicine 2012;29:e445-e448
  207. Deusenberry CM, Coley KC, Korytkowski MT, Donihi AC. Hypoglycemia in hospitalized patients treated with sulfonylureas. Pharmacotherapy 2012;32:613-617
  208. Stuart K, Adderley NJ, Marshall T, Rayman G, Sitch A, Manley S, Ghosh S, Toulis KA, Nirantharakumar K. Predicting inpatient hypoglycaemia in hospitalized patients with diabetes: a retrospective analysis of 9584 admissions with diabetes. Diabetic Medicine 2017;34:1385-1391
  209. Roumie CL, Greevy RA, Grijalva CG, Hung AM, Liu X, Murff HJ, Elasy TA, Griffin MR. Association between intensification of metformin treatment with insulin vs sulfonylureas and cardiovascular events and all-cause mortality among patients with diabetes. JAMA 2014;311:2288-2296
  210. Gerards MC, Venema GE, Patberg KW, Kross M, Potter van Loon BJ, Hageman IM, Snijders D, Brandjes DP, Hoekstra JB, Vriesendorp TM, Gerdes VE. Dapagliflozin for prednisone-induced hyperglycemia in acute exacerbation of chronic obstructive pulmonary disease. Diabetes, Obesity and Metabolism 2018;20:1306-1310
  211. Empagliflozin to be investigated as a possible treatment for COVID-19 in the RECOVERY trial [article online], 2022. Available from https://www.recoverytrial.net/news/empagliflozin-to-be-investigated-as-a-possible-treatment-for-covid-19-in-the-recovery-trial.
  212. Cohen B, Harris YT, Schulman-Rosenbaum R. SGLT2 inhibitors should be avoided for inpatient management of hyperglycemia. Endocrine Practice 2024;
  213. Dhatariya K. Initiation and continuation of sodium-glucose cotransporter 2 inhibitors in hospital inpatients – ready for prime time? Diabetes Care 2022;45:2806-2807
  214. Umpierrez GE, Gianchandani R, Smiley D, Wesorick DH, Newton C, Farrokhi F, Peng L, Lathkar-Pradhan S, Pasquel F. Safety and efficacy of sitagliptin therapy for the inpatient management of general medicine and surgery patients with type 2 diabetes: A pilot, randomized, controlled study. Diabetes Care 2013;36:3430-3435
  215. Pasquel FJ, Gianchandani R, Rubin DJ, Dungan KM, Anzola I, Gomez PC, Peng L, Hodish I, Bodnar T, Wesorick D, Balakrishnan V, Osei K, Umpierrez GE. Efficacy of sitagliptin for the hospital management of general medicine and surgery patients with type 2 diabetes (Sita-Hospital): a multicentre, prospective, open-label, non-inferiority randomised trial. Lancet Diabetes and Endocrinology 2017;5:125-133
  216. Lorenzo-Gonzalez C, Atienza-Sanchez E, Reyes-Umpierrez D, Vellanki P, Davis GM, Pasquel FJ, Cardona S, Fayman M, Peng L, Umpierrez GE. Safety and efficacy of DDP-4 inhibitors for management of hospitalized general medicine and surgery patients with type 2 diabetes. Endocrine Practice 2020;26:722-728
  217. Hulst AH, Plummer MP, Hollmann MW, DeVries JH, Preckel B, Deane AM, Hermanides J. Systematic review of incretin therapy during peri-operative and intensive care. Critical Care 2018;22:299
  218. Kosiborod MN, Angermann CE, Collins SP, Teerlink JR, Ponikowski P, Biegus J, Comin-Colet J, Ferreira JP, Mentz RJ, Nassif ME, Psotka MA, Tromp J, Brueckmann M, Blatchford JP, Salsali A, Voors AA. Effects of empagliflozin on symptoms, physical limitations and quality of life in patients hospitalized for acute heart failure - results from the EMPULSE trial. Circulation 2022;146
  219. Tamaki S, Yamada T, Watanabe T, Morita T, Furukawa Y, Kawasaki M, Kikuchi A, Kawai T, Seo M, Abe M, Nakamura J, Yamamoto K, Kayama K, Kawahira M, Tanabe K, Fujikawa K, Hata M, Fujita Y, Umayahara Y, Taniuchi S, Sanada S, Shintani A, Fukunami M. Effect of empagliflozin as an add-on therapy on decongestion and renal function in patients with diabetes hospitalized for acute decompensated heart failure. Circulation: Heart Failure 2021;14:e007048
  220. Cunningham JW, Vaduganathan M, Claggett BL, Kulac IJ, Desai AS, Jhund PS, de Boer RA, DeMets D, Hernandez AF, Inzucchi SE, Kosiborod MN, Lam CS, Martinez F, Shah SJ, McGrath MM, O’Meara E, Wilderäng U, Lindholm D, Petersson M, Langkilde A, McMurray JJ, Solomon SD. Dapagliflozin in patients recently hospitalized with heart failure and mildly reduced or preserved ejection fraction. Journal of the American College of Cardiology 2022;80:1302-1310
  221. Voors AA, Angermann CE, Teerlink JR, Collins SP, Kosiborod M, Biegus J, Ferreira JP, Nassif ME, Psotka MA, Tromp J, Borleffs CJ, Ma C, Comin-Colet J, Fu M, Janssens SP, Kiss RG, Mentz RJ, Sakata Y, Schirmer H, Schou M, Schulze PC, Spinarova L, Volterrani M, Wranicz JK, Zeymer U, Zieroth S, Brueckmann M, Blatchford JP, Salsali A, Ponikowski P. The SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure: a multinational randomized trial. Nature Medicine 2022;28:568-574
  222. FDA revises labels of SGLT2 inhibitors for diabetes to include warnings about too much acid in the blood and serious urinary tract infections [article online], 2022. Available from https://www.fda.gov/Drugs/DrugSafety/ucm475463.htm.
  223. Guideline for perioperative care for people with diabetes mellitus undergoing elective and emergency surgery [article online], 2022. Available from https://cpoc.org.uk/sites/cpoc/files/documents/2022-12/CPOC-Diabetes-Guideline-Updated2022.pdf.
  224. Rutter CL, Jones C, Dhatariya KK, James J, Irvine L, Wilson EC, Singh H, Walden E, Holland R, Harvey I, Bradley C, Sampson MJ. Determining in-patient diabetes treatment satisfaction in the UK—the DIPSat study. Diabetic Medicine 2013;30:731-738
  225. Avari P, Lumb A, Flanagan D, Rayman G, Misra S, Dhatariya K, Choudhary P. Continuous glucose monitoring within hospital: A scoping review and summary of guidelines from the Joint British Diabetes Societies for Inpatient Care. Journal of Diabetes Science and Technology 2023;17:611-624
  226. Rice MJ, Smith JL, Coursin DB. Glucose measurement in the ICU: Regulatory intersects reality. Critical Care Medicine 2017;45:741-743
  227. Pilackas K, El-Oshar S, Carter C. Clinical reliability of point-of-care glucose testing in critically ill patients. Journal of Diabetes Science and Technology 2020;14:65-69
  228. Misra S, Avari P, Lumb A, Flanagan D, Choudhary P, Rayman G, Dhatariya K. How can point-of-care technologies support in-hospital diabetes care? Journal of Diabetes Science and Technology 2023;17:509-516
  229. Gomez AM, Umpierrez GE. Continuous glucose monitoring in insulin-treated patients in non-ICU settings. Journal of Diabetes Science and Technology 2014;8:930-936
  230. Cardona S, Gomez PC, Vellanki P, Anzola I, Ramos C, Urrutia MA, Haw JS, Fayfman M, Wang H, Galindo RJ, Pasquel FJ, Umpierrez GE. Clinical characteristics and outcomes of symptomatic and asymptomatic hypoglycemia in hospitalized patients with diabetes. BMJ Open Diabetes Research & Care 2018;6:e000607
  231. Galindo RJ, Aleppo G, Klonoff DC, Spanakis EK, Agarwal S, Vellanki P, Olson DE, Umpierrez GE, Davis GM, Pasquel FJ. Implementation of continuous glucose monitoring in the hospital: Emergent considerations for remote glucose monitoring during the COVID-19 pandemic. Journal of Diabetes Science and Technology 2020;14:822-832
  232. Chow KW, Kelly DJ, Rieff MC, Skala PA, Kravets I, Charitou MM, Morley EJ, Gupta R, Miller JD. Outcomes and healthcare provider perceptions of real-time continuous glucose monitoring (rtCGM) in patients with diabetes and COVID-19 admitted to the ICU. Journal of Diabetes Science and Technology 2021;15:607-614
  233. Klarskov CK, Kristensen PL. Experience from implementing telemetric in-hospital continuous glucose monitoring during the COVID-19 pandemic. Journal of Diabetes Science and Technology 2021;15:715-716
  234. Faulds ER, Boutsicaris A, Sumner L, Jones L, McNett M, Smetana KS, May CC, Buschur E, Exline MC, Ringel MD, Dungan K. Use of continuous glucose monitor in critically ill COVID-19 patients requiring insulin infusion: An observational study. Journal of Clinical Endocrinology & Metabolism 2021;106:e4007-e4016
  235. Perez-Guzman MC, Duggan E, Gibanica S, Cardona S, Corujo-Rodriguez A, Faloye A, Halkos M, Umpierrez GE, Peng L, Davis GM, Pasquel FJ. Continuous glucose monitoring in the operating room and cardiac intensive care unit. Diabetes Care 2021;44:e50-e52
  236. Longo RR, Elias H, Khan M, Seley JJ. Use and accuracy of inpatient CGM during the COVID-19 pandemic: An observational study of general medicine and ICU patients. Journal of Diabetes Science and Technology 2022;16:1136-1143
  237. Sweeney AT, Pena S, Sandeep J, Hernandez B, Chen Y, Breeze JL, Bulut A, Feghali K, Abdelrehim M, Abdelazeem M, Srivoleti P, Salvucci L, Cann SB, Norman C. Use of a continuous glucose monitoring system in high-risk hospitalized noncritically ill patients with diabetes after cardiac surgery and during their transition of care from the intensive care unit during COVID-19: A pilot study. Endocrine Practice 2022;28:615-621
  238. Tingsarat W, Buranasupkajorn P, Khovidhunkit W, Boonchaya-anant P, Laichuthai N. The accuracy of continuous glucose monitoring in the medical intensive care unit. Journal of Diabetes Science and Technology 2022;16:1550-1554
  239. Wernerman J, Desaive T, Finfer S, Foubert L, Furnary A, Holzinger U, Hovorka R, Joseph J, Kosiborod M, Krinsley J, Mesotten D, Nasraway S, Rooyackers O, Schultz M, Van Herpe T, Vigersky R, Preiser JC. Continuous glucose control in the ICU: report of a 2013 round table meeting. Critical Care 2014;18:226
  240. Clinical Guideline: Guidelines for managing continuous subcutaneous insulin infusion (CSII, or 'insulin pump') therapy in hospitalised patients [article online], 2019. Available from https://abcd.care/sites/abcd.care/files/CSII_DTN_FINAL%20210218.pdf.
  241. Wang M, Singh LG, Spanakis EK. Advancing the use of CGM devices in a non-ICU setting. Journal of Diabetes Science and Technology 2019;13:674-681
  242. Tsur A, Cahn A, Israel M, Feldhamer I, Hammerman A, Pollack R. Impact of flash glucose monitoring on glucose control and hospitalization in type 1 diabetes: A nationwide cohort study. Diabetes/Metabolism Research and Reviews 2021;37:e3355
  243. Spanakis EK, Urrutia A, Galindo RJ, Vellanki P, Migdal AL, Davis G, Fayfman M, Idrees T, Pasquel FJ, Coronado WZ, Albury B, Moreno E, Singh LG, Marcano I, Lizama S, Gothong C, Munir K, Chesney C, Maguire R, Scott WH, Perez-Guzman MC, Cardona S, Peng L, Umpierrez GE. Continuous glucose monitoring–guided insulin administration in hospitalized patients with diabetes: A randomized clinical trial. Diabetes Care 2022;45:2369-2375
  244. Pasquel FJ, Lansang MC, Dhatariya K, Umpierrez GE. Management of diabetes and hyperglycaemia in the hospital. Lancet Diabetes & Endocrinology 2021;9:174-188
  245. Singh LG, Satyarengga M, Marcano I, Scott WH, Pinault LF, Feng Z, Sorkin JD, Umpierrez GE, Spanakis EK. Reducing inpatient hypoglycemia in the general wards using real-time continuous glucose monitoring: The glucose telemetry system, a randomized clinical trial. Diabetes Care 2020;43:2736-2743
  246. Davis GM, Galindo RJ, Migdal AL, Umpierrez GE. Diabetes technology in the inpatient setting for management of hyperglycemia. Endocrinology and Metabolism Clinics 2020;49:79-93
  247. Wang G, Liu X, Ying Z, Yang G, Chen Z, Liu Z, Zhang M, Yan H, Lu Y, Gao Y, Xue K, Li X, Chen Y. Optimized glycemic control of type 2 diabetes with reinforcement learning: a proof-of-concept trial. Nature Medicine 2023;29:2633-2642
  248. Using technology to support diabetes care in hospital: A guideline from the Joint British Diabetes Societies for Inpatient Care (JBDS-IP) Group and Diabetes Technology Network (DTN) [article online], 2024. Available from https://abcd.care/resource/current/jbds-20-using-technology-support-diabetes-care-hospital.
  249. Lumb A, Misra S, Rayman G, Avari P, Flanagan D, Choudhary P, Dhatariya K. Variation in the current use of technology to support diabetes management in UK hospitals: Results of a survey of health care professionals. Journal of Diabetes Science and Technology 2023;17:733-741
  250. McMahon MM, Nystrom E, Braunschweig C, Miles J, Compher C, Nutrition ASfPaE. A.S.P.E.N. clinical guidelines: Nutrition support of adult patients with hyperglycemia. Journal of Parenteral and Enteral Nutrition 2013;37:23-36
  251. Lobo DN, Gianotti L, Adiamah A, Barazzoni R, Deutz NE, Dhatariya K, Greenhaff PL, Hiesmayr M, Hjort Jakobsen D, Klek S, Krznaric Z, Ljungqvist O, McMillan DC, Rollins KE, Panisic Sekeljic M, Skipworth RJ, Stanga Z, Stockley A, Stockley R, Weimann A. Perioperative nutrition: Recommendations from the ESPEN expert group. Clinical Nutrition 2020;39:3211-3227
  252. JBDS guideline on the glycaemic management during the inpatient enteral feeding of stroke patients with diabetes [article online], 2012. Available from http://www.diabetologists-abcd.org.uk/JBDS/JBDS_IP_Enteral_Feeding_Stroke.pdf.
  253. Flanagan D, Lake AK, Green M, Roberts A, Dhatariya K, on behalf of the Joint British Diabetes Societies for Inpatient Care Group. A UK national survey of enteral feed use in people with diabetes 2022. Diabetic Medicine 2023;n/a:e15216
  254. McMahon MM, Rizza RA. Nutrition support in hospitalized patients with diabetes mellitus. Mayo Clinic Proceedings 1996;71:587-594
  255. Singer P, Blaser AR, Berger MM, Alhazzani W, Calder PC, Casaer MP, Hiesmayr M, Mayer K, Montejo JC, Pichard C, Preiser J-C, van Zanten AR, Oczkowski S, Szczeklik W, Bischoff SC. ESPEN guideline on clinical nutrition in the intensive care unit. Clinical Nutrition 2019;38:48-79
  256. Preiser J-C, Arabi YM, Berger MM, Casaer M, McClave S, Montejo-González JC, Peake S, Reintam Blaser A, Van den Berghe G, van Zanten A, Wernerman J, Wischmeyer P. A guide to enteral nutrition in intensive care units: 10 expert tips for the daily practice. Critical Care 2021;25:424
  257. Via MA, Mechanick JI. Inpatient enteral and parental nutrition for patients with diabetes. Current Diabetes Reports 2011;11:99-105
  258. Elia M, Ceriello A, Laube H, Sinclair AJ, Engfer M, Stratton RJ. Enteral nutritional support and use of diabetes-specific formulas for patients with diabetes: A systematic review and meta-analysis. Diabetes Care 2005;28:2267-2279
  259. Glycaemic management during the inpatient enteral feeding of stroke patients with diabetes [article online], 2024. Available from https://abcd.care/resource/jbds-05-glycaemic-management-during-inpatient-enteral-feeding-stroke-patients-diabetes.
  260. Narwani V, Swafe L, Stavraka C, Dhatariya K. How frequently are bedside glucose levels measured in hospital inpatients on glucocorticoids? Clinical Medicine 2014;14:327-328
  261. Sudlow A, O'Connor HM, Narwani V, Swafe L, Dhatariya K. Assessing the prevalence of dexamethasone use in patients undergoing surgery, and subsequent glucose measurements: a retrospective cohort study. Practical Diabetes 2017;34:117-121
  262. Limbachia V, Nunney I, Page DJ, Barton HA, Patel LK, Thomason GN, Green SL, Lewis KF, Dhatariya K. The effect of different types of oral or intravenous corticosteroids on capillary blood glucose levels in hospitalized inpatients with and without diabetes. Clinical Therapeutics 2024;46
  263. Limbachia V, Nunney I, Page DJ, Barton HA, Patel LK, Thomason GN, Green SL, Lewis KF, Dhatariya K. The effect of different types of oral or intravenous corticosteroids on capillary blood glucose levels in hospitalized inpatients with and without diabetes. Clinical Therapeutics 2024;46:e59-363
  264. Flanagan D, Dhatariya K, Kilvert A, on behalf of the Joint British Diabetes Societies for Inpatient Care (JBDS-IP) Group. Self-management of diabetes in hospital: a guideline from the Joint British Diabetes Societies (JBDS) for Inpatient Care group. Diabetic Medicine 2018;35:992-996
  265. Yatabe T, Yamazaki R, Kitagawa H, Okabayashi T, Yamashita K, Hanazaki K, Yokoama M. The evaluation of the ability of closed-loop glycemic control device to maintain the blood glucose concentration in intensive care unit patients. Critical Care Medicine 2011;39:575-578
  266. Levitt DL, Spanakis EK, Ryan KA, Silver KD. Insulin pump and continuous glucose monitor initiation in hospitalized patients with type 2 diabetes mellitus. Diabetes Technology & Therapeutics 2018;20:32-38
  267. Pelkey MN, Boyle ME, Long A, Castro JC, Cook CB, Thompson B. Hybrid closed loop insulin pump technology can be safely used In the inpatient setting. Endocrine Practice 2023;29:24-28
  268. Bally L, Thabit H, Hartnell S, Andereggen E, Ruan Y, Wilinska ME, Evans ML, Wertli MM, Coll AP, Stettler C, Hovorka R. Closed-loop insulin delivery for glycemic control in noncritical care. New England Journal of Medicine 2018;379:547-556
  269. Thabit H, Hartnell S, Allen JM, Lake A, Wilinska ME, Ruan Y, Evans ML, Coll AP, Hovorka R. Closed-loop insulin delivery in inpatients with type 2 diabetes: a randomised, parallel-group trial. Lancet Diabetes & Endocrinology 2020;5:117-124
  270. Boughton CK, Bally L, Martignoni F, Hartnell S, Herzig D, Vogt A, Wertli MM, Wilinska ME, Evans ML, Coll AP, Stettler C, Hovorka R. Fully closed-loop insulin delivery in inpatients receiving nutritional support: a two-centre, open-label, randomised controlled trial. Lancet Diabetes & Endocrinology 2019;7:368-377
  271. Stewart ZA, Wilinska ME, Hartnell S, Temple RC, Rayman G, Stanley KP, Simmons D, Law GR, Scott EM, Hovorka R, Murphy HR. Closed-loop insulin delivery during pregnancy in women with type 1 diabetes. New England Journal of Medicine 2016;375:644-654

 

Assessing Insulin Sensitivity and Resistance in Humans

ABSTRACT

 

In this chapter we discuss a representative variety of methods currently available for estimating insulin sensitivity/resistance. These range from complex, time consuming, labor-intensive, invasive procedures to simple testsinvolving a single fasting blood sample. It is important to understand the physiological concepts informing each method so that relative merits and limitations of particular approaches are appropriately matched with proposed applications and data is interpreted correctly. The glucose clamp method is the reference standard for direct measurement of insulin sensitivity. Regarding simple surrogates, QUICKI and Log (HOMA) are among the best and most extensively validated. Dynamic tests are useful if information about both insulin secretion and insulin action are needed.

 

INTRODUCTION

 

Insulin resistance plays a major pathophysiological role in type 2 diabetes and is tightly associated with major public health problems including obesity, hypertension, coronary artery disease, dyslipidemias, and a cluster of metabolic and cardiovascular abnormalities that define the metabolic syndrome (1, 2, 3).

 

A global epidemic of obesity is driving the increased incidence and prevalence of type 2 diabetes and its cardiovascular complications (4). Insulin resistance is commonly associated with visceral adiposity, glucose intolerance, hypertension, dyslipidemia, hypercoagulable state, endothelial dysfunction, and/or elevated markers of inflammation. Therefore, the presence of these clinical abnormalities is usually characteristic of an insulin resistant state. In addition to clinical manifestations of the “Insulin Resistance Syndrome,” insulin resistance predisposes to accelerated cardiovascular disease (CVD). Therefore, it is of great importance to develop tools for quantifying insulin sensitivity/resistance in humans that may be used to appropriately investigate the epidemiology, pathophysiological mechanisms, outcomes of therapeutic interventions, and clinical course of patients with insulin resistance (5). In this chapter, we will discuss some currently used methods for assessing insulin sensitivity, their applications, merits, and limitations.

 

INSULIN SENSITIVITY AND RESISTANCE

 

Metabolic actions of insulin help to maintain glucose homeostasis and promote glucose utilization (6). Insulin increases glucose utilization in peripheral organs (e.g., skeletal muscle and adipose tissue) and suppresses hepatic glucose production (HGP) and adipose tissue lipolysis. In addition to these classical metabolic target tissues, insulin has many other important physiological targets. These include the brain, pancreatic β-cells, heart, and vascular endothelium that help to coordinate and couple metabolic and cardiovascular homeostasis under healthy conditions (6-9). Insulin has concentration- dependent saturable actions to increase whole-body glucose disposal. The maximal effect of insulin defines “insulin responsiveness” while the insulin concentration required for a half-maximal response defines “insulin sensitivity” (Fig. 1). Although, other actions of insulin on fat and amino-acid metabolism, cardiovascular, kidney, and brain function also exhibit a concentration-dependent response, the term “insulin sensitivity” typically refers to insulin’s metabolic actions to promote glucose disposal.

 

The concept of insulin resistance was proposed as early as 1936 to describe diabetic patients requiring high doses of insulin (10). Insulin resistance is typically defined as decreased sensitivity and/or responsiveness to insulin- mediatedglucose disposal and/or inhibition of HGP and adipose tissue lipolysis. Rigorous evaluation of altered sensitivity and responsiveness therefore requires a comparison of insulin dose-response curves.

 

Figure 1. Schematic representation of concentration-response relationships between plasma insulin concentrations and insulin-mediated whole-body glucose disposal. Curve a: normal insulin sensitivity and responsiveness. Curve b: rightward shift in insulin concentration-response curve. This represents decreased insulin sensitivity (increased EC50) with normal insulin responsiveness. Curve c: Decreased insulin sensitivity (increased EC50) and reduced insulin responsiveness. Curve d: Leftward shift in the insulin concentration- response curve. This represents increased insulin sensitivity (decreased EC50) with normal insulin responsiveness.

 

DIRECT MEASURES OF INSULIN SENSITIVITY

 

Hyperinsulinemic Euglycemic Glucose Clamp

 

PROCEDURE

 

The glucose clamp technique, originally developed by Andres and DeFronzo is widely accepted as the reference standard for directly determining metabolic insulin sensitivity in humans (11). After an overnight fast, insulin is infused intravenously at a constant rate that may range from 5 - 120 mU/m2/min (dose per body surface area per minute). This constant insulin infusion results in a new steady-state insulin level that is above the fasting level (hyperinsulinemic). As a consequence, glucose disposal in skeletal muscle and adipose tissue is increased while HGP is suppressed. Under these conditions, a bedside glucose analyzer is used to frequently monitor blood glucose levels at 5 – 10 min intervals while 20% dextrose is given intravenously at a variable rate in order to “clamp” blood glucose concentrations in the normal range (euglycemic). An infusion of potassium phosphate is also given to prevent hypokalemia resulting from hyperinsulinemia and increased glucose disposal. After several hours of constant insulin infusion, steady-state conditions are typically achieved for plasma insulin, blood glucose, and the glucose infusion rate (GIR). Assuming that the hyperinsulinemic state is sufficient to completely suppress hepatic glucose production, and since there is no net change in blood glucose concentrations under steady- state clamp conditions, the GIR must be equal to the glucose disposal rate (M) (Fig. 2). Thus, whole body glucose disposal at a given level of hyperinsulinemia can be directly determined. M is typically normalized to body weight or fat-free mass to generate an estimate of insulin sensitivity.Alternatively, an insulin sensitivity index derived from clamp data can be defined as SIClamp = M/(G x ΔI), where M is normalized for G (steady-state blood glucose concentration) and ΔI (difference between fasting and steady-state plasma insulin concentrations) (12).

 

Figure 2 Schematic representation of the “steady state” dynamics of glucose and insulin during an euglycemic hyperinsulinemic glucose clamp.

 

The validity of glucose clamp measurements of insulin sensitivity depends on achieving steady-state conditions. “Steady-state” is often defined as a period greater than 30- min (at least 1 h after initiation of insulin infusion) during which the coefficient of variation for blood glucose, plasma insulin, and GIR are less than 5% (12, 13). It is possible to use stable isotope or radio-labeled glucose tracer under clamp conditions to estimate HGP so that appropriate corrections can be made to M in the event HGP is not completely suppressed (14, 15, 16,17). An alternative approach is to use an insulin infusion rate sufficiently high to completely suppress HGP according to the insulin sensitivity/resistance of the population to be studied. M is routinely obtained at only a single insulin infusion rate and therefore comparisons between M or SIClamp among different subjects is valid only if the same insulin infusion rate is used for all subjects. When glucose tracers are used during a clamp study, the tracer is infused at constant rate throughout the study. HGP estimated during the last 20 or 30 min of the clamp is a measure of insulin- mediated suppression of HGP, an estimate of hepatic insulin sensitivity. Similarly, lipolytic rates can be assessed at baseline and hyperinsulinemia during clamp by using isotopic tracers (e.g., palmitate). A single or multistep hyperinsulinemic euglycemic clamp can be used to measure adipose tissue insulin sensitivity. The linear relationship between log transformed rates of palmitate flux and plasma insulin concentrations provides an IC50 (pmol/L) for suppression of lipolysis (18).

 

ADVANTAGES AND LIMITATIONS

 

The principal advantage of the glucose clamp in humans is that it directly measures whole body glucose disposal at a given level of insulinemia under steady-state conditions. Conceptually, the approach is straightforward and there are a limited number of assumptions which are clearly defined. In research settings where assessing insulin sensitivity/resistance is of primary interest and feasibility is not an issue (e.g., study population < 100) it is appropriate to use the reference standard glucose clamp technique. The main limitations of the clamp approach are that it is time-consuming, labor intensive, expensive, and requires an experienced operator to manage technical difficulties. Thus, for epidemiological studies, large clinical investigations, or routine clinical applications (e.g., following changes in insulinresistance after therapeutic intervention in individual patients) application of the glucose clamp is not feasible. Nevertheless, when measured in relatively large cohorts, the M values showed a bimodal pattern, with an optimal cutoff of 5 mg/min/kg-FFM using a 40 mU/min·m2. However, IR was defined as a glucose disposal rate below 4.9 mg/min/kg, using a 120 mU/min·m2 dose (80, 81).

 

Insulin-Suppression Test (IST)

 

PROCEDURE

 

The insulin-suppression test, another method that directly measures metabolic insulin sensitivity/resistance, was introduced by Shen et. al. in 1970 and subsequently modified by Harano et. al. (19, 20). After an overnight fast, somatostatin (250 μg/h) or the somatostatin analogue octreotide (25 µg bolus, followed by 0.5 µg/min) (21) is intravenously infused to suppress endogenous secretion of insulin and glucagon. Simultaneously, insulin (25 mU/m2/min) and glucose (240 mg/m2/min) are infused into the same antecubital vein over 3 h. From the contralateral arm, blood samples for glucose and insulin determinations are taken every 30 min for 2.5 h and then at 10 min intervals from 150 - 180 min of the IST. The constant infusions of insulin and glucose determine steady-state plasma insulin (SSPI) andglucose (SSPG) concentrations. The steady-state period is assumed to be from 150 - 180 min after initiation of the IST. SSPI concentrations are generally (but not always) similar among subjects. Therefore, the SSPG concentration will be higher in insulin resistant subjects and lower in insulin sensitive subjects. That is, SSPG values are inversely related to insulin sensitivity. The IST provides a direct measure (SSPG) of the ability of exogenous insulin to mediate disposal of an intravenous glucose load under steady-state conditions where endogenous insulin secretion is suppressed.

 

ADVANTAGES AND LIMITATIONS

 

The SSPG is a highly reproducible direct measure of metabolic actions of insulin that is less labor-intensive and less technically demanding than the glucose clamp. Indeed, since there are no variable infusions with the IST, steady-state conditions are more easily achieved with the IST than with the glucose clamp. Estimates of insulin sensitivity determined by SSPG correlate well with reference standard glucose clamp estimates in normal subjects (r = 0.93) and in patients with type 2 diabetes mellitus (r = 0.91). (22, 23). Indeed, SSPG has positive predictive power for cardiovascular disease events and onset of type 2 diabetes (24, 25). In research settings where assessing insulin sensitivity/resistance is of primary interest and feasibility is not an issue, it is appropriate to use the IST. Moreover, the IST can be used for larger populations that may pose difficulties for application of the glucose clamp (26). Many of the limitations of the IST are similar to those described above for the glucose clamp (with the exception that the IST is less technically demanding).Thus, it is impractical to apply the IST in large epidemiological studies or in the clinical care setting. SSPG under ideal conditions determines primarily skeletal muscle insulin sensitivity and is not designed to reflect hepatic insulin sensitivity.

 

INDIRECT MEASURES OF INSULIN SENSITIVITY

 

Minimal Model Analysis of Frequently Sampled Intravenous Glucose Tolerance Test (FSIVGTT)

 

PROCEDURE

 

The minimal model, developed by Bergman, Cobelli, and colleagues in 1979, provides an indirect measure of metabolic insulin sensitivity/resistance based on glucose and insulin data obtained during an FSIVGTT (27). After an overnight fast, an intravenous bolus of glucose (0.3 g/kg body weight) is infused over 2 min starting at time 0. Currently, a modified FSIVGTT is used where exogenous insulin (4 mU/kg/min) is also infused over 5 min beginning 20 min after the intravenous glucose bolus (28, 29,30). Some studies use tolbutamide instead of insulin in the modified FSIVGTT to stimulate endogenous insulin secretion (15, 29, 31, 32, 27). Blood samples are taken for plasma glucose and insulinmeasurements at -10, -1, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 14, 16, 20, 22, 23, 24, 25, 27, 30, 40, 50, 60, 70, 80, 90, 100,120, 160, and 180 min. These data are then subjected to minimal model analysis using the computer program MINMOD to generate an index of insulin sensitivity (SI).

 

The minimal model is defined by two coupled differential equations with four model parameters (Fig. 3). The first equation describes plasma glucose dynamics in a single compartment. The second equation describes insulin dynamics in a “remote compartment”. The structure of the minimal model allows MINMOD to uniquely identify model parameters that determine a best fit to glucose disappearance during the modified FSIVGTT. SI is calculated from two of these model parameters and is defined as fractional glucose disappearance per insulin concentration unit. In addition to SI, other minimal model parameters may be used to estimate a “glucose effectiveness” index (SG). SG is defined as the ability of glucose per se to promote its own disposal and inhibit HGP in the absence of an incremental insulin effect (i.e., when insulin is at basal or constant concentrations).

 

Recently the minimal model has been used to assess free fatty acid (FFA) insulin sensitivity. Using a one compartment nonlinear model of FFA kinetics during FSIVGTT, showed that the FFA insulin sensitivity parameter correlated well with minimal model indices (33). Furthermore, this model also showed that glucose modulates disposal of FFAs.

 

Figure 3. Schematic equations, and parameters for the minimal model of glucose metabolism. Differential equations describing glucose dynamics (G(t)) in a monocompartmental “glucose space” and insulin dynamics in a “remote compartment” (X(t)) are shown at the top. Glucose leaves or enters its space at a rate proportional to the difference between plasma glucose level, G(t) and the basal fasting level, Gb. In addition, glucose also disappears from its compartment at a rate proportional to insulin levels in the “remote” compartment (X(t)). In this model, t = time; G(t) = plasma glucose at time t; I(t) = plasma insulin concentration at time t; X(t) = insulin concentration in “remote” compartment at time t; Gb = basal plasma concentration; Ib = basal plasma insulin concentration; G(0) = G0 (assuming instantaneous mixing of the IV glucose load); p1, p2, p3, and G0 = unknown parameters in the model that are uniquely identifiable from FSIVGTT; glucose effectiveness, SG = p1; and insulin sensitivity, SI = p3/p2.

 

ADVANTAGES AND LIMITATIONS

 

Minimal model analysis of the modified FSIVGTT is easier than the glucose clamp method because it is slightly less labor intensive, steady-state conditions are not required, and there are no intravenous infusions that require constant adjustment. Unlike the glucose clamp or IST, information about insulin sensitivity, glucose effectiveness, and β-cellfunction can be derived from a single dynamic test. The minimal model generates excellent predictions of glucose disappearance during the FSIVGTT. SI is a strong predictor of the development of diabetes in a prospective study of children of diabetic parents (34). Moreover, the insulin-modified FSIVGT may be used in relatively large- scale population studies (35). Therefore, in research settings where assessing insulin sensitivity along with glucose effectiveness and β-cell function is of interest, minimal model analysis of the insulin-modified FSIVGTT may be appropriate. The minimal model approach is simpler than direct methods for determining insulin sensitivity. Nevertheless, it still involves intravenous infusions with multiple blood sampling over a 3 h period that is nearly as labor intensive as the glucose clamp or IST. In addition, many limitations of minimal model analysis stem from the fact that the model oversimplifies the physiology of glucose homeostasis and is discussed in detail elsewhere (5).

 

Oral Glucose Tolerance Test (OGTT)

 

The oral glucose tolerance test (OGTT) is a simple test widely used in clinical practice to diagnose glucose intolerance and type 2 diabetes (36). After overnight fast, blood samples for determinations of glucose and insulin concentrations are taken at 0, 30, 60, and 120 min following a standard 75g oral glucose load. Oral glucose tolerance reflects the efficiency of the body to dispose of glucose after an oral glucose load or meal. The OGTT mimics the glucose and insulin dynamics of physiological conditions more closely than conditions of the glucose clamp, IST, or FSIVGTT. However, it is important to recognize that glucose tolerance and insulin sensitivity are not equivalent concepts. In addition to metabolic actions of insulin, insulin secretion, incretin effects, and other factors contribute importantly to glucose tolerance. Thus, the OGTT and meal tolerance tests provide useful information about glucose tolerance but not insulin sensitivity/resistance per se.

 

Intravenous and Oral Tracer Studies

 

The use of tracers for estimation of insulin sensitivity was first introduced in 1986 to overcome the shortcomings of FSIVGTT (37) The minimal model method does not allow segregation of glucose production from liver from exogenously administered glucose during calculations of insulin sensitivity and thus induces error in the insulin sensitivity calculations. Labeled intravenous glucose can be differentiated from endogenously produced glucose and thus use of labeled glucose during IVGTT provides more precise and accurate measurements (38,39) Similarly, labeled glucose has been used in oral glucose tolerance test and insulin sensitivity has been calculated by minimal model technique similar toFSIVGTT(40). There is a strong correlation of insulin sensitivity calculated from labeled oral minimal model with insulin sensitivity calculated from gold standard euglycemic hyperinsulinemic clamp, r=0.7, p<0.001 (41). There are dual tracer and triple tracer methods as well to estimate the hepatic/endogenous glucose production and discussion of these methods is beyond the scope of this review (42). Basal hepatic insulin resistance index can then be estimated as the product of HGP rate and the fasting plasma insulin concentration. Use of tracer definitely allows for improvement over the FSIVGTT. Use of labeled oral glucose allows for more precise measurements of insulin sensitivity and glucose disposal from a simple OGTT and this can be a useful tool in large studies. The triple tracer method is cumbersome and cannot be employed in large studies.

 

SIMPLE SURROGATE INDEXES FOR INSULIN SENSITIVITY/RESISTANCE

 

Surrogates Derived from Fasting Steady-state Conditions

 

PROCEDURE

 

After an overnight fast, a single blood sample is taken for determination of blood glucose and plasma insulin. In healthy humans, the fasting condition represents a basal steady-state where glucose is homeostatically maintained in the normal range such that insulin levels are not significantly changing and HGP is constant. That is, basal insulin secretion by pancreatic β cells determines a relatively constant level of insulinemia that will be lower or higher in accordance with insulin sensitivity/resistance such that HGP matches whole body glucose disposal under fasting conditions. Surrogate indexes based on fasting glucose and insulin concentrations reflect primarily hepatic insulin sensitivity/resistance. However, under most conditions, hepatic and skeletal muscle insulin sensitivity/resistance are proportional to each other. In the diabetic state with fasting hyperglycemia, fasting insulin levels are inappropriately low and insufficient to maintain euglycemia. Therefore, definitions of the more useful surrogate indexes take these considerations into account. Due to lack of a standardized insulin assay, it is not possible to use surrogate indexes to define universal cutoff points for insulin resistance.

 

ADVANTAGES AND LIMITATIONS

 

Simple surrogate indexes of insulin sensitivity/resistance are inexpensive quantitative tools that can be easily applied in almost every setting including epidemiological studies, large clinical trials, clinical research investigations, and clinical practice. If a direct measure of insulin sensitivity is not required, not feasible to obtain, or if insulin sensitivity is ofsecondary interest, it may be appropriate to use a surrogate index. The relative merits and limitations of individual surrogate indexes are discussed below.

 

The Homeostasis Model Assessment (HOMA)

 

HOMA, developed in 1985, is a model of interactions between glucose and insulin dynamics that is then used to predict fasting steady-state glucose and insulin concentrations for a wide range of possible combinations of insulin resistance and β-cell function (43). The model assumes a feedback loop between the liver and β-cell (43, 44, 45); glucose concentrations are regulated by insulin- dependent HGP while insulin levels depend on the pancreatic β-cell response to glucose concentrations. Thus, deficient β-cell function reflects a diminished response to glucose-stimulated insulinsecretion. Likewise, insulin resistance is reflected by diminished suppressive effect of insulin on HGP. HOMA model describes this glucose-insulin homeostasis by a set of empirically derived non-linear equations. The model predicts fasting steady- state levels of plasma glucose and insulin for any given combination of pancreatic β-cell function and insulin sensitivity. Computer simulations have been used to generate a grid from which mathematical transformations of fasting glucose and insulin β-cell function (HOMA %B) from steady-state conditions. An important caveat for HOMA is that it imputes dynamic β-cell function (i.e., glucose-stimulated insulin secretion) from fasting steady- state data. In the absence of dynamic data, it is difficult, if not impossible, to determine the true dynamic function of β-cell insulin secretion.

 

In practical terms, most studies using HOMA employ an approximation described by a simple equation to determine a surrogate index of insulin resistance. This is defined by the product of the fasting glucose and fasting insulin divided by a constant. Thus, HOMA-IR = fasting insulin (μU/ml) × fasting glucose (mmol/l) / 22.5. The constant is a normalizing factor, the product of fasting plasma insulin of 5 µU/mL and plasma glucose of 4.5 mmol/L obtained from an “ideal” and “normal” individual. Therefore, for an individual with normal insulin sensitivity, HOMA-IR = 1. It is important to note that over wide ranges of insulin sensitivity/resistance Log (HOMA-IR), (which normalizes the skewed distribution of fasting insulinvalues) determines a much stronger linear correlation with glucose clamp estimates of insulin sensitivity (12). HOMA orLog  (HOMA) is extensively used in large epidemiological studies, prospective clinical trials, and clinical research studies (45, 46, 47). In research settings where assessing insulin sensitivity/resistance is of secondary interest or feasibility issues preclude the use of direct measures by glucose clamp, it may be appropriate to use Log (HOMA-IR). However, as discussed below, other surrogate indexes have certain advantages over HOMA or Log (HOMA) in some circumstances.

 

Quantitative Insulin Sensitivity Check Index (QUICKI)

 

QUICKI is an empirically-derived mathematical transformation of fasting blood glucose and plasma insulin concentrations that provides a reliable, reproducible, and accurate index of insulin sensitivity with excellent positive predictive power (12, 48,13, 49, 50). Since fasting insulin levels have a non-normal skewed distribution, log transformation improves its linear correlation with SIclamp. However, as with 1/(fasting insulin) and the G/I ratio, this correlation is not maintained in diabetic subjects with fasting hyperglycemia and impaired β-cell function that is insufficient to maintain euglycemia. To accommodate these clinically important circumstances where fasting glucose is inappropriately high and insulin is inappropriately low, addition of log (fasting glucose) to log (fasting insulin) provides a reasonable correction such that the linear correlation with SIClamp is maintained in both diabetic and non-diabetic subjects. The reciprocal of this sum results in further transformation of the data generating an insulin sensitivity index that has a positive correlation with SIclamp. Thus, QUICKI = 1/Log (Fasting Insulin, µU/ml) + Log (Fasting Glucose, mg/dl). Over a wide range of insulin sensitivity/resistance, QUICKI has a substantially better linear correlation with SIclamp (r ≈ 0.8 – 0.9) than SI derived from the minimal model or HOMA-IR (12, 48, 49). Log (HOMA) is roughly comparable to QUICKI in this regard. Multiple independent studies find excellent linear correlations between QUICKI and glucose clampestimates (either GIR or SIClamp) in healthy subjects, obesity, diabetes, hypertension, and many other insulin- resistant states (49, 51, 52, 53, 54, 55, 56). QUICKI is among the most thoroughly evaluated and validated surrogate index for insulin sensitivity. As a simple, useful, inexpensive, and minimally invasive surrogate for glucose clamp-derived measures of insulin sensitivity, QUICKI is appropriate and effective for use in large epidemiological or clinical researchstudies, to follow changes after therapeutic interventions, and for use in studies where evaluation of insulin sensitivity is not of primary interest.

 

Adipose Tissue Insulin Resistance Index (Adipo-IR)

 

Adipo-IR is a measure similar to HOMA-IR in that it is obtained from a fasting level of FFA and insulin (product of FFA and insulin levels). Recent studies have shown that Adipo-IR correlates well with the gold standard measure of adipose tissue insulin sensitivity derived from one-step hyperinsulinemic-euglycemic clamp technique using a palmitate tracer (57). Age and physical fitness were however shown to affect the predictive values. Thus, Adipo-IR may be suitable for larger population studies, however the multistep pancreatic clamp technique is probably needed for mechanistic studies of adipose tissue insulin action.

 

Surrogates Derived from Dynamic Tests

 

PROCEDURE

 

Surrogate indexes of insulin sensitivity that use information derived from dynamic tests include OGTT, meal tolerance tests, and IVGTT. Procedures for these tests have been described in a previous section. Specific indexes including Matsuda index (58), Stumvoll index (59), Avignon index (60), oral glucose insulin sensitivity index (OGSI) (61), Gutt index (62), and Belfiore index (63) use particular sampling protocols during the OGTT or the meal. In addition, minimal model approaches have been used to model plasma glucose and insulin dynamics during an OGTT or a meal to determine insulin sensitivity/resistance (64). Glucose disposal of an oral glucose load or a meal is mediated by a complex dynamic process that includes gut absorption, glucose effectiveness, neurohormonal actions, incretin actions, insulin secretion, and metabolic actions of insulin that primarily determine the balance between peripheral glucose utilization and HGP. Surrogate indexes that depend on dynamic testing take into account both fasting steady-state and dynamic post-glucose load plasma glucose and insulin levels. After an oral glucose challenge, the HGP is maximally suppressed for approximately 60 min and remains suppressed at a constant level for the subsequent 60–120 min time period. Therefore, glucose uptake by peripheral tissues (e.g., muscle and adipose tissue) primarily determines the rate ofdecrease in plasma glucose concentration from its peak value to its nadir during an OGTT. Based on this observation, surrogate indices of hepatic and muscle insulin sensitivity/ resistance from an OGTT has been widely used (65). Recent studies comparing the OGTT- derived, tissue-specific surrogate indices hepatic insulin resistance index (HIRI) andmuscle insulin sensitivity index (MISI) with clamp measurements showed that surrogate indices derived from an OGTT are accurate in predicting insulin sensitivity but are not tissue-specific (66). Studies using oral tracers in OGTT, with measurement of insulin sensitivity from OGTT and then comparing these to clamp measurements, would be crucial to ascertain the validity these measures. Indeed, recent studies have shown that it is possible to measure hepatic insulin sensitivity in healthy volunteers and in prediabetes with the use of single tracer (67).

 

ADVANTAGES AND LIMITATIONS

 

Many surrogate measures derived from dynamic data correlate reasonably well with glucose clamp estimates of insulin sensitivity (58, 61,62). Estimates of insulin sensitivity derived from OGTT predict the development of type 2 diabetes in epidemiologic studies ( 50, 68, 65). The advantage of surrogates based on dynamic testing is that information about insulin secretion can be obtained at the same time as information about insulin action. However, if one is only interested in estimating insulin sensitivity/resistance, fasting surrogates may be preferable to dynamic surrogates because they are simpler to obtain. The oral route of glucose delivery is more physiological than intravenous glucose infusion. However, poor reproducibility of the OGTT and meal tolerance test due to variable glucose absorption, splanchnic glucose uptake, and additional incretin effects need to be considered. Thus, distinguishing direct metabolic actions of insulin following oral ingestion of glucose or a mixed meal is more problematic than after FSIVGTT. In addition, as with many other measures of insulin sensitivity, surrogates derived from dynamic testing generally incorporate both peripheral and hepatic insulin sensitivity. Although OGTT involves considerably less work than FSIVGTT, dynamic testing in general requires more effort and cost than fasting blood sampling.

 

ETHNIC DIFFERENCES

 

Hispanics, African Americans, and South Asians are highly prone to develop diabetes. A meta-analysis showed that non-diabetic Africans have lower insulin sensitivity and higher insulin response after an intravenous glucose load comparedto Caucasians and East Asians ( 69). In a study that compared euglycemic hyperinsulinemic clamp derived glucosedisposal rates (GDR) with HOMA-IR, QUICKI, and OGTT-derived indices, fasting insulin levels and HOMA-IR did not correlate with GDR whereas Matsuda index derived from OGTT significantly correlated with GDR in African American men (70). Similarly, in another study in Afro-Caribbean adults, HOMA-IR did not correlate with insulin sensitivity calculated from FSIVGTT and M-value calculated from hyperinsulinemic euglycemic clamp (71). Likewise, IR predictive ability of QUICKI and HOMA-IR was limited in Asian-Indian men (72). Recent studies highlight that minimal model may underestimate insulin sensitivity between groups when acute insulin response (AIR) is higher in one group (73). African Americans have reduced insulin clearance and higher AIR than Whites, suggesting that the minimal model may underestimate insulin sensitivity in African Americans (73). These studies suggest that at least in some ethnic groups, QUICKI and HOMA-IR may only be useful as secondary outcome measurements in assessing insulin sensitivity/resistance and studies inferring lower insulin sensitivity in non- diabetic African Americans based on FSIVGTT and minimal modeling should be interpreted cautiously.

 

METABOLOMICS

 

Metabolomics is an interrogation and quantification of small-molecule metabolites in body fluids and tissues. It aims at identifying and quantifying small molecules in the sample by either using mass spectrometry (MS) or nuclear magnetic resonance (NMR) spectroscopy. The details of the methodology and its application in diabetes research are beyond the scope of this chapter. In this chapter, we will focus on new markers of insulin resistance that have been discovered using this approach. Using a non- targeted approach, Gall et al. metabolically profiled fasting plasma samples from 399 non-diabetic, clinically healthy subjects (74). Insulin sensitivity was measured using euglycemic hyperinsulinemic clamps. Individuals in the bottom tertile of the cohort were designated as insulin- resistant. Among the 485 candidate biomarkers identified, plasma α-hydroxybutyrate levels were inversely related to insulin sensitivity and this association was independent of age, sex and BMI. Other metabolites such as linoleoyl- glycerophosphocholine (L-GPC), glycine, and creatine were also highly correlated with insulin sensitivity. Using 26 metabolites from this study, the group went on to develop a model called Quantose algorithm to predict insulin resistance. Fasting insulin, α-hydroxybutyrate, L- GPC and oleate levels were included in this model. Quantose IR as a fasting surrogate of insulin sensitivity was superior to other simple surrogate measures and was able to predict the progression from normal glucose tolerance to impaired glucose tolerance (75). Branched chain amino acids (BCAAs) were found to significantly increase in obese compared to lean subjects and a BCAA based index correlated with HOMA (76). The elevation of BCAA in subjects with impaired fasting glucose and diabetes has been confirmed in subsequent studies (77).

 

Lipoprotein insulin resistance score (LPIR) is a novel metabolomic biomarker based on nuclear magnetic resonance (NMR) quantification of lipoprotein levels and sizes. This index has been shown to predict future type 2 diabetes mellitus is some cohorts (78). LPIR is derived from the weighted score of six lipoproteins (VLDL, LDL, and HDL sizes and concentrations) that are more strongly related to IR than each of its individual subclasses (79). A risk score of between0-100 is estimated, with a score of 100 denoting being most insulin resistant. These metabolomic studies are promising since they can measure hundreds of metabolites in a very small sample. However, the pricing, technology, and access, precludes its use clinically. Further studies using this approach are necessary in larger more heterogeneous cohorts to replicate and validate surrogate insulin resistance markers derived through metabolomics

 

Table 1. Methods for Assessing Insulin Sensitivity and Resistance in Humans

Method

Measure of Insulin sensitivity

Direct Measures

Hyperinsulinemic Euglycemic Glucose Clamp

Average glucose infusion rate (GIR) = glucose disposal rate (M). SIClamp = M/(G x ΔI), where M is normalized for G (steady-state blood glucose concentration) and ΔI (difference between fastingand steady-state plasma insulin concentrations)

Insulin-suppression Test (IST)

Steady-state plasma glucose (SSPG) concentrations duringconstant infusions of insulin and glucose with suppressed endogenous insulin secretion

Indirect Measures

Minimal Model Analysis of Frequently Sampled IntravenousGlucose Tolerance Test (FSIVGTT)

Minimal model uniquely identifies model parameters thatdetermine a best fit to glucose disappearance during the modified FSIVGTT. SI : fractional glucose disappearance per insulin concentration unit; SG (glucose effectiveness): ability of glucose per se to promote its own disposal and inhibit HGP in the absenceof an incremental insulin effect (i.e., when insulin is at basal levels).

Simple Surrogate Indexes

Surrogates Derived from Fasting Steady-state Conditions

The Homeostasis Model Assessment (HOMA)

HOMA-IR = [(Fasting Insulin (µU/mL)) X (Fasting Glucose(mmol/L))]/22.5

Quantitative Insulin Sensitivity Check Index (QUICKI)

QUICKI = 1/[Log (Fasting Insulin, µU/ml) + Log (Fasting Glucose,mg/dl)]

Surrogates Derived from Dynamic Tests (OGTT)

Matsuda Index

ISI(Matsuda) = 10000/√[(Gfasting (mg/dl) x Ifasting (µU/ml) x(Gmean x Imean)]

Gutt Index - ISI (0, 120) (mg.l2.mmol-1.mIU-1.min-1)

ISI (0, 120) = 75000 + (G0-G120)(mg/l) x 0.19 x BW / 120 xGmean (0,

120min) (mmol/l) x Log [Imean (0, 120min)](mU/l)

Gmean, mean plasma glucose concentration during OGTT; Go, plasma glucose concentration during fasting; G120, plasma glucose concentration at 120 min; Gmean, mean plasma glucose concentration during OGTT; Imean, mean insulin concentration during OGTT; Io, plasma insulin concentration during fasting; I120, plasma insulin concentration at 120 min.

 

SUMMARY

 

In this chapter we have discussed a representative variety of methods currently available for estimating insulin sensitivity/resistance (but this is by no means an exhaustive review) (Table 1). These range from complex, timeconsuming, labor-intensive, invasive procedures to simple tests involving a single fasting blood sample. It is important to understand the physiological concepts informing each method so that relative merits and limitations of particular approaches are appropriately matched with proposed applications and data is interpreted correctly. The glucose clamp method is the reference standard for direct measurement of insulin sensitivity. Regarding simple surrogates, QUICKI and Log (HOMA) are among the best and most extensively validated. Dynamic tests are useful if information about both insulin secretion and insulin action are needed.

 

ACKNOWLEDGEMENTS

 

This work was supported by the Intramural Research Program, NIDDK, NIH

 

REFERENCES

 

  1. Reaven, G.M., The insulin resistance syndrome: definition and dietary approaches to treatment. Annu Rev Nutr, 2005. 25: p. 391-406.
  2. Petersen, K.F., et al., The role of skeletal muscle insulin resistance in the pathogenesis of the metabolic syndrome. Proc Natl Acad Sci U S A, 2007. 104(31): p. 12587-94.
  3. DeFronzo, R.A. and E. Ferrannini, Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care, 1991. 14(3): p. 173-94.
  4. Poirier, P., et al., Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation, 2006. 113(6): p. 898-918.
  5. Muniyappa, R., et al., Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage. Am J Physiol Endocrinol Metab, 2008. 294(1): p. E15-26.
  6. Accili, D., Lilly lecture 2003: the struggle for mastery in insulin action: from triumvirate to republic. Diabetes, 2004. 53(7): p. 1633-42.
  7. Prodi, E. and S. Obici, Minireview: the brain as a molecular target for diabetic therapy. Endocrinology, 2006. 147(6): p. 2664-9.
  8. Muniyappa, R., et al., Cardiovascular actions of insulin. Endocr Rev, 2007. 28(5): p. 463-91.
  9. Kahn, S.E., R.L. Hull, and K.M. Utzschneider, Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature, 2006. 444(7121): p. 840-6.
  10. Himsworth, H.P., Diabetes mellitus: its differentiation into insulin-sensitive and insulin-insensitive types. 1936. Int J Epidemiol, 2013. 42(6): p. 1594-8.
  11. DeFronzo, R.A., J.D. Tobin, and R. Andres, Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol, 1979. 237(3): p. E214-23.
  12. Katz, A., et al., Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab, 2000. 85(7): p. 2402-10.
  13. Chen, H., G. Sullivan, and M.J. Quon, Assessing the predictive accuracy of QUICKI as a surrogate index for insulin sensitivity using a calibration model. Diabetes, 2005. 54(7): p. 1914-25.
  14. Rizza, R.A., L.J. Mandarino, and J.E. Gerich, Dose- response characteristics for effects of insulin on production and utilization of glucose in man. Am J Physiol, 1981. 240(6): p. E630-9.
  15. Finegood, D.T., R.N. Bergman, and M. Vranic, Estimation of endogenous glucose production during hyperinsulinemic-euglycemic glucose clamps. Comparison of unlabeled and labeled exogenous glucose infusates. Diabetes, 1987. 36(8): p. 914-24.
  16. McMahon, M.M., et al., Underestimation of glucose turnover measured with 6-3H- and 6,6-2H- but not 6-14Cglucose during hyperinsulinemia in humans. Diabetes, 1989. 38(1): p. 97-107.
  17. Radziuk, J. and S. Pye, Quantitation of basal endogenous glucose production in Type II diabetes: importance of the volume of distribution. Diabetologia, 2002. 45(8): p. 1053- 84.
  18. Sondergaard, E. and M.D. Jensen, Quantification of adipose tissue insulin sensitivity. J Investig Med, 2016. 64(5): p. 989-91.
  19. Shen, S.W., G.M. Reaven, and J.W. Farquhar, Comparison of impedance to insulin-mediated glucose uptake in normal subjects and in subjects with latent diabetes. J Clin Invest, 1970. 49(12): p. 2151-60.
  20. Harano, Y., et al., Glucose, insulin, and somatostatin infusion for the determination of insulin sensitivity in vivo. Metabolism, 1978. 27(9 Suppl 1): p. 1449-52.
  21. Pei, D., et al., Evaluation of octreotide to assess insulin- mediated glucose disposal by the insulin suppression test. Diabetologia, 1994. 37(8): p. 843-5.
  22. Greenfield, M.S., et al., Assessment of insulin resistance with the insulin suppression test and the euglycemic clamp. Diabetes, 1981. 30(5): p. 387-92.
  23. Mimura, A., et al., Insulin sensitivity test using a somatostatin analogue, octreotide (Sandostatin). Horm Metab Res, 1994. 26(4): p. 184-7.
  24. Yip, J., F.S. Facchini, and G.M. Reaven, Resistance to insulin-mediated glucose disposal as a predictor of cardiovascular disease. J Clin Endocrinol Metab, 1998. 83(8): p. 2773-6.
  25. Facchini, F.S., et al., Insulin resistance as a predictor of age-related diseases. J Clin Endocrinol Metab, 2001. 86(8): p. 3574-8.
  26. Yeni-Komshian, H., et al., Relationship between several surrogate estimates of insulin resistance and quantification of insulin-mediated glucose disposal in 490 healthy nondiabetic volunteers. Diabetes Care, 2000. 23(2): p. 171-5.
  27. Bergman, R.N., et al., Equivalence of the insulin sensitivity index in man derived by the minimal model method and the euglycemic glucose clamp. J Clin Invest, 1987. 79(3): p. 790-800.
  28. Finegood, D.T., I.M. Hramiak, and J. Dupre, A modified protocol for estimation of insulin sensitivity with the minimal model of glucose kinetics in patients with insulin-dependent diabetes. J Clin Endocrinol Metab, 1990. 70(6): p. 1538-49.
  29. Saad, M.F., et al., Differences between the tolbutamide- boosted and the insulin-modified minimal model protocols. Diabetes, 1997. 46(7): p. 1167-71.
  30. Quon, M.J., et al., Direct comparison of standard and insulin modified protocols for minimal model estimation of insulin sensitivity in normal subjects. Diabetes Res, 1994. 25(4): p. 139- 49.
  31. Yang, Y.J., J.H. Youn, and R.N. Bergman, Modified protocols improve insulin sensitivity estimation using the minimal model. Am J Physiol, 1987. 253(6 Pt 1): p. E595-602.
  32. Beard, J.C., et al., The insulin sensitivity index in nondiabetic man. Correlation between clamp-derived and IVGTT- derived values. Diabetes, 1986. 35(3): p. 362-9.
  33. Stefanovski, D., et al., Insulin Action, Glucose Homeostasis and Free Fatty Acid Metabolism: Insights From a Novel Model. Front Endocrinol (Lausanne), 2021. 12: p. 625701.
  34. Martin, B.C., et al., Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet, 1992. 340(8825): p. 925-9.
  35. Howard, G., et al., Insulin sensitivity and atherosclerosis. The Insulin Resistance Atherosclerosis Study (IRAS) Investigators. Circulation, 1996. 93(10): p. 1809-17.
  36. American Diabetes, A., Diagnosis and classification of diabetes mellitus. Diabetes Care, 2007. 30 Suppl 1: p. S42-7.
  37. Cobelli, C., et al., Estimation of insulin sensitivity and glucose clearance from minimal model: new insights from labeled IVGTT. Am J Physiol, 1986. 250(5 Pt 1): p. E591-8.
  38. Toffolo, G. and C. Cobelli, The hot IVGTT two- compartment minimal model: an improved version. Am J Physiol Endocrinol Metab, 2003. 284(2): p. E317-21.
  39. Krudys, K.M., et al., Integrated model of hepatic and peripheral glucose regulation for estimation of endogenous glucose production during the hot IVGTT. Am J Physiol Endocrinol Metab, 2005. 288(5): p. E1038-46.
  40. Cobelli, C., et al., The oral minimal model method. Diabetes, 2014. 63(4): p. 1203-13.
  41. Dalla Man, C., et al., Insulin sensitivity by oral glucose minimal models: validation against clamp. Am J Physiol Endocrinol Metab, 2005. 289(6): p. E954-9.
  42. Dube, S., et al., Assessment of insulin action on carbohydrate metabolism: physiological and non-physiological methods. Diabet Med, 2013. 30(6): p. 664-70.
  43. Matthews, D.R., et al., Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia, 1985. 28(7): p. 412-9.
  44. Levy, J.C., D.R. Matthews, and M.P. Hermans, Correct homeostasis model assessment (HOMA) evaluation uses the computer program. Diabetes Care, 1998. 21(12): p. 2191-2.
  45. Wallace, T.M., J.C. Levy, and D.R. Matthews, Use and abuse of HOMA modeling. Diabetes Care, 2004. 27(6): p. 1487- 95.
  46. Haffner, S.M., H. Miettinen, and M.P. Stern, The homeostasis model in the San Antonio Heart Study. Diabetes Care, 1997. 20(7): p. 1087-92.
  47. Dabelea, D., et al., Birth weight, type 2 diabetes, and insulin resistance in Pima Indian children and young adults. Diabetes Care, 1999. 22(6): p. 944-50.
  48. Chen, H., et al., QUICKI is a useful index of insulin sensitivity in subjects with hypertension. Am J Physiol Endocrinol Metab, 2003. 284(4): p. E804-12.
  49. Mather, K.J., et al., Repeatability characteristics of simple indices of insulin resistance: implications for research applications. J Clin Endocrinol Metab, 2001. 86(11): p. 5457-64.
  50. Hanley, A.J., et al., Prediction of type 2 diabetes using simple measures of insulin resistance: combined results from the San Antonio Heart Study, the Mexico City Diabetes Study, and the Insulin Resistance Atherosclerosis Study. Diabetes, 2003. 52(2): p. 463-9.
  51. Bastard, J.P., et al., Is quantitative insulin sensitivity check index, a fair insulin sensitivity index in humans? Diabetes Metab, 2001. 27(1): p. 69-70.
  52. Yokoyama, H., et al., Quantitative insulin sensitivity check index and the reciprocal index of homeostasis model assessment are useful indexes of insulin resistance in type 2 diabetic patients with wide range of fasting plasma glucose. J Clin Endocrinol Metab, 2004. 89(3): p. 1481-4.
  53. Skrha, J., et al., Comparison of the insulin action parameters from hyperinsulinemic clamps with homeostasis model assessment and QUICKI indexes in subjects with different endocrine disorders. J Clin Endocrinol Metab, 2004. 89(1): p. 135-41.
  54. Rabasa-Lhoret, R., et al., Modified quantitative insulin sensitivity check index is better correlated to hyperinsulinemic glucose clamp than other fasting-based index of insulin sensitivity in different insulin-resistant states. J Clin Endocrinol Metab, 2003. 88(10): p. 4917-23.
  55. Katsuki, A., et al., QUICKI is useful for following improvements in insulin sensitivity after therapy in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab, 2002. 87(6): p. 2906-8.
  56. Uwaifo, G.I., et al., Indices of insulin action, disposal, and secretion derived from fasting samples and clamps in normal glucose-tolerant black and white children. Diabetes Care, 2002. 25(11): p. 2081-7.
  57. Sondergaard, E., et al., How to Measure Adipose Tissue Insulin Sensitivity. J Clin Endocrinol Metab, 2017. 102(4): p. 1193-1199.
  58. Matsuda, M. and R.A. DeFronzo, Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care, 1999. 22(9): p. 1462-70.
  59. Stumvoll, M., et al., Use of the oral glucose tolerance test to assess insulin release and insulin sensitivity. Diabetes Care, 2000. 23(3): p. 295-301.
  60. Avignon, A., et al., Assessment of insulin sensitivity from plasma insulin and glucose in the fasting or post oral glucose- load state. Int J Obes Relat Metab Disord, 1999. 23(5): p. 512-7.
  61. Mari, A., et al., A model-based method for assessing insulin sensitivity from the oral glucose tolerance test. Diabetes Care, 2001. 24(3): p. 539-48.
  62. Gutt, M., et al., Validation of the insulin sensitivity index (ISI(0,120)): comparison with other measures. Diabetes Res Clin Pract, 2000. 47(3): p. 177-84.
  63. Belfiore, F., S. Iannello, and G. Volpicelli, Insulin sensitivity indices calculated from basal and OGTT-induced insulin, glucose, and FFA levels. Mol Genet Metab, 1998. 63(2):
  64. 134-41.
  65. Cobelli, C., et al., Assessment of beta-cell function in humans, simultaneously with insulin sensitivity and hepatic extraction, from intravenous and oral glucose tests. Am J Physiol Endocrinol Metab, 2007. 293(1): p. E1-E15.
  66. Abdul-Ghani, M.A., et al., What is the best predictor of future type 2 diabetes? Diabetes Care, 2007. 30(6): p. 1544-8.
  67. Muniyappa, R., et al., Predictive Accuracy of Surrogate Indices for Hepatic and Skeletal Muscle Insulin Sensitivity. J Endocr Soc, 2019. 3(1): p. 108-118.
  68. Visentin, R., et al., Hepatic insulin sensitivity in healthy and prediabetic subjects: from a dual- to a single-tracer oral minimal model. Am J Physiol Endocrinol Metab, 2015. 309(2): p. E161-7.
  69. Hanson, R.L., et al., Evaluation of simple indices of insulin sensitivity and insulin secretion for use in epidemiologic studies. Am J Epidemiol, 2000. 151(2): p. 190-8.
  70. Kodama, K., et al., Ethnic differences in the relationship between insulin sensitivity and insulin response: a systematic review and meta-analysis. Diabetes Care, 2013. 36(6): p. 1789-96.
  71. Pisprasert, V., et al., Limitations in the use of indices using glucose and insulin levels to predict insulin sensitivity: impact of race and gender and superiority of the indices derived from oral glucose tolerance test in African Americans. Diabetes Care, 2013. 36(4): p. 845-53.
  72. Thompson, D.S., et al., Limitations of fasting indices in the measurement of insulin sensitivity in Afro-Caribbean adults. BMC Res Notes, 2014. 7: p. 98.
  73. Muniyappa, R., et al., Limited predictive ability of surrogate indices of insulin sensitivity/resistance in Asian-Indian men. Am J Physiol Endocrinol Metab, 2010. 299(6): p. E1106-12.
  74. Ha, J., et al., When MINMOD Artifactually Interprets Strong Insulin Secretion as Weak Insulin Action. Front Physiol, 2021. 12: p. 601894.
  75. Gall, W.E., et al., alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One, 2010. 5(5): p. e10883.
  76. Cobb, J., et al., A novel fasting blood test for insulin resistance and prediabetes. J Diabetes Sci Technol, 2013. 7(1):p. 100-10.
  77. Newgard, C.B., et al., A branched-chain amino acid- related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab, 2009. 9(4): p. 311-26.
  78. Menni, C., et al., Biomarkers for type 2 diabetes and impaired fasting glucose using a nontargeted metabolomics approach. Diabetes, 2013. 62(12): p. 4270-6.
  79. Mackey, R.H., et al., Lipoprotein particles and incident type 2 diabetes in the multi-ethnic study of atherosclerosis. Diabetes Care, 2015. 38(4): p. 628-36.
  80. Shalaurova, I., et al., Lipoprotein insulin resistance index: a lipoprotein particle-derived measure of insulin resistance. Metab Syndr Relat Disord, 2014. 12(8): p. 422-9.
  81. Stern, S.E., et al., Identification of individuals with insulin resistance using routine clinical measurements. Diabetes, 2005. 54(2): p. 333-339.
  82. Tam, C.S., et al., Defining insulin resistance from hyperinsulinemic-euglycemic clamps. Diabetes Care, 2012. 35(7): p.1605-1610.

 

Physiology of GnRH and Gonadotropin Secretion

ABSTRACT

 

Gonadotropin hormone-releasing hormone (GnRH) is the key regulator of the reproductive axis.  Its pulsatile secretion determines the pattern of secretion of the gonadotropins, follicle stimulating hormone and luteinizing hormone, which then regulate both the endocrine function and gamete maturation in the gonads. Recent years have seen rapid developments in how GnRH secretion is regulated, with the discovery of the kisspeptin-neurokinin-dynorphin neuronal network in the hypothalamus. This mediates both positive and negative sex steroid feedback control of GnRH secretion, in conjunction with other neuropeptides and neurotransmitters. This chapter describes the main features of this regulatory system, including how its anatomical arrangements interact with functional control, and describes key differences between rodent and larger mammalian systems.

 

INTRODUCTION

 

Since the discovery of Gonadotropin Releasing Hormone (GnRH), an extensive body of literature has established it as the pivotal central regulator of human reproduction. However, the GnRH neuronal network, per se lacks the cellular machinery to fully integrate developmental, environmental, endocrine, and metabolic factors that influence its secretion. For example, GnRH neurons do not express the principal estrogen receptor alpha (ER-alpha), which is required for sex-steroid mediated control of gonadotropin secretion (1). Intermediate signaling pathways must therefore exist to mediate gonadal steroid feedback. Current evidence, accumulated since the discovery of Kisspeptin-Neurokinin B-Dynorphin (KNDy) neuronal network in the last decade, suggests a pivotal role for this network in the regulation of pulsatile GnRH secretion by integrating nutrient, endocrine, and environmental signals, and thus the control of downstream hypothalamic-pituitary-gonadal (HPG) axis.

 

The HPG axis anatomically comprises:

 

  1. The hypothalamus (especially the infundibular nucleus, the human homologue of the arcuate nucleus) where the KNDy and GnRH-producing neurons are located.
  2. The anterior pituitary, where Luteinizing Hormone (LH) and Follicle-Stimulating Hormone (FSH) are secreted by gonadotropes.
  3. The gonads, responsible for the production of both sex steroids and gametes, under the influences of LH and FSH.

 

As with other endocrine systems, positive and negative feedback regulate HPG axis (2,3). In this chapter, we have focused on human data. Where human data is limited, data from other species are leveraged.

 

GONADOTROPIN RELEASING HORMONE (GnRH) – THE PRINCIPAL REGULATOR OF REPRODUCTION

 

The Discovery of GnRH

 

GnRH was isolated from porcine hypothalami and structurally identified as a decapeptide (pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly·NH2) five decades ago (4-6). This decapeptide was shown to potently stimulate LH and FSH release from the pituitary in a number of mammalian species (6,7). Early literature referred to this peptide as the ‘Luteinizing Hormone-Releasing Hormone (LH-RH)’, but more recently, it is widely referenced as Gonadotropin Releasing Hormone (GnRH) -reflecting the stimulatory role on the secretion of both gonadotropins – i.e., LH and FSH (8).

 

Diverse forms of GnRH and its receptor exist among vertebrates, with over twenty primary structures across species, suggesting that GnRH system developed early in the evolutionary sequence (9,10). The GnRH structure was first identified in mammals and is therefore referred to as GnRH I (9,11).  Subsequently, another structurally different vertebrate GnRH sequence was first identified from chicken brain -this is now referred to as GnRH II (pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH2) (10,12). A third form has also been described in fish - GnRH III (9,12). In mammals, hypophysiotropic functions are limited to GnRH I, therefore in the human context GnRH I is referred to as GnRH (13) and we will use this terminology for this review.

 

Neuroanatomy of GnRH Neurons

 

GnRH neurons originate in the medial olfactory placode during embryological development and migrate along the olfactory bulb to their final positions within the hypothalamus (14,15). A number of factors contributing to this GnRH neuron migratory process have been identified: anosmin-1 (the product of KAL gene) (16), neuropilins (17), leukemia inhibitory factor (18), fibroblast growth factor receptor 1 (19), fibroblast growth factor receptor 8 (20), polysialic form of neural adhesion molecule (PSA-NCAM) (21), among others (22). Defective GnRH migration leads to Kallmann syndrome, characterized by hypogonadotropic hypogonadism due to GnRH deficiency and anosmia (15). Mutations in prokineticin genes (PROK1 and PROK2) lead to hypogonadotropic hypogonadism without anosmia, suggesting that factors other than suboptimal migration can also lead to functional deficiencies in GnRH (15,23,24).

 

GnRH cell bodies are located in the medial preoptic area (POA) and in the arcuate/infundibular nucleus of the hypothalamus, forming a neuronal network with projections to the median eminence (25). GnRH is secreted from the median eminence into the fenestrated capillaries of portal circulation, carried to the anterior pituitary (25). In humans, the number of GnRH neurons has been estimated to range between 1000 to 1500 (9,14). The co-location of GnRH neurons with other central regulators allows the GnRH network to be influenced by a range of neuroendocrine and metabolic inputs (26).

 

GnRH Secretion and Pulsatility

 

Two distinct modes of GnRH secretion have been described: pulsatile and surge modes (26). Pulsatile mode refers to episodic release of GnRH, with distinct pulses of GnRH secretion into the portal circulation with undetectable GnRH concentrations between pulses. The surge mode of GnRH secretion occurs in females, during the pre-ovulatory phase, in which the presence of GnRH in the portal circulation appears to be persistent (26,27).

 

Direct pulsatile GnRH release was initially demonstrated in ovariectomized rhesus monkeys using serial samples of portal blood (28). Pulsatile pattern of GnRH secretion was demonstrated subsequently in humans through serial blood sampling during pituitary surgery (29). Abolishment of LH pulses by GnRH antisera (30,31) and its reestablishment with GnRH analogues (30) suggest that LH pulses are determined by the underlying GnRH pulsatility.  The LH pulsatility was first detected during an attempt to validate a radioimmunoassay to measure serum LH in rhesus monkeys, where marked variations in LH levels was noticed (32). Further studies confirmed the pulsatile nature of LH secretion (33-35). In women, the frequency and amplitude of LH pulses were noted to be dependent on the menstrual cycle phase, with pulses every 1 to 2 hours during the early follicular phase eventually merging into a continuous mid-cycle surge, and decreased pulse frequency to every 4 hours during the luteal phase (36). In humans, LH pulse frequency is used as a surrogate of GnRH pulsatility, as ethical considerations and technical challenges preclude sampling of hypophyseal blood or cerebrospinal fluid to measure GnRH concentrations directly (37,38).

 

The importance of GnRH pulsatility on LH and FSH secretion was first demonstrated in rhesus monkeys, where endogenous GnRH secretion was abolished by hypothalamic radiofrequency. Pulsatile GnRH reinstated gonadotropin secretion in these animals, whereas continuous GnRH only elicited a transient response. Moreover, the switch from continuous to pulsatile GnRH administration allowed recovery of gonadotropin secretion (39).

 

GnRH neurons coordinate their activity, but the precise mechanism of this remains unclear (27,40), and is the subject of continuing investigations. Episodic multi-unit electrical activity at medial basal hypothalami (MBH) is correlated with LH release, suggesting that ‘GnRH pulse generator’ is anatomically located at MBH – or closely linked to it neurohormonally(41,42). GnRH neurons show intrinsic electrical pulsatility. GnRH cell lines derived from mouse hypothalamic and fetal olfactory placode GnRH neurons both demonstrate intrinsic pulsatility in vitro (26,43,44). Functionally, the ‘GnRH pulse generator’ relies on complex relations between glutamatergic cells, GnRH and other neurons, and likely other elements are involved, of which the kisspeptin-neurokinin B-opioid pathway may have a pivotal intermediary role in the regulation of GnRH pulsatility (45).

 

Differential Regulation of LH and FSH

 

The stimulatory effects of GnRH on LH and FSH secretion are not identical (46).  FSH secretion is more irregular than LH in both humans and sheep, which is essentially related to the pulsatility and different stimulatory effects of GnRH, but other factors also might be relevant, such as differences in LH and FSH storage (more scarce for the FSH), existence of different gonadotropes subpopulations, or diverse response times to GnRH (47). In ovariectomized sheep administered GnRH antisera, pulsatile secretion of LH was completely inhibited (undetectable LH levels within 24 hours), while the FSH concentration fell more slowly and remained detectable (30). It has been estimated that 93% of the GnRH pulses were associated with FSH pulses and, unlike LH, a constitutive secretion of FSH appears to exist (48). The frequency of GnRH input has been demonstrated to selectively regulate gonadotropin subunit gene transcription: rapid GnRH pulse rates increase α and LH-β and slow GnRH pulse frequency increases FSH-β gene transcription (49-51). Moreover, with progressive increases in GnRH frequency (from one pulse every 120 to 60 min, from 60 to 30 min, and from 30 to 15 min) in GnRH deficient men, mean LH rose concurrently with a decrease in LH pulse amplitude, while FSH remained unchanged (52).

 

Biological and Clinical Relevance of GnRH Pulsatility

 

Appropriate modulation of LH pulse frequency is essential for pubertal maturation and reproductive function. In infancy, LH pulsatile secretion is increased (often termed mini-puberty), likely reflecting pulsatile GnRH secretion, but soon becomes quiescent (53). This pre-pubertal suppression of HPG axis has been shown to occur in agonadal humans (54)and primates (55), suggesting that hypothalamo-hypophyseal factors play a role in post-natal quiescence of the reproductive axis, until puberty sets in.

 

The onset of pubertal maturation is heralded by the development of a pattern of steady acceleration in LH pulsatility (56). In children, higher basal and GnRH stimulated LH concentrations are observed in early childhood (<5 years). This is subdued mid-childhood (5-11 years) and increase thereafter with pubertal development (54,57).  Conceptually, an abnormal reactivation of GnRH pulse frequency is the central mechanism associated with precocious or delayed puberty (14).

 

In women, the pattern of GnRH secretion is essential for the regulation of the menstrual cycle (Figure 1) (58,59). LH pulse frequency is slow in the luteal phase, and increasingly speeds up during the follicular and the pre-ovulatory phases, presumably reflecting changes in GnRH pulse frequency (60). Abnormalities in GnRH - and hence LH pulse frequency - are associated with a number of reproductive endocrine disorders. In hypothalamic amenorrhea, a condition associated with anovulatory amenorrhea and hypoestrogenism, LH pulse frequency (and by inference GnRH) is lower than expected for the prevailing steroid profile and is comparable to luteal phase pulsatility (37). LH pulse frequency in hyperprolactinemic women is also lower than in healthy women, requiring dopaminergic agonist preparations, such as bromocriptine to regulate prolactin secretion and restore LH pulse frequency (38). In polycystic ovary syndrome LH pulse frequency and amplitude are higher throughout the menstrual cycle in comparison to that observed in healthy women, contributing to chronic anovulation (61-64).

 

Figure 1. Hormonal oscillations through the menstrual cycle. In the early follicular phase of the menstrual cycle, the initial increase in FSH stimulates follicular recruitment and maturation. The consequent secretion of estradiol (E2) selectively inhibits FSH release (needed for selection of the dominant follicle) and maintains rapid GnRH pulsatility during the late follicular phase. The persistent rapid GnRH pulses increase LH, which further stimulates E2 secretion, culminating in positive E2 feedback to produce the mid-cycle LH surge. During the LH surge, GnRH levels appear to be consistently elevated and remain elevated as LH declines, suggesting that the frequency of GnRH pulse has become very rapid or continuous, which results in desensitization of LH secretion (possibly the mechanism to terminate the LH surge). After ovulation, luteinization of the ruptured follicle results in progesterone secretion which reduces the frequency of GnRH pulses. With the demise of corpus luteum, E2, progesterone and inhibin levels fall, and the GnRH pulse frequency increases, leading to follicular maturation in the next cycle. (Adapted from: Marshall JC, Dalkin AC, Haisenleder DJ, Paul SJ, Ortolano GA, Kelch RP. Gonadotropin-releasing hormone pulses: regulators of gonadotropin synthesis and ovulatory cycles. Recent Prog Horm Res. 1991;47:155-187).

 

NEURONAL REGULATION OF GnRH SECRETION: THE KISSPEPTIN-NEUROKININ B-DYNORPHIN (KNDy) NEURONAL NETWORK

 

Whilst the central role attributed to GnRH remains undisputed, its effective function requires input from other neuronal networks. For instance, the absence of estrogen receptor alpha (ER-alpha) expression on GnRH neurons suggests the need for an intermediate signaling pathway to mediate gonadal steroid feedback (1). The discovery of kisspeptin signaling in neuroendocrine regulation of human reproduction revolutionized the current understanding of the HPG axis. Kisspeptin signaling pathway is increasingly recognized as essential for normal puberty, gonadotropin secretion, and regulation of reproduction (65-67). Other relevant kisspeptin roles have been identified such as regulation of sexual and social behavior, emotional brain processing, mood, audition, olfaction, metabolism, body composition, cardiac function, among others (68-74).

 

Discovery of KNDy Neuronal Network

 

KiSS1, the gene encoding kisspeptins, was first described in 1996 as a suppressor of metastasis in human malignant melanoma (75,76). This gene was discovered in Hershey and named in accordance with the famous chocolates ‘Hershey’s Kisses’; the inclusion of ‘SS’ is indicative of ‘suppressor sequence’. The KiSS1 gene maps to chromosome 1q32 and includes four exons of which the first two are not translated (77). The gene encodes the precursor 145 amino acid peptide, which is cleaved down to a 54 amino acid peptide. This peptide can be truncated to 14, 13 and 10 amino-acid peptides, all sharing the C-terminal sequence (78,79). These peptides are collectively referred to as kisspeptins - and Kp-10, Kp-13, Kp-14 and Kp-54 are suggested abbreviations for human kisspeptins (80). In 2001, kisspeptins were identified as ligands for the orphan G–protein receptor 54 (GPR54) (81-83), currently named KISS1R (80). KISS1R is localized to human chromosome 19p13.3 and it has five exons, encoding a 398-amino acid protein with seven trans-membrane domains (79,82). Upon binding by kisspeptin, KISS1R activates phospholipase C and recruits intracellular messengers, inositol triphosphate and diacylglycerol, which in turn lead to the release of calcium and activation of protein kinase C (82-84).

 

A reproductive role for kisspeptin in humans became apparent from patients with pubertal disorders which were associated with KISS1R mutations (85-87). A number of inactivating mutations of Kiss1 and Kiss1r have since been reported in animal models with phenotypes characterized by pubertal delay (88). An activating mutation in KISS1R has been described in a girl with precocious puberty: when compared to cells with wild-type transfected GPR54, cells with this mutation showed prolonged inositol phosphate accumulation and phosphorylation of extracellular signal–regulated kinase, suggesting extended activation of intracellular signaling by the mutant GPR54 (89). Missense mutations have also been reported in KISS1 gene in three unrelated children with central precocious puberty (90). Functional studies of these mutant peptides demonstrated higher resistance to in vitro degradation but normal affinity to KISS1R, thus suggestive of increased bioavailability as the mechanism by which these abnormal kisspeptins induce precocious puberty (90).

 

Recently in an Asian cohort, potentially regulatory polymorphisms, as rs5780218 and rs12998, in KiSS1 gene were significantly associated to genetic susceptibility to central precocious puberty in Chinese girls by single-locus analysis (91). Nevertheless, these findings are inconsistently reported in literature and require additional validation in functional studies.

 

A role for neurokinin B in the hypothalamic regulation was also demonstrated when genetic studies in patients from consanguineous families with hypogonadotropic hypogonadism were found to have missense mutations in TAC3 (encodes neurokinin B) and TACR3 (encodes neurokinin B receptor) (92). Other cases have been reported since (93-96).

 

There is also long-standing evidence for the role of opioid systems in reproduction. In 1980, Wilkes reported the localization of β endorphin in the human hypothalamus (97). Studies involving the administration of naloxone and naltrexone (opioid antagonists) to humans showed stimulatory effects on LH secretion (98,99), and other studies supported the notion that endogenous opioids play a role in the control of HPG axis (100-104). In 2007, it was demonstrated that dynorphin and kisspeptin are co-localized along with neurokinin B in the same hypothalamic neuronal population in sheep, therefore termed KNDy (Kisspeptin-Neurokinin B-Dynorphin) neurons, highlighting the possible interconnection between these neuropeptides in the control of GnRH and gonadotropin secretion (105-107). The co-localization of kisspeptin, neurokinin B and dynorphin has also been demonstrated in humans (108).

 

Kisspeptin neurons have also other important neuroanatomical relationships, such as with neuronal nitric oxide synthase neurons as demonstrated in prepubertal female sheep (109), or with somatostatin neurons in the rat hypothalamus (110).

 

Neuroanatomy of KNDy Neuronal Network

 

The location of kisspeptin neurons is different between rodents and human species. In humans, kisspeptin neurons are distributed in the rostral Pre-optic Area (POA) and in the infundibular nucleus in the hypothalamus (Figure 2) (108,111). In both male and female autopsy samples, the majority of kisspeptin cell bodies are identified in the infundibular nucleus, and a second dense population of kisspeptin neurons in the rostral POA (108). The infundibular nucleus (arcuate nucleus in non-human species) is similar across species, but the rostral region is more species specific (108,112,113). In rodents, the rostral population is located in the anteroventral periventricular nucleus (AVPV) and the periventricular nucleus (PeN), the continuum of this region named as the rostral periventricular region of the third ventricle (RP3V) (112,114). Humans and ruminants lack this well-defined RP3V population of kisspeptin neurons, which are more scattered within the preoptic region (113,115).

 

Kisspeptin axons form dense plexuses in the human infundibular stalk, where the secretion of GnRH occurs (108). Axo-somatic, axo-dendritic, and axo-axonal contacts between kisspeptin and GnRH axons were demonstrated at this level, showing that kisspeptin and GnRH networks are in close proximity (108,116). Moreover, GnRH neurons express Kiss1rmRNA, reinforcing the notion of kisspeptin involvement in GnRH secretion (117-119).

 

Figure 2. Neuroanatomy of kisspeptin-GnRH pathway and the control of HPG axis in humans and rodents. Kisspeptin signals directly to GnRH neurons, which express KISS1R. The location of kisspeptin neurons within the hypothalamus is species specific, residing within the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus in rodents, and within the preoptic area (POA) and the infundibular nucleus in humans. Kisspeptin neurons in the infundibular nucleus (humans)/arcuate nucleus (rodents) co-express neurokinin B and dynorphin (KNDy neurons), which autosynaptically regulate kisspeptin secretion (via neurokinin B receptor and kappa opioid peptide receptor). In humans, infundibular KNDy neurons relay negative (red) and positive (green) feedback, whereas in rodents the negative and positive steroid feedback are mediated via arcuate nucleus and AVPV respectively. The role of human POA kisspeptin neurons in sex steroid feedback is not yet clear. (Adapted from: Skorupskaite K, George JT, Anderson RA. The kisspeptin-GnRH pathway in human reproductive health and disease. Human Reproduction Update. 2014;20:485-500).

 

Three-quarters of kisspeptin-immunoreactive cells in the human infundibular nucleus of the hypothalamus co-express neurokinin B and dynorphin (KNDy neurons) (108,120). KNDy neurons in rodents and ruminants are localized in the arcuate nucleus of the hypothalamus. However, neurokinin B and dynorphin are absent from kisspeptin neurons in the hypothalamic POA (Figure 2) (67,115). This differential expression of neuropeptides may reflect distinct functions of these two kisspeptin populations with kisspeptin neurons in the AVPV acting as LH surge generators, while those in the ARC (including KNDy neurons) acting as LH pulse generators.

 

Significant kisspeptin expression was also demonstrated in central extra-hypothalamic sites, including in limbic and paralimbic brain regions, such as medial amygdala, cingulate, globus pallidus, hippocampus, putamen and thalamus, key areas of neurobiological control of sexual and emotional behaviors (reviewed in detail in (121)), as well as peripherally in organs like ovary, testis, uterus and placenta where the kisspeptin system may also play a part in reproduction function (122,123).

 

Apart from reproductive and central kisspeptin expression, the kisspeptin signaling system has been demonstrated in several peripheral tissues, namely, in pancreas (involved in glucose-stimulated insulin secretion); in endothelial cells of different vascular beds as coronary artery, aorta and umbilical vein (triggering vasoconstriction); in the kidney, namely, in tubular cells, collecting duct cells and vascular smooth muscle cells (involved in function and renal morphogenesis); as well as in bone, fat and liver tissue (78,124,125).

 

Interactions Between Kisspeptin, Neurokinin B and Dynorphin

 

KDNy neurons act synergistically to induce coordinated and pulsatile GnRH secretion by regulating the neuroactivity of other KDNy cells. This is supported by the existence of neurokinin B and kappa opioid peptide receptors (receptor for dynorphin) within the KNDy cells, but not kisspeptin receptors, which are predominantly expressed on GnRH neurons (107,120,126). Neuron-neuron and neuron-glia communications via gap junctions contribute for the synchronized activities among KNDy neurons (127).  

 

Neurokinins (A and B) are members of the tachykinin family of peptides, which stimulate three related GPCRs (encoded by TACR1, TACR2 and TACR3) (128) This family also contains substance P, neuropeptide K, neuropeptide γ, hemokinin-1, and more recently endokinins. Neurokinin B acts predominantly on TACR3. Neurokinin B increases the membrane potential of KNDy neurons, leading to an increase in KNDy neuron pulsatile activity which, in turn, will promote the secretion of kisspeptin leading, ultimately, to GnRH secretion (67,129,130). Neurokinin B signaling regulates GnRH/LH secretion in healthy women, and it is crucial for the mediation of the estrogenic positive and negative feedback on LH secretion (131-133). There is rapidly increasing interest in the therapeutic value of neurokinin antagonists in several indications in reproductive health, recently reviewed in (134).

 

In women with polycystic ovary syndrome, the relationship between kisspeptin and gonadotropin levels has been widely explored in those with anovulatory cycles (135-138). Most studies have shown higher serum levels of kisspeptin and LH when the oligomenorrhea phenotype is present, despite the high heterogeneity observed. In this context, potential treatments targeting neuroendocrine dysfunction emerged. The administration of neurokinin 3 receptor antagonists markedly reduced serum LH concentration and pulse frequency, as well as serum testosterone (139-141). A recent study confirmed a complex crosstalk between neurokinin B and kisspeptin pathways in the regulation of GnRH secretion in polycystic ovary syndrome. In this study, kisspeptin-10 infusion given to women with polycystic ovary syndrome increased LH secretion with a direct relationship to estradiol exposure. Neurokinin 3 receptor antagonism reduced LH secretion and pulsatility, and whilst LH response to kisspeptin-10 was preserved, its relationship with circulating estradiol was not. More interestingly, although kisspeptin-10 increased LH pulse frequency, changes in other parameters of LH secretory pattern were prevented when co-administered with neurokinin 3 receptor antagonists (141).

 

In postmenopausal women,  seven day treatment with neurokinin 3 receptor antagonist decreased LH secretion, but not FSH secretion, as well as lead to a remarkable reduction in hot flushes (142). Neurokinin 3 receptor antagonism efficiency in treating menopausal hot flushes has been also demonstrated in other clinical trials (143,144). Fezolinetant administrated in a single daily dose regimen (30 or 45mg/daily) for treatment of moderate to severe vasomotor symptoms reduced these symptoms by over 50% from baseline within the first week and persistently during the 52-week treatment period, and is now approved for the treatment of menopausal vasomotor symptoms (145-148).

 

A second comparable drug, elinzanetant, which differs in its pharmacology in that it is an antagonist at the NK1 as well as NK3 receptor, has also been recently demonstrated to reduce the severity and frequency of moderate-to-severe vasomotor symptoms and also to improve sleep quality and menopause-related quality of life (149). The importance of NK1 receptor antagonism in these effects is unclear.

 

In healthy men, neurokinin B signaling display a central role for the reproductive function, and this is functionally upstream of kisspeptin-mediated GnRH secretion: LH, FSH and testosterone secretion decreased during the administration of a neurokinin 3 receptor antagonist, while kisspeptin-10 administration restored LH secretion to the same degree before and during neurokinin 3 receptor antagonist treatment (150).

 

An increase in the expression of Kiss1 in the hypothalamic neurons was observed following senktide (agonist of neurokinin B) administration (151), and its stimulatory effects were abolished in Gpr54 knock-out male (152). In ovariectomized goats, neurokinin B stimulated LH secretion through electrical multi-unit activity corresponded to LH secretion, suggesting a hypothalamic site for this GnRH pulse generation (153). GnRH antagonists abolished the stimulatory effect of neurokinin B, demonstrating its site of action to be functionally higher than the GnRH receptor (154,155).

 

Dynorphins act as the decelerator that inhibits KNDy neurons pulsatility. Studies involving the administration of opioid antagonists to humans have shown stimulatory effects on LH secretion in late follicular and mid-luteal phase (98,99), and together with other studies (100-104), highlight the inhibitory input by dynorphins on kisspeptin signalling, and consequently on GnRH/gonadotropin secretion. Through the stimulatory effects of neurokinin B and kisspeptin, and the inhibitory action of dynorphin, these neuropeptides coordinate pulsatile GnRH and LH secretion (Figure 2) (156,157).

 

Kisspeptin-mediated GnRH secretion is sex steroid dependent. Estrogen and progesterone directly modulate kisspeptin activity though the sex-steroid receptors expressed on kisspeptin neurons at both AVPV and the arcuate nucleus (158-160). Furthermore, two distinct populations of kisspeptin neurons, the infundibular/arcuate region of which interacts with neurokinin B and dynorphin, appear to mediate distinct sex-steroid pathways (discussed in more detail in sections 4.1-4.4).

 

Briefly, in humans, KNDy neurons in the infundibular nucleus alone are involved in negative and positive sex-steroid feedback, whereas in rodents positive sex-steroid feedback seems to be mediated via kisspeptin neurons in the AVPV region and negative sex-steroid feedback via the arcuate KNDy neurons (Figure 2) (67,111,160,161).

 

Stimulatory Effect of Kisspeptin on GnRH and Gonadotropin Secretion

 

Kisspeptin is a potent stimulator of the HPG axis – and in fact, it is the most potent GnRH secretagogue currently known. Kisspeptin signals directly to the hypothalamic GnRH neurons via kisspeptin receptor to release GnRH into the portal circulation, which in turn stimulates the anterior pituitary gonadotropes to produce LH and FSH (129,162).

 

The stimulatory effects of kisspeptin on LH secretion have been documented in animal models (163-166). This is consistent with human studies, where kisspeptin increases both LH and FSH secretion with the preferential stimulatory effect on the former (67,167-177). Kissppetin-54 was first administered in healthy men as an intravenous infusion with dose-dependent rise in LH secretion (169). Since then kisspeptin was administered in different isoforms (kisspeptin-54 and kisspeptin-10), different routes (subcutaneous and intravenous), different types of exposure (continuous and bolus), to healthy men and women and in endocrine disease models with low gonadotropin output, all showing stimulatory effect of kisspeptin on LH secretion (fully reviewed in (67)).

 

Pulsatile GnRH secretion correlates with LH pulsatility, prompting investigation of the effect of kisspeptin on regulating LH pulse frequency. LH pulse frequency and amplitude were increased following intravenous infusion of kisspeptin-10 in healthy men (172), and subcutaneous bolus of kisspeptin-54 in healthy women (174). The hypothalamic response to kisspeptin-54 and the pituitary response to GnRH are preserved in healthy older men (178). Kisspeptin also stimulates LH pulse frequency in reproductive endocrine disorders of low LH pulsatility, including hypothalamic amenorrhea, defects in the neurokinin B pathway and hypogonadal men with diabetes (96,179,180). Indeed, kisspeptin-54 and kisspeptin-10, as well as kisspeptin agonists like MVT-602 (previously known as TAK-448) are able to stimulate physiological reproductive hormone secretion in individuals with functional hypogonadism related to deficient GnRH secretion, such as in hypothalamic amenorrhea or polycystic ovary syndrome (181,182).

 

In addition, recent findings have explored further the effects of MVT-602. LH concentration increased in a dose-dependent manner, resembling the amplitude and duration found in the physiological mid-cycle LH surge, proving to be safe and well tolerated throughout the dose range (0.3 – 3.0 mg) (183). This approach to mimicking the physiological response during oocyte maturation and ovulation may have clinical utility for women during medically assisted reproduction.

 

Kisspeptin regulates GnRH and subsequently gonadotropin secretion through Kiss1r, as suggested by Messager who demonstrated no detectable LH levels in response to kisspeptin in Kiss1r knockout mice (119). The prevention of the stimulatory effect of kisspeptin on LH secretion by GnRH antagonists indicate that kisspeptin action is GnRH-mediated (118,164,184-186). This is further supported by the observation that kisspeptin cause depolarization of GnRH neurons (117) and stimulate GnRH release from hypothalamic explants (187,188). The expression of GnRH mRNA is upregulated in GnRH neurons following kisspeptin administration (189). Moreover, in patients with impaired functional capacity of GnRH neurons (idiopathic hypogonadotropic hypogonadism), the same dose of kisspeptin failed to induce LH response seen in healthy men and women (190). In female rats, ablation of KNDy neurons resulted in hypogonadotropic hypogonadism, confirming its role in the maintenance of normal LH levels and to estrous cyclicity (191).

 

Some investigators have demonstrated a direct stimulatory effect of kisspeptin on gonadotropes, but this direct stimulatory action of kisspeptin on gonadotropes remains debatable (192-196). Kiss1 and Kiss1r gene expression has been shown in gonadotropes, and gonadotropin secretion from the pituitary explants was observed following exposure to kisspeptin (78,192-195). Moreover, LHβ and FSHβ gene expression was upregulated in the primary pituitary cells treated with kisspeptin. Whilst kisspeptin can directly regulate gonadotropins at the transcriptional level, it appears to be less relevant than the GnRH-mediated action (67,195,196).

 

Desensitization Effect of Chronic or Continuous Exposure to Kisspeptin

 

Continuous administration of GnRH desensitizes the HPG axis by downregulation of GnRH receptors and desensitization of gonadotropes, following an initial stimulatory effect (39). It is therefore important to ascertain the effects of continuous exposure to kisspeptin on the HPG axis. Efforts have been made to assess the impact of continuous infusions of kisspeptin in a number of animal experiments (119,197-200).

 

In adult rats, continuous administration of kisspeptin-54 increased serum LH and free testosterone on day one, but this stimulatory effect was lost after 2 days, indicative of kisspeptin receptor desensitization (200). In rhesus monkeys, the continuous administration of kisspeptin-10 resulted in suppression of LH secretion, indicating desensitization of kisspeptin receptor (198). The kisspeptin receptor has been shown to desensitize in vitro (197). In sheep, infusion of kisspeptin-10 resulted in acute increase in serum LH levels, which declined by the end of 4-hour infusion, while GnRH remained elevated following the discontinuation of kisspeptin-10 administration. This suggests that desensitization to GnRH could be occurring at the level of pituitary gonadotropes (119).  

 

Consistent with animal studies, Jayasena et al. demonstrated that in women with hypothalamic amenorrhea an initial increase in LH and FSH secretion was not sustained following twice daily subcutaneous kisspeptin-54 administration for two weeks (176). Other studies in humans employing continuous or repeated kisspeptin administration provide conflicting evidence for kisspeptin-mediated desensitization and appear to be dose-related (172,180). High doses of kisspeptin may induce desensitization, but this is not apparent at lower doses (67). Sustained LH secretion and increased LH pulsatility was demonstrated with lower dose of kisspeptin-54 (0.01-1nmol/kg/h) infusion for 8 hours in women with hypothalamic amenorrhea (180) and kisspeptin-10 (3.1 nmol/kg/h) infusion for 22.5 hours in healthy men (172). In contrast, LH secretion was not maintained in three healthy men during the 24 hour infusion of kisspeptin-10 at 9.2 nmol/kg/h (the highest dose used in humans so far), although serum LH did not fall to the castrate levels and remained well above baseline at end of infusion (201).

 

Kisspeptin receptor agonist analogues, TAK-488 and TAK-683, induce desensitization when administered to healthy men (202,203). However, the ability of natural kisspeptin fragments to downregulate the HPG axis in humans remains to be established, and is to date complicated by differences in study protocols, in terms of isoform of kisspeptin used, duration (8 hours-2 weeks), mode and route of kisspeptin administration, lower doses of kisspeptin in human studies compared to animal, and the endocrine profile of the study participants (men versus women versus hypothalamic amenorrhea). 

 

Sexual Dimorphism in Kisspeptin Signaling

 

The response to kisspeptin is different in men and women. In men, kisspeptin potently stimulates the release of LH, but in women the effect of kisspeptin is variable and dependent on the phase of menstrual cycle (67). Whilst men respond to the modest doses of kisspeptin, LH response to kisspeptin in healthy women is minimal and inconsistent in the early follicular phase but greatest in the pre-ovulatory phase of the menstrual cycle (169-171,177). This indicates that in addition to the fluctuations in sex-steroid milieu, other mechanisms, such as changes in pituitary sensitivity to GnRH or GnRH network responsiveness to kisspeptin regulate the sensitivity to kisspeptin throughout the menstrual cycle (67,126,204).

 

Not only there is sexual dimorphism in gonadotropin response to kisspeptin, but there are also anatomical differences. Female hypothalami have significantly more kisspeptin fibers and kisspeptin cell bodies than men (173). Only a few kisspeptin cell bodies are present in the male infundibular nucleus and none in the rostral periventricular nucleus, which is on contrary to the female hypothalami with abundant kisspeptin network in both of these hypothalamic nuclei (108). These sex differences in kisspeptin neurons appear to be established early during perinatal development through the action of sex steroids (126,205).

 

These marked functional and anatomical differences may reflect sexually dimorphic roles of kisspeptin between both sexes, influencing their reproductive functions, namely the sex steroid feedback in GnRH and gonadotropin secretion (67).

 

Kisspeptin, GnRH and Puberty

 

Kisspeptin is crucial for normal pubertal development, the discovery of which formed the basis for the obligate role of kisspeptin signaling in the control of reproductive function (206). More than a decade ago two independent groups identified ‘inactivating’ mutations in KISS1R in patients with hypogonadotropic hypogonadism presenting with pubertal delay (85,86). Recently, a male patient with a biallelic loss-of-function KISS1R mutation was described who had undergone a normal and timely puberty, although as a child he had presented with microphallus and bilateral cryptorchidism. This suggests different levels of dependence of the hypothalamic-pituitary-gonadal axis on kisspeptin signaling during the reproductive life span, with the mini-puberty of infancy appearing more dependent on the kisspeptin system than is adolescent puberty (207). On the other hand, activating mutations in KISS1R and KISS1 were then described in children with central precocious puberty (89,90).

 

Hypothalamic expression of Kiss1 and Kiss1R mRNA is upregulated at puberty (117,165,208), and the percentage of GnRH neurons depolarizing in response to kisspeptin increases from juvenile (25%) to pubertal (50%) and to adult mice (>90%) (117), suggesting that GnRH neurons may acquire sensitivity to kisspeptin across puberty. In monkeys, kisspeptin-54 secretion and pulsatility increased at the onset of puberty (209). Moreover, the exogenous administration of kisspeptin resulted in earlier puberty in rats and monkeys (208,210), whereas kisspeptin antagonists delayed puberty in rats (186) and inhibited GnRH release in pubertal monkeys (211). In other study, daily injections of a synthetic kisspeptin analogue have been shown to significantly advance puberty in prepubertal female mice (212). GnRH neuron-specific Kiss1r knockout mouse showed a delay in pubertal onset, abnormal estrous cyclicity in female and abnormal external genitalia in male (microphallus, decreased anogenital distance associated with failure of preputial gland separation) (213).

 

Exogenous kisspeptin stimulated GnRH-induced LH secretion in patients with hypogonadotropism resulted in a spontaneous and permanent activation of their hypothalamic-pituitary-gonadal axis, whereas patients with idiopathic hypogonadotropic hypogonadism and no spontaneous LH pulsatility did not respond to kisspeptin, suggesting that the reversal of hypogonadism, sexual maturation and puberty may well be associated with the acquisition of kisspeptin responsiveness which in turn signals the emergence of reproductive endocrine activity (214). A recent study, 15 children with delayed puberty were administered intravenous kisspeptin and displayed divergent responses, with seven subjects having no response to kisspeptin, whereas others having either robust response (comparable to those of adults) or intermediate responses as perceived in one case (215).

 

GnRH release during puberty appears to require a cooperative mechanism between the kisspeptin/NKB networks in close interaction with different neuropeptides, as substance P, NKA, RFRP-3 and alpha-MSH, working as partners to regulate puberty timing influenced, naturally, by a combination of genetic, environmental, and gene-environment interactions (216).

 

Agonists and antagonists of kisspeptin and NKB were administered into the stalk-median eminence (region with high concentration of GnRH, kisspeptin and NKB neuroterminal fibers), and it was found that both kisspeptin-10 and the NK3R agonist senktide stimulated GnRH release in a dose-responsive manner in prepubertal and pubertal monkeys. However, senktide-induced GnRH release was blocked in the presence of a KISS1R antagonist and the kisspeptin-induced GnRH release was blocked in the presence of NK3R antagonist in pubertal monkeys, leading to the notion that a reciprocal signaling mechanism between kisspeptin and NKB exists and is possibly necessary for a normal puberty (217). These data together emphasizes that disrupted kisspeptin-GPR54-NKB signaling leads to hypogonadotropic hypogonadism, reinforcing the critical role of kisspeptin in puberty.

 

REGULATION OF GnRH AND GONADOTROPIN SECRETION

 

Development and maintenance of normal reproductive function requires a coordinated interplay between neuroendocrine, metabolic, and environmental factors. The GnRH-gonadotropin system plays a central role in the regulation of reproduction by integrating different signals and factors (Figure 3) (126,204). 

 

Figure 3. Neuroendocrine regulation of GnRH/gonadotropin secretion.
The GnRH-gonadotropin system plays a central role in the regulation of reproduction by integrating different neuroendocrine, metabolic and environmental signals/factors. The KNDy signaling has a key role in this process by integrating some of these signals and by regulating GnRH neurons.

 

Overview of Sex Steroid Feedback

 

A crucial role for sex steroids in the regulation of GnRH neurons and/or gonadotropes in humans was initially proposed as serial blood sampling and gonadotropin assays in women through phases of menstrual cycle showed an uneven distribution, with a clear mid-cycle surge in LH and FSH. Two mechanisms were proposed to mediate this effect: first, GnRH secretion is altered in response to the steroid milieu; second, sensitivity of the gonadotropes to a GnRH input is sex-steroid dependent, although the exact mechanism remains controversial due to inter-species variation (218).

 

Hypothalamic secretion of GnRH increases during proesterus in rats (219), sheep (220), and non-human primates (221). Pulsatile once hourly administration of exogenous GnRH restored ovulation in Rhesus monkeys with hypothalamic lesions which abolished GnRH secretion, suggesting that it was the ‘ebb and flow’ of ovarian estrogen feedback acting directly on the pituitary which triggered an LH surge (222). In humans, endogenous GnRH secretion is potentially diminished during the pre-ovulatory LH surge and the suppression of gonadotropin secretion is greater with lower doses of a GnRH receptor antagonist during the mid-cycle surge in comparison to the other phases of the menstrual cycle (223). This suggests that pituitary gonadotrope sensitivity to GnRH is enhanced during the mid-cycle surge. Administration of exogenous estradiol or testosterone in men with hypogonadotropic hypogonadism receiving pulsatile GnRH therapy, decreased gonadotropin concentrations, demonstrating inhibitory effects of sex-steroids at the level of pituitary (224). A direct effect of estrogen on gonadotropes is further demonstrated by the inhibition of LH secretion from rat pituitary gonadotropes in vitro (225). Literature to date suggests that there is dual-site sex-steroid feedback in the regulation of gonadotropin secretion, occurring at the level of both pituitary and hypothalamus (226-231).

 

Estrogen Feedback

 

Patterns of GnRH and LH secretion across the menstrual cycle are modulated by estradiol feedback. A biphasic effect of estradiol on gonadotropin secretion has long been established and it is essential for normal menstrual cycle, with an initial negative feedback (greater suppression of FSH) and a subsequent positive feedback (more prominent for LH) (32). However, the basis for estrogen feedback has been unclear for a long time. GnRH neurons do not express estrogen receptor alpha (ER-alpha) (232,233), and therefore a mediator between gonads and hypothalamus was missed. Recent evidence suggests that kisspeptin and neurokinin B (132) appears to be providing this “missing link” as a key regulator of both negative and positive estrogen feedback (67,126).

 

KNDy neurons in the infundibular nucleus in humans and the arcuate nucleus in other mammals mediate negative estrogen feedback. Estrogen suppresses kisspeptin and neurokinin B release from KNDy neurons, which reduce their stimulatory input to GnRH neurons. Simultaneously, there is a relative deficiency in dynorphin signaling as part of this negative feedback, releasing the inhibitory action on kisspeptin signaling (Figure 2) (67). Immunohistochemical staining of the postmenopausal women hypothalami showed up-regulated expression of KISS1 mRNA and hypertrophy of kisspeptin neurons in the infundibular nucleus when compared to the premenopausal women (111). These hypertrophied kisspeptin neurons co-localized with ER-alpha, had increased expression of neurokinin B and decreased levels of prodynorphin mRNA (234-236). The above evidence for the involvement of the infundibular KNDy system in mediating negative estrogen feedback in humans is consistent with animal studies. Kisspeptin neurons in the arcuate nucleus show frequent co-localization with ER-alpha (160,237). In ovariectomized animals, the expression of Kiss1 and neurokinin B mRNA was up-regulated but prodynorphin mRNA reduced in the arcuate nucleus (equivalent to the infundibular nucleus in humans), and this was reversed by estrogen replacement (102,115,120,238-242). Postmenopausal women are resistant to the stimulatory effect of kisspeptin on LH secretion (142,243), but postmenopausal women receiving estradiol replacement therapy are only resistant to kisspeptin initially and then they do demonstrate a remarkable increase in LH pulse amplitude with direct correlation to the circulating levels of estradiol and duration of kisspeptin administration (243). However, neurokinin B regulates gonadotropin secretion in postmenopausal women, and antagonizing the neurokinin 3 receptor modestly decreases LH secretion in this context (142).

 

Interestingly, the use of neurokinin 3 receptor antagonists has been shown to effectively reduce the frequency and severity of menopause-related vasomotor symptoms owing to their inhibitory effect in the hypothalamic thermoregulatory center, and thus presenting a potential non-hormonal treatment option for menopausal women (144,148,149).

 

Negative estrogen feedback switches to positive feedback in the late follicular phase of menstrual cycle, in order to induce the pre-ovulatory LH surge. Ovarian estradiol seems to be the predominant signal to trigger this switch, via ER-alpha, stimulating RP3V kisspeptin neurons while it inhibits arcuate kisspeptin neurons. Recent evidence supports the role of kisspeptin in generating the LH surge: during an assisted conception cycle, kisspeptin-54, used instead of a routinely administered human chorionic gonadotropin, induced an LH surge, and oocyte maturation, with a subsequent live term birth (241). Repeated twice-daily administration of kisspeptin-54 shortened the menstrual cycle, suggesting that the onset of LH surge was advanced (173). This is further supported by antagonistic studies in animal models, where the administration of kisspeptin antiserum or antagonists blunt/prevent LH peak, whilst kisspeptin advances LH surge (211,244,245).

 

However, kisspeptin-mediated positive estrogen feedback has marked anatomical variations between humans and other species. In rodents, positive estrogen feedback is mediated via the AVPV nucleus, which is absent in humans, other primates and sheep (Figure 2). AVPV neurons are sexually dimorphic, with higher density of ER-alpha described in females and AVPV kisspeptin neurons, as a subset of AVPV neurons, share this pattern (246). There seems to be functional specialization, since only a subset of AVPV kisspeptin neurons (~1/3) are synaptically connected to GnRH cell bodies, but of these, nearly all express estrogen sensitivity and most co-express tyrosine hydroxylase to facilitate positive feedback (247). The expression of Kiss1 mRNA in the AVPV nucleus is low following an ovariectomy but is dramatically increased with estrogen treatment and at the time of LH surge (160,161). In sheep, positive estrogen feedback is mediated though the arcuate nucleus, where the expression of Kiss1 mRNA is the greatest at the pre-ovulatory LH surge (195).

There are no studies looking at the anatomical region of estrogen mediating positive feedback in humans. Although there does not appear to be two distinct anatomical populations of kisspeptin neurons to relay negative and positive sex-steroid feedback in humans, it is possible that separate signaling pathways exists to mediate gonadal steroid feedback.

 

Whilst it is clear that kisspeptin is involved in estrogen-induced mid-cycle gonadotropin surge, the role of KNDy neurons in positive estrogen feedback is less obvious. In sheep, the expression of neurokinin B mRNA was increased during the LH surge, and neurokinin B receptor agonist senktide induced LH secretion mimicking its mid-cycle surge (248,249). However, this has not been reproduced in other species, including humans (180). In summary, KNDy neurons mediate negative estrogen feedback in the infundibular nucleus in humans and the arcuate nucleus in other species. Positive estrogen feedback is mediated via kisspeptin neurons, which show marked inter-species anatomical variation.

 

In addition to the gonads, the brain is one of the major organs producing estradiol, and recently a number of studies demonstrated that estradiol is synthesized and released in the hypothalamus (i.e. neuroestradiol) contributing to the regulation of GnRH release, particularly regarding its positive feedback effect during the preovulatory GnRH/LH surge (250).

 

Progesterone Feedback

 

Progesterone reduces LH pulse frequency in healthy women. LH secretory pattern in women exposed to exogenous progesterone was comparable to LH profile observed in the mid-luteal phase, demonstrating that progesterone plays a central role in the luteal phase of menstrual cycle (251). These inhibitory effects of progesterone on gonadotropin secretion are mediated by the progesterone receptor (PR) (252). The suppressive effect of progesterone on LH secretion was diminished in the context of estrogen deficiency, while co-administration of estradiol restored it (252), suggesting an interplay between these sex steroids. However, the presence of PR on only a small subset of GnRH neurons (253-255)led to the notion that intermediaries are involved in mediating inhibitory progesterone signal to GnRH neurons.

 

There is evidence that KNDy neurons play a role in mediating progesterone feedback on GnRH through dynorphin signaling (Figure 2) (102,120). PR have been demonstrated to be co-localized with dynorphin in the KNDy neurons (159)and progesterone increased dynorphin concentrations (256). Moreover, the number of preprodynorphin mRNA expressing cells decreased in postmenopausal women (236) and in ovariectomized ewes, but normalized with exogenous progesterone to luteal levels (256).

 

Testosterone Feedback

 

Testosterone exerts negative feedback on gonadotropin secretion. Early studies verified that LH and FSH pulse frequency are enhanced in hypogonadal men and exogenous testosterone decreases gonadotropin secretion, suggesting that testosterone have an inhibitory effect on GnRH secretion (230,257).

 

Few GnRH neurons express androgen receptors (AR) (258). GnRH neurons were thus considered to be reliant on an intermediary neuronal population to mediate testosterone feedback. A key role for KNDy neurons in this mediation has been suggested, as these neurons express AR which directly mediate the androgen feedback. The androgen feedback may also rely on the aromatization of testosterone, as testosterone-induced suppression of Kiss1 mRNA in the rodent arcuate nucleus is identical to that observed with estradiol, but more than that observed with dihydrotestosterone administration (259). The cross-talk between AR and ER was suggested from animal studies: AR expression was downregulated in the prostate following neonatal estrogen exposure (260), and AR transcription was modulated following a co-transfection of AR and ER (261).

 

Navarro has described a role for KNDy neurons in mediating the negative testosterone feedback on GnRH secretion, and provided evidence that neurokinin B released from KNDy neurons is part of an auto-feedback loop that generates the pulsatile secretion of Kiss1 and GnRH in male mice: Kiss1 and dynorphin mRNA are regulated by testosterone through estrogen and androgen receptor-dependent pathways; KNDy neurons express neurokinin B receptor whereas GnRH neurons do not, and senktide (an agonist for the neurokinin B receptor) activates KNDy neurons leading to gonadotropin secretion but has no discernible effect on GnRH neurons (262). Other studies demonstrated that the suppression of gonadotropin secretion using testosterone is associated with a reduction of Kiss1 mRNA in the hypothalamus (118,208,263). Moreover, post-orchidectomy rise in LH in rodents can be blocked by kisspeptin antagonists, further suggesting that kisspeptin system mediates the hypothalamic androgen feedback (186).

 

Stress and Glucocorticoids

 

Physical and psychological stress is associated with hypothalamic amenorrhea, possibly though the activation of hypothalamic-pituitary-adrenal (HPA) axis (264,265). Experimental evidence points towards a cortisol-mediated suppression of gonadotropin secretion as the main key pathway to explain stress-induced gonadotropin suppression(55,266-273). The negative effect of cortisol on HPG axis is recognized to occur at both pituitary and hypothalamic levels. There are also data suggesting that upstream factors in the HPA axis, such as Corticotropin Releasing Hormone (CRH) and vasopressin may play a mediatory role (274,275).

 

Cortisol secretion in women with hypothalamic amenorrhea is elevated (267), and evening adrenocorticotropic hormone (ACTH) and cortisol concentrations are higher in excessive exercise (266,270). Administration of exogenous glucocorticoids to eugonadal women was associated with a decrease in LH pulse frequency, suggesting that glucocorticoids have a negative action on GnRH secretion (273). In ovine portal blood, cortisol administration led to a decrease in GnRH pulse frequency (272). Inferences of cortisol effects on gonadotropin secretion were also derived from observations in women and men with Cushing’s syndrome (condition associated with excessive cortisol secretion), where exogenous GnRH preferentially stimulates FSH whilst LH remains unchanged (268,271). The resolution of male hypogonadotropic hypogonadism was also observed in men with remission of Cushing’s disease (271). This negative input of cortisol on the HPG axis may be modulated by sex-steroid hormones, and kisspeptin signaling has also been implicated in the process.

 

Cortisol alone had no impact on GnRH pulsatility in ovariectomized ewes, but the co-administration of estradiol and progesterone led to a 70% decrease in GnRH secretion (272). Decreased hypothalamic Kiss1 mRNA expression has been observed during exposure to stress or exogenous glucocorticoids. The role of kisspeptin in mediating stress inputs is further supported by the expression of glucocorticoid receptor on murine kisspeptin neurons (276). Colocalization by immunohistochemistry of CRH receptor (CRH-R) in most hypothalamic kisspeptin neurons in the AVPV/PeN and ARC nuclei as well as glucocorticoid receptor (GR) in AVPV/PeN kisspeptin neurons support a relevant direct role of kisspeptin neurons in the inhibitory effects of CRH/ glucocorticoids (277).

 

Hypothalamic CRH neurons, important regulators of the stress response, also directly modulate GnRH excitability in a dose-dependent and receptor-specific manner, and the GnRH response to CRH is influenced by estrogens (278). Intracerebroventricular administration of CRH in female rats suppressed LH pulsatility and the LH surge, and this suppression was enhanced by estrogens (279).

 

Animal models have also linked increased exposure to RFamide-related peptide-3 (RFRP-3) during acute and chronic stress and hypothalamic expression of GnIH mRNA. Along these lines, the surface of GnIH neurons has glucocorticoid receptors and hydrocortisone administration was associated to an increased GnIH mRNA expression, ultimately leading to lower GnRH activity and dysregulation of the HPG axis (280). Together, these findings emphasize that kisspeptin as GnIH provide relevant inputs that contribute to an inhibitory effect of corticosteroids on gonadal axis during stress.

 

Prolactin

 

Prolactin is a well-known inhibitor of GnRH release and a suppressor of the HPG axis. The association between hyperprolactinemia and reproductive dysfunction has long been established, accounting for 14% of secondary amenorrhea and hypogonadism cases (281) and for a third of women presenting with infertility (282,283). Hyperprolactinemia is evident in 16% of men with erectile dysfunction and in 11% of men with oligospermia (284). The decreased pulsatility of LH in hyperprolactinemia responds to bromocriptine (285). GnRH therapy has restored ovulation and normal luteal function in bromocriptine resistant hyperprolactinemia women (286,287), suggesting that prolactin exerts inhibition through direct reduction of GnRH secretion.

 

The neuroendocrine pathway by which prolactin inhibits GnRH pulse frequency remains to be fully elucidated. A direct action of prolactin on the GnRH neuronal network is possible (288,289). Prolactin has also been demonstrated to influence other systems, including GABA (290), β endorphins (291), neuropeptide Y (292) and dopaminergic systems (via tuberoinfundibular dopamine (TIDA) neurons) (293).

 

Nevertheless, data suggest that prolactin receptors are expressed in most kisspeptin neurons but only in a small proportion of GnRH neurons, indicating that kisspeptin signaling may have a role in this context (288,294). In rodent models, kisspeptin neurons in the arcuate nucleus modulate dopamine release from dopaminergic neurons, thereby regulating prolactin secretion (295). Kiss1 expression is decreased in lactation, a physiological state associated with hyperprolactinemia (296). Prolactin-sensitive GABA and kisspeptin neurons were identified in regions of the rat hypothalamus (294). Moreover, in a mouse model of anovulatory hyperprolactinemia (induced by a continuous infusion of prolactin), Kiss1 mRNA levels were diminished and peripheral administration of kisspeptin restored gonadotropin secretion and ovarian cyclicity (297). There are also other animal studies reporting an inhibitory effect of prolactin on Kiss1 expression (298,299). This data suggests that kisspeptin is a possible link between hyperprolactinemia and GnRH deficiency. The administration of kisspeptin-10 reactivated the gonadotropin secretion in women with hyperprolactinemia-induced hypogonadotropic amenorrhea, suggesting that GnRH deficiency in the context of hyperprolactinemia is, at least in part, mediated by an impaired hypothalamic kisspeptin secretion (300).

 

On the other hand, kisspeptins appears to have a stimulatory effect on prolactin release, as demonstrated in a recent study in ovariectomized rats which had intracerebroventricular injections of kisspeptin-10 with subsequent increase in prolactin release, and this required the estrogen receptor-alpha and was potentiated by progesterone via progesterone receptor activation (301).

 

Nutrition and Metabolism

 

A link between energy balance and reproductive function enables organisms to survive to reproductive maturity and to withstand the energy needs of parturition, lactation, and other parental behaviors. This link optimizes reproductive success under fluctuating metabolic conditions (302). Kisspeptin signaling may link nutrition/metabolic status and reproduction by sensing energy stores and translate this information into GnRH secretion (303). These relations elucidate further associations between reproductive dysfunction and metabolic disturbances, such as diabetes, obesity or anorexia nervosa (67,304,305).

 

Food deprivation impairs GnRH and gonadotropin secretion, and leptin (a satiety hormone secreted by adipose tissue, the levels of which drop in response to fasting) plays a role in this inter-regulation by stimulating LH release (67,306-308). Periods of fasting and calorie restriction decrease LH pulse frequency and increase pulse amplitude (302,309-311). Administration of recombinant leptin increased LH pulse frequency in women with hypothalamic amenorrhea (312) and prevented fasting-induced drop in testosterone and LH pulsatility in healthy men (313). Moreover, humans with mutations in leptin or in leptin receptor show hypogonadism (314). Thus, the crosstalk between kisspeptin and leptin is relevant for reproduction and fertility (71), including in the setting of assisted reproduction techniques (315).

 

Kisspeptin neurons may have a role in mediating the metabolic signals of leptin on the control of HPG axis, as 40% of the arcuate kisspeptin neurons express leptin receptors in contrast to the GnRH neurons, where leptin receptors are absent (316-319). Food deprivation is associated with a decrease in kisspeptin, and subsequent reduction in gonadotropin secretion (320-323). Levels of low Kiss1 mRNA expression in the leptin-deficient ob/ob mice are partially upregulated by exogenous leptin (161). Moreover, exogenous kisspeptin restored vaginal opening (marker of sexual maturation) in malnourished rodents (320). Animal models of type 1 diabetes, characterized by insulin deficiency and impaired cellular nutrition, had hypogonadotropic hypogonadism and decreased Kiss1 mRNA expression. Repeated administration of kisspeptin to these rodents increased prostate and testis weight (324). It is plausible that a relative deficiency of kisspeptin secretion is a mechanism for hypogonadotropic hypogonadism in patients with obesity and diabetes (179). In hypogonadal men with type 2 diabetes, kisspeptin-10 increased LH secretion and pulse frequency (179). Although early studies appeared to suggest a direct link between kisspeptin and leptin, it seems that the neuronal pathway whereby leptin modulates GnRH is far more complex (325,326). Only partial restoration in Kiss1 mRNA in leptin-deficiency and normal pubertal development and fertility observed in selective leptin receptor deletion from kisspeptin neurons suggest that kisspeptin may link reproduction and metabolism through other ways than leptin (161,327). Proopiomelanocortin (POMC), agouti-related peptide, neuropeptide Y, ghrelin, and cocaine- and amphetamine-regulated transcript (CART) expressing neurons have been linked to this process (303,319). Kisspeptin neurons communicate with POMC and neuropeptide Y neurons and are able to modulate the expression of relevant genes in these cells (316). This link between kisspeptin and other peptides classically associated to food intake (as POMC and neuropeptide Y) was explored due to the anorexigenic effect of intracerebroventricular administration of KP-10 in male rats mediated via anorectic neuropeptides, nesfatin-1 and oxytocin, expressed in various hypothalamic nuclei. Diminished food intake and anorexia was significantly abolished by pretreatment with oxytocin receptor antagonist (328,329).

 

Several studies have also suggested that ghrelin can interact directly with hypothalamic neurons leading to suppression of gonadotropins release, and thus impairing fertility, an effect that is dependent of the estradiol milieu (303,330-332).

 

GABA (Gamma-Amino Butyric Acid)

 

GABA has also been implicated as a regulator of GnRH secretion. Although GABA is classically an inhibitory neurotransmitter in the central nervous system, most mature GnRH neurons are stimulated by GABA, which has attributed to GABA an excitatory action in HPG axis. The precise physiology of this mechanism is still unclear (333-337), but it may be related to the bidirectional interactions between GABA and kisspeptin pathways, as well as between these and GnRH neurons, in a variety of ways throughout development (338). In early development, GABA seems to increase KISS1 expression in embryonic phase and early postnatally, while in the absence of GABAergic input the expression of KISS1 declines (338,339). In the prepubertal period, the central restraint on GnRH secretion seems to be mediated by GABA possibly acting directly via kisspeptin neurons (338). In the peri-pubertal phase, the antagonism of GABA and the intrinsic disinhibition of kisspeptin neurons seem to be critical in puberty initiation and development (340,341). In adulthood, the interactions between GnRH-GABA-kisspeptin become more complex with HPG axis function critically dependent on such interactions. For instance, the preovulatory surge does not occur in the absence of GABA signaling, thus neurons co-expressing GABA and kisspeptin seem crucial in providing double excitatory input to GnRH neurons at the time of ovulation (338,342).

 

Additionally, in healthy men, total endogenous GABA levels in the anterior cingulate cortex, a key limbic structure, significantly decreased after intravenous infusion of kisspeptin (1 nmol/kg/h) demonstrating a potent inhibitory effect of kisspeptin on GABA levels which could be a fundamental concept in understanding the central limbic effect of kisspeptin in the human brain (343).

 

Other Neuropeptides

 

In addition to KNDy system and GABA, other peptides and neurotransmitters have been shown to influence GnRH-gonadotrope system: vasoactive intestinal polypeptide (VIP), vasopressin, catecholamines, nitric oxide, neurotensin, gonadotropin-inhibitory hormone (GnIH) /RFamide related peptide-3 (RFRP-3) (337), nucleobindin-2/nesfatin-1 (344). Excitatory inputs to the HPG axis may be mediated by VIP, catecholamines, glutamate and possibly vasopressin, whereas GnIH in birds, or its mammalian homolog RFRP-3, provide inhibitory inputs (345-349). RFRP neuronal populations have been detected mainly in the hypothalamic dorsomedial nucleus or adjacent regions, and they have projections to several hypothalamic areas including the arcuate nucleus, paraventricular nucleus, ventromedial nucleus and the lateral hypothalamus, all areas with major roles in the regulation of reproduction and energy balance (350,351). RFRP-3, encoded by the gene Rfrp, inhibits the electric firing of GnRH and kisspeptin neurons (346,352), which results in a suppression of GnRH-induced gonadotropin release with consequent inhibition of the reproductive axis (353). This RFRP-3 inhibitory input on the gonadotropin release is influenced by estrogens and may well be involved in their negative feedback. Estrogens reduce RFRP-3 expression and RFRP-3 neuronal activation (354,355).

 

Particular attention has been paid to the role of glutamate as a stimulatory modulator of the activity of ARC kisspeptin neurons, reaffirming the role of kisspeptin as a major neural integrator of inputs to GnRH neurons. Data from Kiss1 KO rats showed failure to increase GnRH/LH secretion following monosodium glutamate/NMDA administration (356).

 

SUMMARY

 

Complex neuroendocrine networks coordinate the regulation of reproduction, integrating a wide range of internal and external environmental inputs and signals. GnRH, the principal regulator of reproduction integrates cues from sex steroids, stress, glucocorticoids, nutritional and metabolic status, prolactin and other peptides, to controls gonadotropin secretion and subsequently gonadal function. Recently, the KNDy neuronal network has emerged as essential gatekeeper of GnRH release and thus reproduction, fertility and puberty. Translational clinical studies, exploring kisspeptin and neurokinin B activity in various physiological and pathological states are pivotal to explore potential clinical applications for these novel neuropeptides and their agonists as well as antagonists, may underpin future management of some disorders with dysfunctional GnRH pulsatility, such polycystic ovary syndrome, hypothalamic amenorrhea, infertility, obesity, pubertal disorders and menopause-related symptoms.

 

REFERENCES

 

  1. Herbison AE, Theodosis DT. Localization of oestrogen receptors in preoptic neurons containing neurotensin but not tyrosine hydroxylase, cholecystokinin or luteinizing hormone-releasing hormone in the male and female rat. Neuroscience. 1992;50(2):283-298.
  2. Fink G. Neuroendocrine regulation of pituitary function: general principles. Neuroendocrinology in Physiology and Medicine. 2000:p.107-134.
  3. Tena-Sempere M. Hypothalamic KiSS-1: the missing link in gonadotropin feedback control? Endocrinology. 2005;146(9):3683-3685.
  4. Baba Y, Matsuo H, Schally AV. Structure of the porcine LH- and FSH-releasing hormone. II. Confirmation of the proposed structure by conventional sequential analyses. Biochem Biophys Res Commun. 1971;44(2):459-463.
  5. Matsuo H, Arimura A, Nair RM, Schally AV. Synthesis of the porcine LH- and FSH-releasing hormone by the solid-phase method. Biochem Biophys Res Commun. 1971;45(3):822-827.
  6. Schally AV, Arimura A, Kastin AJ, Matsuo H, Baba Y, Redding TW, Nair RM, Debeljuk L, White WF. Gonadotropin-releasing hormone: one polypeptide regulates secretion of luteinizing and follicle-stimulating hormones. Science. 1971;173(4001):1036-1038.
  7. Schally AV, Arimura A, Baba Y, Nair RM, Matsuo H, Redding TW, Debeljuk L. Isolation and properties of the FSH and LH-releasing hormone. Biochem Biophys Res Commun. 1971;43(2):393-399.
  8. Schally AV. Use of GnRH in preference to LH-RH terminology in scientific papers. Hum Reprod. 2000;15(9):2059-2061.
  9. Millar RP. GnRHs and GnRH receptors. Anim Reprod Sci. 2005;88(1-2):5-28.
  10. Okubo K, Nagahama Y. Structural and functional evolution of gonadotropin-releasing hormone in vertebrates. Acta Physiol (Oxf). 2008;193(1):3-15.
  11. Miyamoto K, Hasegawa Y, Nomura M, Igarashi M, Kangawa K, Matsuo H. Identification of the second gonadotropin-releasing hormone in chicken hypothalamus: evidence that gonadotropin secretion is probably controlled by two distinct gonadotropin-releasing hormones in avian species. Proc Natl Acad Sci U S A. 1984;81(12):3874-3878.
  12. White SA, Bond CT, Francis RC, Kasten TL, Fernald RD, Adelman JP. A second gene for gonadotropin-releasing hormone: cDNA and expression pattern in the brain. Proc Natl Acad Sci U S A. 1994;91(4):1423-1427.
  13. Gault PM, Maudsley S, Lincoln GA. Evidence that gonadotropin-releasing hormone II is not a physiological regulator of gonadotropin secretion in mammals. J Neuroendocrinol. 2003;15(9):831-839.
  14. Balasubramanian R, Dwyer A, Seminara SB, Pitteloud N, Kaiser UB, Crowley WF, Jr. Human GnRH deficiency: a unique disease model to unravel the ontogeny of GnRH neurons. Neuroendocrinology. 2010;92(2):81-99.
  15. Mitchell AL, Dwyer A, Pitteloud N, Quinton R. Genetic basis and variable phenotypic expression of Kallmann syndrome: towards a unifying theory. Trends Endocrinol Metab. 2011;22(7):249-258.
  16. Cariboni A, Pimpinelli F, Colamarino S, Zaninetti R, Piccolella M, Rumio C, Piva F, Rugarli EI, Maggi R. The product of X-linked Kallmann's syndrome gene (KAL1) affects the migratory activity of gonadotropin-releasing hormone (GnRH)-producing neurons. Hum Mol Genet. 2004;13(22):2781-2791.
  17. Cariboni A, Hickok J, Rakic S, Andrews W, Maggi R, Tischkau S, Parnavelas JG. Neuropilins and their ligands are important in the migration of gonadotropin-releasing hormone neurons. J Neurosci. 2007;27(9):2387-2395.
  18. Magni P, Dozio E, Ruscica M, Watanobe H, Cariboni A, Zaninetti R, Motta M, Maggi R. Leukemia inhibitory factor induces the chemomigration of immortalized gonadotropin-releasing hormone neurons through the independent activation of the Janus kinase/signal transducer and activator of transcription 3, mitogen-activated protein kinase/extracellularly regulated kinase 1/2, and phosphatidylinositol 3-kinase/Akt signaling pathways. Mol Endocrinol. 2007;21(5):1163-1174.
  19. 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. Nat Genet. 2003;33(4):463-465.
  20. Kim SH, Hu Y, Cadman S, Bouloux P. Diversity in fibroblast growth factor receptor 1 regulation: learning from the investigation of Kallmann syndrome. J Neuroendocrinol. 2008;20(2):141-163.
  21. Yoshida K, Rutishauser U, Crandall JE, Schwarting GA. Polysialic acid facilitates migration of luteinizing hormone-releasing hormone neurons on vomeronasal axons. J Neurosci. 1999;19(2):794-801.
  22. Kaprara A, Huhtaniemi IT. The hypothalamus-pituitary-gonad axis: Tales of mice and men. Metabolism. 2017.
  23. Martin C, Balasubramanian R, Dwyer AA, Au MG, Sidis Y, Kaiser UB, Seminara SB, Pitteloud N, Zhou QY, Crowley WF, Jr. The role of the prokineticin 2 pathway in human reproduction: evidence from the study of human and murine gene mutations. Endocr Rev. 2011;32(2):225-246.
  24. Pitteloud N, Zhang C, Pignatelli D, Li JD, Raivio T, Cole LW, Plummer L, Jacobson-Dickman EE, Mellon PL, Zhou QY, Crowley WF, Jr. Loss-of-function mutation in the prokineticin 2 gene causes Kallmann syndrome and normosmic idiopathic hypogonadotropic hypogonadism. Proc Natl Acad Sci U S A. 2007;104(44):17447-17452.
  25. Clifton DKS, R.A. . Neuroendocrinology of reproduction. Yen & Jaffe's Reproductive Endocrinology JF Strauss and RL Barberi, Elsevier. 2009.
  26. Maeda K, Ohkura S, Uenoyama Y, Wakabayashi Y, Oka Y, Tsukamura H, Okamura H. Neurobiological mechanisms underlying GnRH pulse generation by the hypothalamus. Brain Res. 2010;1364:103-115.
  27. Moenter SM, DeFazio AR, Pitts GR, Nunemaker CS. Mechanisms underlying episodic gonadotropin-releasing hormone secretion. Front Neuroendocrinol. 2003;24(2):79-93.
  28. Carmel PW, Araki S, Ferin M. Pituitary stalk portal blood collection in rhesus monkeys: evidence for pulsatile release of gonadotropin-releasing hormone (GnRH). Endocrinology. 1976;99(1):243-248.
  29. Antunes JL, Carmel PW, Housepian EM, Ferin M. Luteinizing hormone-releasing hormone in human pituitary blood. J Neurosurg. 1978;49(3):382-386.
  30. Caraty A, Martin GB, Montgomery G. A new method for studying pituitary responsiveness in vivo using pulses of LH-RH analogue in ewes passively immunized against native LH-RH. Reprod Nutr Dev. 1984;24(4):439-448.
  31. Lincoln GA, Fraser HM. Blockade of episodic secretion of luteinizing hormone in the ram by the administration of antibodies to luteinizing hormone releasing hormone. Biol Reprod. 1979;21(5):1239-1245.
  32. Knobil E, Plant TM, Wildt L, Belchetz PE, Marshall G. Control of the rhesus monkey menstrual cycle: permissive role of hypothalamic gonadotropin-releasing hormone. Science. 1980;207(4437):1371-1373.
  33. Bolt DJ. Changes in the concentration of luteinizing hormone in plasma of rams following administration of oestradiol, progesterone or testosterone. J Reprod Fertil. 1971;24(3):435-438.
  34. Dierschke DJ, Bhattacharya AN, Atkinson LE, Knobil E. Circhoral oscillations of plasma LH levels in the ovariectomized rhesus monkey. Endocrinology. 1970;87(5):850-853.
  35. Naftolin F, Yen SS, Tsai CC. Rapid cycling of plasma gonadotrophins in normal men as demonstrated by frequent sampling. Nat New Biol. 1972;236(64):92-93.
  36. Yen SS, Tsai CC. The biphasic pattern in the feedback action of ethinyl estradiol on the release of pituitary FSH and LH. J Clin Endocrinol Metab. 1971;33(6):882-887.
  37. Reame NE, Sauder SE, Case GD, Kelch RP, Marshall JC. Pulsatile gonadotropin secretion in women with hypothalamic amenorrhea: evidence that reduced frequency of gonadotropin-releasing hormone secretion is the mechanism of persistent anovulation. J Clin Endocrinol Metab. 1985;61(5):851-858.
  38. Sauder SE, Frager M, Case GD, Kelch RP, Marshall JC. Abnormal patterns of pulsatile luteinizing hormone secretion in women with hyperprolactinemia and amenorrhea: responses to bromocriptine. J Clin Endocrinol Metab. 1984;59(5):941-948.
  39. Belchetz PE, Plant TM, Nakai Y, Keogh EJ, Knobil E. Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin-releasing hormone. Science. 1978;202(4368):631-633.
  40. Rasmussen DD. Episodic gonadotropin-releasing hormone release from the rat isolated median eminence in vitro. Neuroendocrinology. 1993;58(5):511-518.
  41. Thiery JC, Pelletier J. Multiunit activity in the anterior median eminence and adjacent areas of the hypothalamus of the ewe in relation to LH secretion. Neuroendocrinology. 1981;32(4):217-224.
  42. Wilson RC, Kesner JS, Kaufman JM, Uemura T, Akema T, Knobil E. Central electrophysiologic correlates of pulsatile luteinizing hormone secretion in the rhesus monkey. Neuroendocrinology. 1984;39(3):256-260.
  43. Martinez de la Escalera G, Choi AL, Weiner RI. Generation and synchronization of gonadotropin-releasing hormone (GnRH) pulses: intrinsic properties of the GT1-1 GnRH neuronal cell line. Proc Natl Acad Sci U S A. 1992;89(5):1852-1855.
  44. Terasawa E, Keen KL, Mogi K, Claude P. Pulsatile release of luteinizing hormone-releasing hormone (LHRH) in cultured LHRH neurons derived from the embryonic olfactory placode of the rhesus monkey. Endocrinology. 1999;140(3):1432-1441.
  45. Ezzat A, Pereira A, Clarke IJ. Kisspeptin is a component of the pulse generator for GnRH secretion in female sheep but not the pulse generator. Endocrinology. 2015;156(5):1828-1837.
  46. Millar RP, Lu ZL, Pawson AJ, Flanagan CA, Morgan K, Maudsley SR. Gonadotropin-releasing hormone receptors. Endocr Rev. 2004;25(2):235-275.
  47. Pincus SM, Padmanabhan V, Lemon W, Randolph J, Rees Midgley A. Follicle-stimulating hormone is secreted more irregularly than luteinizing hormone in both humans and sheep. J Clin Invest. 1998;101(6):1318-1324.
  48. Padmanabhan V, McFadden K, Mauger DT, Karsch FJ, Midgley AR, Jr. Neuroendocrine control of follicle-stimulating hormone (FSH) secretion. I. Direct evidence for separate episodic and basal components of FSH secretion. Endocrinology. 1997;138(1):424-432.
  49. Dalkin AC, Haisenleder DJ, Ortolano GA, Ellis TR, Marshall JC. The frequency of gonadotropin-releasing-hormone stimulation differentially regulates gonadotropin subunit messenger ribonucleic acid expression. Endocrinology. 1989;125(2):917-924.
  50. Haisenleder DJ, Dalkin AC, Ortolano GA, Marshall JC, Shupnik MA. A pulsatile gonadotropin-releasing hormone stimulus is required to increase transcription of the gonadotropin subunit genes: evidence for differential regulation of transcription by pulse frequency in vivo. Endocrinology. 1991;128(1):509-517.
  51. Kaiser UB, Jakubowiak A, Steinberger A, Chin WW. Differential effects of gonadotropin-releasing hormone (GnRH) pulse frequency on gonadotropin subunit and GnRH receptor messenger ribonucleic acid levels in vitro. Endocrinology. 1997;138(3):1224-1231.
  52. Spratt DI, Finkelstein JS, Butler JP, Badger TM, Crowley WF, Jr. Effects of increasing the frequency of low doses of gonadotropin-releasing hormone (GnRH) on gonadotropin secretion in GnRH-deficient men. J Clin Endocrinol Metab. 1987;64(6):1179-1186.
  53. Waldhauser F, Weissenbacher G, Frisch H, Pollak A. Pulsatile secretion of gonadotropins in early infancy. Eur J Pediatr. 1981;137(1):71-74.
  54. Conte FA, Grumbach MM, Kaplan SL, Reiter EO. Correlation of luteinizing hormone-releasing factor-induced luteinizing hormone and follicle-stimulating hormone release from infancy to 19 years with the changing pattern of gonadotropin secretion in agonadal patients: relation to the restraint of puberty. J Clin Endocrinol Metab. 1980;50(1):163-168.
  55. Pohl CR, deRidder CM, Plant TM. Gonadal and nongonadal mechanisms contribute to the prepubertal hiatus in gonadotropin secretion in the female rhesus monkey (Macaca mulatta). J Clin Endocrinol Metab. 1995;80(7):2094-2101.
  56. Boyar R, Finkelstein J, Roffwarg H, Kapen S, Weitzman E, Hellman L. Synchronization of augmented luteinizing hormone secretion with sleep during puberty. N Engl J Med. 1972;287(12):582-586.
  57. Roth JC, Kelch RP, Kaplan SL, Grumbach MM. FSH and LH response to luteinizing hormone-releasing factor in prepubertal and pubertal children, adult males and patients with hypogonadotropic and hypertropic hypogonadism. J Clin Endocrinol Metab. 1972;35(6):926-930.
  58. Marshall JC, Dalkin AC, Haisenleder DJ, Griffin ML, Kelch RP. GnRH pulses--the regulators of human reproduction. Trans Am Clin Climatol Assoc. 1993;104:31-46.
  59. Marshall JC, Dalkin AC, Haisenleder DJ, Paul SJ, Ortolano GA, Kelch RP. Gonadotropin-releasing hormone pulses: regulators of gonadotropin synthesis and ovulatory cycles. Recent Prog Horm Res. 1991;47:155-187; discussion 188-159.
  60. Yen SS, Tsai CC, Naftolin F, Vandenberg G, Ajabor L. Pulsatile patterns of gonadotropin release in subjects with and without ovarian function. J Clin Endocrinol Metab. 1972;34(4):671-675.
  61. Arroyo A, Laughlin GA, Morales AJ, Yen SS. Inappropriate gonadotropin secretion in polycystic ovary syndrome: influence of adiposity. J Clin Endocrinol Metab. 1997;82(11):3728-3733.
  62. Morales AJ, Laughlin GA, Butzow T, Maheshwari H, Baumann G, Yen SS. Insulin, somatotropic, and luteinizing hormone axes in lean and obese women with polycystic ovary syndrome: common and distinct features. J Clin Endocrinol Metab. 1996;81(8):2854-2864.
  63. Waldstreicher J, Santoro NF, Hall JE, Filicori M, Crowley WF, Jr. Hyperfunction of the hypothalamic-pituitary axis in women with polycystic ovarian disease: indirect evidence for partial gonadotroph desensitization. J Clin Endocrinol Metab. 1988;66(1):165-172.
  64. Yen SS, Vela P, Rankin J. Inappropriate secretion of follicle-stimulating hormone and luteinizing hormone in polycystic ovarian disease. J Clin Endocrinol Metab. 1970;30(4):435-442.
  65. Beltramo M, Dardente H, Cayla X, Caraty A. Cellular mechanisms and integrative timing of neuroendocrine control of GnRH secretion by kisspeptin. Mol Cell Endocrinol. 2014;382(1):387-399.
  66. Jayasena CN, Abbara A, Izzi-Engbeaya C, Comninos AN, Harvey RA, Gonzalez Maffe J, Sarang Z, Ganiyu-Dada Z, Padilha AI, Dhanjal M, Williamson C, Regan L, Ghatei MA, Bloom SR, Dhillo WS. Reduced levels of plasma kisspeptin during the antenatal booking visit are associated with increased risk of miscarriage. J Clin Endocrinol Metab. 2014;99(12):E2652-2660.
  67. Skorupskaite K, George JT, Anderson RA. The kisspeptin-GnRH pathway in human reproductive health and disease. Hum Reprod Update. 2014;20(4):485-500.
  68. Clarke SA, Dhillo WS. Kisspeptin across the human lifespan:evidence from animal studies and beyond. J Endocrinol. 2016;229(3):R83-98.
  69. Comninos AN, Dhillo WS. Emerging Roles of Kisspeptin in Sexual and Emotional Brain Processing. Neuroendocrinology. 2018;106(2):195-202.
  70. Dudek M, Ziarniak K, Sliwowska JH. Kisspeptin and Metabolism: The Brain and Beyond. Front Endocrinol (Lausanne). 2018;9:145.
  71. Wahab F, Atika B, Ullah F, Shahab M, Behr R. Metabolic Impact on the Hypothalamic Kisspeptin-Kiss1r Signaling Pathway. Front Endocrinol (Lausanne). 2018;9:123.
  72. Wolfe A, Hussain MA. The Emerging Role(s) for Kisspeptin in Metabolism in Mammals. Front Endocrinol (Lausanne). 2018;9:184.
  73. Yang L, Comninos AN, Dhillo WS. Intrinsic links among sex, emotion, and reproduction. Cell Mol Life Sci. 2018;75(12):2197-2210.
  74. Mills EG, Yang L, Abbara A, Dhillo WS, Comninos AN. Current Perspectives on Kisspeptins Role in Behaviour. Front Endocrinol (Lausanne). 2022;13:928143.
  75. Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman BE, Welch DR. KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J Natl Cancer Inst. 1996;88(23):1731-1737.
  76. Ciaramella V, Della Corte CM, Ciardiello F, Morgillo F. Kisspeptin and Cancer: Molecular Interaction, Biological Functions, and Future Perspectives. Front Endocrinol (Lausanne). 2018;9:115.
  77. West A, Vojta PJ, Welch DR, Weissman BE. Chromosome localization and genomic structure of the KiSS-1 metastasis suppressor gene (KISS1). Genomics. 1998;54(1):145-148.
  78. Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E, Brezillon S, Tyldesley R, Suarez-Huerta N, Vandeput F, Blanpain C, Schiffmann SN, Vassart G, Parmentier M. The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem. 2001;276(37):34631-34636.
  79. Pasquier J, Kamech N, Lafont AG, Vaudry H, Rousseau K, Dufour S. Molecular evolution of GPCRs: Kisspeptin/kisspeptin receptors. J Mol Endocrinol. 2014;52(3):T101-117.
  80. Gottsch ML, Navarro VM, Zhao Z, Glidewell-Kenney C, Weiss J, Jameson JL, Clifton DK, Levine JE, Steiner RA. Regulation of Kiss1 and dynorphin gene expression in the murine brain by classical and nonclassical estrogen receptor pathways. J Neurosci. 2009;29(29):9390-9395.
  81. Lee DK, Nguyen T, O'Neill GP, Cheng R, Liu Y, Howard AD, Coulombe N, Tan CP, Tang-Nguyen AT, George SR, O'Dowd BF. Discovery of a receptor related to the galanin receptors. FEBS Lett. 1999;446(1):103-107.
  82. Muir AI, Chamberlain L, Elshourbagy NA, Michalovich D, Moore DJ, Calamari A, Szekeres PG, Sarau HM, Chambers JK, Murdock P, Steplewski K, Shabon U, Miller JE, Middleton SE, Darker JG, Larminie CG, Wilson S, Bergsma DJ, Emson P, Faull R, Philpott KL, Harrison DC. AXOR12, a novel human G protein-coupled receptor, activated by the peptide KiSS-1. J Biol Chem. 2001;276(31):28969-28975.
  83. Ohtaki T, Shintani Y, Honda S, Matsumoto H, Hori A, Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y, Ishibashi Y, Watanabe T, Asada M, Yamada T, Suenaga M, Kitada C, Usuki S, Kurokawa T, Onda H, Nishimura O, Fujino M. Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature. 2001;411(6837):613-617.
  84. Liu X, Lee K, Herbison AE. Kisspeptin excites gonadotropin-releasing hormone neurons through a phospholipase C/calcium-dependent pathway regulating multiple ion channels. Endocrinology. 2008;149(9):4605-4614.
  85. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100(19):10972-10976.
  86. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Jr., Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB, Slaugenhaupt SA, Gusella JF, O'Rahilly S, Carlton MB, Crowley WF, Jr., Aparicio SA, Colledge WH. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349(17):1614-1627.
  87. Tenenbaum-Rakover Y, Commenges-Ducos M, Iovane A, Aumas C, Admoni O, de Roux N. Neuroendocrine phenotype analysis in five patients with isolated hypogonadotropic hypogonadism due to a L102P inactivating mutation of GPR54. J Clin Endocrinol Metab. 2007;92(3):1137-1144.
  88. Chan YM, Broder-Fingert S, Wong KM, Seminara SB. Kisspeptin/Gpr54-independent gonadotrophin-releasing hormone activity in Kiss1 and Gpr54 mutant mice. J Neuroendocrinol. 2009;21(12):1015-1023.
  89. Teles MG, Bianco SD, Brito VN, Trarbach EB, Kuohung W, Xu S, Seminara SB, Mendonca BB, Kaiser UB, Latronico AC. A GPR54-activating mutation in a patient with central precocious puberty. N Engl J Med. 2008;358(7):709-715.
  90. Silveira LG, Noel SD, Silveira-Neto AP, Abreu AP, Brito VN, Santos MG, Bianco SD, Kuohung W, Xu S, Gryngarten M, Escobar ME, Arnhold IJ, Mendonca BB, Kaiser UB, Latronico AC. Mutations of the KISS1 gene in disorders of puberty. J Clin Endocrinol Metab. 2010;95(5):2276-2280.
  91. Dong L, Zhou W, Lin Z, Tang L, Deng X, Chen B, Huang W, Xiong Q. Polymorphism rs5780218, rs12998 and rs10158616 in KISS1 gene among the Hubei province Chinese girls with the central precocious puberty. Endocrine. 2024;84(3):1229-1237.
  92. Topaloglu AK, Reimann F, Guclu M, Yalin AS, Kotan LD, Porter KM, Serin A, Mungan NO, Cook JR, Imamoglu S, Akalin NS, Yuksel B, O'Rahilly S, Semple RK. TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for Neurokinin B in the central control of reproduction. Nat Genet. 2009;41(3):354-358.
  93. Gianetti E, Tusset C, Noel SD, Au MG, Dwyer AA, Hughes VA, Abreu AP, Carroll J, Trarbach E, Silveira LF, Costa EM, de Mendonca BB, de Castro M, Lofrano A, Hall JE, Bolu E, Ozata M, Quinton R, Amory JK, Stewart SE, Arlt W, Cole TR, Crowley WF, Kaiser UB, Latronico AC, Seminara SB. TAC3/TACR3 mutations reveal preferential activation of gonadotropin-releasing hormone release by neurokinin B in neonatal life followed by reversal in adulthood. J Clin Endocrinol Metab. 2010;95(6):2857-2867.
  94. Guran T, Tolhurst G, Bereket A, Rocha N, Porter K, Turan S, Gribble FM, Kotan LD, Akcay T, Atay Z, Canan H, Serin A, O'Rahilly S, Reimann F, Semple RK, Topaloglu AK. Hypogonadotropic hypogonadism due to a novel missense mutation in the first extracellular loop of the neurokinin B receptor. J Clin Endocrinol Metab. 2009;94(10):3633-3639.
  95. Young J, Bouligand J, Francou B, Raffin-Sanson ML, Gaillez S, Jeanpierre M, Grynberg M, Kamenicky P, Chanson P, Brailly-Tabard S, Guiochon-Mantel A. TAC3 and TACR3 defects cause hypothalamic congenital hypogonadotropic hypogonadism in humans. J Clin Endocrinol Metab. 2010;95(5):2287-2295.
  96. Young J, George JT, Tello JA, Francou B, Bouligand J, Guiochon-Mantel A, Brailly-Tabard S, Anderson RA, Millar RP. Kisspeptin restores pulsatile LH secretion in patients with neurokinin B signaling deficiencies: physiological, pathophysiological and therapeutic implications. Neuroendocrinology. 2013;97(2):193-202.
  97. Wilkes MM, Watkins WB, Stewart RD, Yen SS. Localization and quantitation of beta-endorphin in human brain and pituitary. Neuroendocrinology. 1980;30(2):113-121.
  98. Quigley ME, Yen SS. The role of endogenous opiates in LH secretion during the menstrual cycle. J Clin Endocrinol Metab. 1980;51(1):179-181.
  99. Veldhuis JD, Rogol AD, Samojlik E, Ertel NH. Role of endogenous opiates in the expression of negative feedback actions of androgen and estrogen on pulsatile properties of luteinizing hormone secretion in man. J Clin Invest. 1984;74(1):47-55.
  100. Boukhliq R, Goodman RL, Berriman SJ, Adrian B, Lehman MN. A subset of gonadotropin-releasing hormone neurons in the ovine medial basal hypothalamus is activated during increased pulsatile luteinizing hormone secretion. Endocrinology. 1999;140(12):5929-5936.
  101. Genazzani AD, Gastaldi M, Petraglia F, Battaglia C, Surico N, Volpe A, Genazzani AR. Naltrexone administration modulates the neuroendocrine control of luteinizing hormone secretion in hypothalamic amenorrhoea. Hum Reprod. 1995;10(11):2868-2871.
  102. Goodman RL, Coolen LM, Anderson GM, Hardy SL, Valent M, Connors JM, Fitzgerald ME, Lehman MN. Evidence that dynorphin plays a major role in mediating progesterone negative feedback on gonadotropin-releasing hormone neurons in sheep. Endocrinology. 2004;145(6):2959-2967.
  103. Shoupe D, Montz FJ, Lobo RA. The effects of estrogen and progestin on endogenous opioid activity in oophorectomized women. J Clin Endocrinol Metab. 1985;60(1):178-183.
  104. Walsh JP, Clarke IJ. Effects of central administration of highly selective opioid mu-, delta- and kappa-receptor agonists on plasma luteinizing hormone (LH), prolactin, and the estrogen-induced LH surge in ovariectomized ewes. Endocrinology. 1996;137(9):3640-3648.
  105. Cheng G, Coolen LM, Padmanabhan V, Goodman RL, Lehman MN. The kisspeptin/neurokinin B/dynorphin (KNDy) cell population of the arcuate nucleus: sex differences and effects of prenatal testosterone in sheep. Endocrinology. 2010;151(1):301-311.
  106. Goodman RL, Lehman MN, Smith JT, Coolen LM, de Oliveira CV, Jafarzadehshirazi MR, Pereira A, Iqbal J, Caraty A, Ciofi P, Clarke IJ. Kisspeptin neurons in the arcuate nucleus of the ewe express both dynorphin A and neurokinin B. Endocrinology. 2007;148(12):5752-5760.
  107. Burke MC, Letts PA, Krajewski SJ, Rance NE. Coexpression of dynorphin and neurokinin B immunoreactivity in the rat hypothalamus: Morphologic evidence of interrelated function within the arcuate nucleus. J Comp Neurol. 2006;498(5):712-726.
  108. Hrabovszky E, Ciofi P, Vida B, Horvath MC, Keller E, Caraty A, Bloom SR, Ghatei MA, Dhillo WS, Liposits Z, Kallo I. The kisspeptin system of the human hypothalamus: sexual dimorphism and relationship with gonadotropin-releasing hormone and neurokinin B neurons. Eur J Neurosci. 2010;31(11):1984-1998.
  109. Bedenbaugh MN, O'Connell RC, Lopez JA, McCosh RB, Goodman RL, Hileman SM. Kisspeptin, gonadotrophin-releasing hormone and oestrogen receptor alpha colocalise with neuronal nitric oxide synthase neurones in prepubertal female sheep. J Neuroendocrinol. 2018;30(1).
  110. Dufourny L, Delmas O, Teixeira-Gomes AP, Decourt C, Sliwowska JH. Neuroanatomical connections between kisspeptin neurones and somatostatin neurones in female and male rat hypothalamus: a possible involvement of SSTR1 in kisspeptin release. J Neuroendocrinol. 2018:e12593.
  111. Rometo AM, Krajewski SJ, Voytko ML, Rance NE. Hypertrophy and increased kisspeptin gene expression in the hypothalamic infundibular nucleus of postmenopausal women and ovariectomized monkeys. J Clin Endocrinol Metab. 2007;92(7):2744-2750.
  112. Clarkson J, Herbison AE. Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons. Endocrinology. 2006;147(12):5817-5825.
  113. Pompolo S, Pereira A, Estrada KM, Clarke IJ. Colocalization of kisspeptin and gonadotropin-releasing hormone in the ovine brain. Endocrinology. 2006;147(2):804-810.
  114. Clarkson J, d'Anglemont de Tassigny X, Colledge WH, Caraty A, Herbison AE. Distribution of kisspeptin neurones in the adult female mouse brain. J Neuroendocrinol. 2009;21(8):673-682.
  115. Oakley AE, Clifton DK, Steiner RA. Kisspeptin signaling in the brain. Endocr Rev. 2009;30(6):713-743.
  116. Uenoyama Y, Inoue N, Pheng V, Homma T, Takase K, Yamada S, Ajiki K, Ichikawa M, Okamura H, Maeda KI, Tsukamura H. Ultrastructural evidence of kisspeptin-gonadotrophin-releasing hormone (GnRH) interaction in the median eminence of female rats: implication of axo-axonal regulation of GnRH release. J Neuroendocrinol. 2011;23(10):863-870.
  117. Han SK, Gottsch ML, Lee KJ, Popa SM, Smith JT, Jakawich SK, Clifton DK, Steiner RA, Herbison AE. Activation of gonadotropin-releasing hormone neurons by kisspeptin as a neuroendocrine switch for the onset of puberty. J Neurosci. 2005;25(49):11349-11356.
  118. Irwig MS, Fraley GS, Smith JT, Acohido BV, Popa SM, Cunningham MJ, Gottsch ML, Clifton DK, Steiner RA. Kisspeptin activation of gonadotropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat. Neuroendocrinology. 2004;80(4):264-272.
  119. Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D, Dixon J, Thresher RR, Malinge I, Lomet D, Carlton MB, Colledge WH, Caraty A, Aparicio SA. Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. Proc Natl Acad Sci U S A. 2005;102(5):1761-1766.
  120. Lehman MN, Coolen LM, Goodman RL. Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion. Endocrinology. 2010;151(8):3479-3489.
  121. Mills EGA, Dhillo WS, Comninos AN. Kisspeptin and the control of emotions, mood and reproductive behaviour. J Endocrinol. 2018;239(1):R1-R12.
  122. Cao Y, Li Z, Jiang W, Ling Y, Kuang H. Reproductive functions of Kisspeptin/KISS1R Systems in the Periphery. Reprod Biol Endocrinol. 2019;17(1):65.
  123. Hu KL, Chang HM, Zhao HC, Yu Y, Li R, Qiao J. Potential roles for the kisspeptin/kisspeptin receptor system in implantation and placentation. Hum Reprod Update. 2019;25(3):326-343.
  124. Bhattacharya M, Babwah AV. Kisspeptin: beyond the brain. Endocrinology. 2015;156(4):1218-1227.
  125. Guzman S, Dragan M, Kwon H, de Oliveira V, Rao S, Bhatt V, Kalemba KM, Shah A, Rustgi VK, Wang H, Bech PR, Abbara A, Izzi-Engbeaya C, Manousou P, Guo JY, Guo GL, Radovick S, Dhillo WS, Wondisford FE, Babwah AV, Bhattacharya M. Targeting hepatic kisspeptin receptor ameliorates nonalcoholic fatty liver disease in a mouse model. J Clin Invest. 2022;132(10).
  126. Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev. 2012;92(3):1235-1316.
  127. Ikegami K, Minabe S, Ieda N, Goto T, Sugimoto A, Nakamura S, Inoue N, Oishi S, Maturana AD, Sanbo M, Hirabayashi M, Maeda KI, Tsukamura H, Uenoyama Y. Evidence of involvement of neurone-glia/neurone-neurone communications via gap junctions in synchronised activity of KNDy neurones. J Neuroendocrinol. 2017;29(6).
  128. Topaloglu AK, Semple RK. Neurokinin B signalling in the human reproductive axis. Mol Cell Endocrinol. 2011;346(1-2):57-64.
  129. Navarro VM, Bosch MA, Leon S, Simavli S, True C, Pinilla L, Carroll RS, Seminara SB, Tena-Sempere M, Ronnekleiv OK, Kaiser UB. The integrated hypothalamic tachykinin-kisspeptin system as a central coordinator for reproduction. Endocrinology. 2015;156(2):627-637.
  130. Ikegami K, Watanabe Y, Nakamura S, Goto T, Inoue N, Uenoyama Y, Tsukamura H. Cellular and molecular mechanisms regulating the KNDy neuronal activities to generate and modulate GnRH pulse in mammals. Front Neuroendocrinol. 2022;64:100968.
  131. Skorupskaite K, George JT, Veldhuis JD, Anderson RA. Neurokinin B Regulates Gonadotropin Secretion, Ovarian Follicle Growth, and the Timing of Ovulation in Healthy Women. J Clin Endocrinol Metab. 2018;103(1):95-104.
  132. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Interactions Between Neurokinin B and Kisspeptin in Mediating Estrogen Feedback in Healthy Women. J Clin Endocrinol Metab. 2016;101(12):4628-4636.
  133. Skorupskaite K, George J, Anderson RA. Role of a neurokinin B receptor antagonist in the regulation of ovarian function in healthy women. Lancet. 2015;385 Suppl 1:S92.
  134. Skorupskaite K, Anderson RA. Hypothalamic neurokinin signalling and its application in reproductive medicine. Pharmacol Ther. 2021:107960.
  135. Akad M, Socolov R, Furnica C, Covali R, Stan CD, Crauciuc E, Pavaleanu I. Kisspeptin Variations in Patients with Polycystic Ovary Syndrome-A Prospective Case Control Study. Medicina (Kaunas). 2022;58(6).
  136. de Assis Rodrigues NP, Lagana AS, Zaia V, Vitagliano A, Barbosa CP, de Oliveira R, Trevisan CM, Montagna E. The role of Kisspeptin levels in polycystic ovary syndrome: a systematic review and meta-analysis. Arch Gynecol Obstet. 2019;300(5):1423-1434.
  137. Zhao T, Zhang Q, Xiao X, Tao X, Gao M, He W, Wu X, Yuan T. Associations of the KiSS-1 and GPR54 genetic polymorphism with polycystic ovary syndrome in Yunnan, China. Gynecol Endocrinol. 2022;38(9):790-794.
  138. Zhao T, Xiao X, Li L, Tao X, He W, Zhang Q, Wu X, Yuan T. Role of kisspeptin in polycystic ovarian syndrome: A metabolomics study. Clin Endocrinol (Oxf). 2023;99(3):315-325.
  139. George JT, Kakkar R, Marshall J, Scott ML, Finkelman RD, Ho TW, Veldhuis J, Skorupskaite K, Anderson RA, McIntosh S, Webber L. Neurokinin B Receptor Antagonism in Women With Polycystic Ovary Syndrome: A Randomized, Placebo-Controlled Trial. J Clin Endocrinol Metab. 2016;101(11):4313-4321.
  140. Fraser GL, Obermayer-Pietsch B, Laven J, Griesinger G, Pintiaux A, Timmerman D, Fauser B, Lademacher C, Combalbert J, Hoveyda HR, Ramael S. Randomized Controlled Trial of Neurokinin 3 Receptor Antagonist Fezolinetant for Treatment of Polycystic Ovary Syndrome. J Clin Endocrinol Metab. 2021.
  141. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Kisspeptin and neurokinin B interactions in modulating gonadotropin secretion in women with polycystic ovary syndrome. Hum Reprod. 2020;35(6):1421-1431.
  142. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Neurokinin 3 Receptor Antagonism Reveals Roles for Neurokinin B in the Regulation of Gonadotropin Secretion and Hot Flashes in Postmenopausal Women. Neuroendocrinology. 2018;106(2):148-157.
  143. Prague JK, Roberts RE, Comninos AN, Clarke S, Jayasena CN, Nash Z, Doyle C, Papadopoulou DA, Bloom SR, Mohideen P, Panay N, Hunter MS, Veldhuis JD, Webber LC, Huson L, Dhillo WS. Neurokinin 3 receptor antagonism as a novel treatment for menopausal hot flushes: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet. 2017;389(10081):1809-1820.
  144. Depypere H, Timmerman D, Donders G, Sieprath P, Ramael S, Combalbert J, Hoveyda HR, Fraser GL. Treatment of Menopausal Vasomotor Symptoms With Fezolinetant, a Neurokinin 3 Receptor Antagonist: A Phase 2a Trial. J Clin Endocrinol Metab. 2019;104(12):5893-5905.
  145. Prague JK, Dhillo WS. Treating hot flushes with a neurokinin 3 receptor antagonist. Oncotarget. 2017;8(63):106153-106154.
  146. Prague JK, Dhillo WS. Neurokinin 3 receptor antagonism - the magic bullet for hot flushes? Climacteric. 2017;20(6):505-509.
  147. Anderson RA, Skorupskaite K, Sassarini J. The neurokinin B pathway in the treatment of menopausal hot flushes. Climacteric. 2019;22(1):51-54.
  148. Johnson KA, Martin N, Nappi RE, Neal-Perry G, Shapiro M, Stute P, Thurston RC, Wolfman W, English M, Franklin C, Lee M, Santoro N. Efficacy and Safety of Fezolinetant in Moderate to Severe Vasomotor Symptoms Associated With Menopause: A Phase 3 RCT. J Clin Endocrinol Metab. 2023;108(8):1981-1997.
  149. Pinkerton JV, Simon JA, Joffe H, Maki PM, Nappi RE, Panay N, Soares CN, Thurston RC, Caetano C, Haberland C, Haseli Mashhadi N, Krahn U, Mellinger U, Parke S, Seitz C, Zuurman L. Elinzanetant for the Treatment of Vasomotor Symptoms Associated With Menopause: OASIS 1 and 2 Randomized Clinical Trials. JAMA. 2024.
  150. Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Neurokinin 3 receptor antagonism decreases gonadotropin and testosterone secretion in healthy men. Clin Endocrinol (Oxf). 2017;87(6):748-756.
  151. Navarro VM, Castellano JM, McConkey SM, Pineda R, Ruiz-Pino F, Pinilla L, Clifton DK, Tena-Sempere M, Steiner RA. Interactions between kisspeptin and neurokinin B in the control of GnRH secretion in the female rat. Am J Physiol Endocrinol Metab. 2011;300(1):E202-210.
  152. Garcia-Galiano D, van Ingen Schenau D, Leon S, Krajnc-Franken MA, Manfredi-Lozano M, Romero-Ruiz A, Navarro VM, Gaytan F, van Noort PI, Pinilla L, Blomenrohr M, Tena-Sempere M. Kisspeptin signaling is indispensable for neurokinin B, but not glutamate, stimulation of gonadotropin secretion in mice. Endocrinology. 2012;153(1):316-328.
  153. Wakabayashi Y, Nakada T, Murata K, Ohkura S, Mogi K, Navarro VM, Clifton DK, Mori Y, Tsukamura H, Maeda K, Steiner RA, Okamura H. Neurokinin B and dynorphin A in kisspeptin neurons of the arcuate nucleus participate in generation of periodic oscillation of neural activity driving pulsatile gonadotropin-releasing hormone secretion in the goat. J Neurosci. 2010;30(8):3124-3132.
  154. Ramaswamy S, Seminara SB, Ali B, Ciofi P, Amin NA, Plant TM. Neurokinin B stimulates GnRH release in the male monkey (Macaca mulatta) and is colocalized with kisspeptin in the arcuate nucleus. Endocrinology. 2010;151(9):4494-4503.
  155. Ramaswamy S, Seminara SB, Plant TM. Evidence from the agonadal juvenile male rhesus monkey (Macaca mulatta) for the view that the action of neurokinin B to trigger gonadotropin-releasing hormone release is upstream from the kisspeptin receptor. Neuroendocrinology. 2011;94(3):237-245.
  156. Navarro VM. Interactions between kisspeptins and neurokinin B. Adv Exp Med Biol. 2013;784:325-347.
  157. Navarro VM, Gottsch ML, Chavkin C, Okamura H, Clifton DK, Steiner RA. Regulation of gonadotropin-releasing hormone secretion by kisspeptin/dynorphin/neurokinin B neurons in the arcuate nucleus of the mouse. J Neurosci. 2009;29(38):11859-11866.
  158. Ciofi P, Krause JE, Prins GS, Mazzuca M. Presence of nuclear androgen receptor-like immunoreactivity in neurokinin B-containing neurons of the hypothalamic arcuate nucleus of the adult male rat. Neurosci Lett. 1994;182(2):193-196.
  159. Foradori CD, Coolen LM, Fitzgerald ME, Skinner DC, Goodman RL, Lehman MN. Colocalization of progesterone receptors in parvicellular dynorphin neurons of the ovine preoptic area and hypothalamus. Endocrinology. 2002;143(11):4366-4374.
  160. Smith JT, Cunningham MJ, Rissman EF, Clifton DK, Steiner RA. Regulation of Kiss1 gene expression in the brain of the female mouse. Endocrinology. 2005;146(9):3686-3692.
  161. Smith JT, Acohido BV, Clifton DK, Steiner RA. KiSS-1 neurones are direct targets for leptin in the ob/ob mouse. J Neuroendocrinol. 2006;18(4):298-303.
  162. George JT, Seminara SB. Kisspeptin and the hypothalamic control of reproduction: lessons from the human. Endocrinology. 2012;153(11):5130-5136.
  163. Arreguin-Arevalo JA, Lents CA, Farmerie TA, Nett TM, Clay CM. KiSS-1 peptide induces release of LH by a direct effect on the hypothalamus of ovariectomized ewes. Anim Reprod Sci. 2007;101(3-4):265-275.
  164. Gottsch ML, Cunningham MJ, Smith JT, Popa SM, Acohido BV, Crowley WF, Seminara S, Clifton DK, Steiner RA. A role for kisspeptins in the regulation of gonadotropin secretion in the mouse. Endocrinology. 2004;145(9):4073-4077.
  165. Shahab M, Mastronardi C, Seminara SB, Crowley WF, Ojeda SR, Plant TM. Increased hypothalamic GPR54 signaling: a potential mechanism for initiation of puberty in primates. Proc Natl Acad Sci U S A. 2005;102(6):2129-2134.
  166. Smith JT, Li Q, Pereira A, Clarke IJ. Kisspeptin neurons in the ovine arcuate nucleus and preoptic area are involved in the preovulatory luteinizing hormone surge. Endocrinology. 2009;150(12):5530-5538.
  167. Chan YM, Butler JP, Pinnell NE, Pralong FP, Crowley WF, Jr., Ren C, Chan KK, Seminara SB. Kisspeptin resets the hypothalamic GnRH clock in men. J Clin Endocrinol Metab. 2011;96(6):E908-915.
  168. Chan YM, Butler JP, Sidhoum VF, Pinnell NE, Seminara SB. Kisspeptin administration to women: a window into endogenous kisspeptin secretion and GnRH responsiveness across the menstrual cycle. J Clin Endocrinol Metab. 2012;97(8):E1458-1467.
  169. Dhillo WS, Chaudhri OB, Patterson M, Thompson EL, Murphy KG, Badman MK, McGowan BM, Amber V, Patel S, Ghatei MA, Bloom SR. Kisspeptin-54 stimulates the hypothalamic-pituitary gonadal axis in human males. J Clin Endocrinol Metab. 2005;90(12):6609-6615.
  170. Dhillo WS, Chaudhri OB, Thompson EL, Murphy KG, Patterson M, Ramachandran R, Nijher GK, Amber V, Kokkinos A, Donaldson M, Ghatei MA, Bloom SR. Kisspeptin-54 stimulates gonadotropin release most potently during the preovulatory phase of the menstrual cycle in women. J Clin Endocrinol Metab. 2007;92(10):3958-3966.
  171. George JT, Anderson RA, Millar RP. Kisspeptin-10 stimulation of gonadotrophin secretion in women is modulated by sex steroid feedback. Hum Reprod. 2012;27(12):3552-3559.
  172. George JT, Veldhuis JD, Roseweir AK, Newton CL, Faccenda E, Millar RP, Anderson RA. Kisspeptin-10 is a potent stimulator of LH and increases pulse frequency in men. J Clin Endocrinol Metab. 2011;96(8):E1228-1236.
  173. Jayasena CN, Comninos AN, Nijher GM, Abbara A, De Silva A, Veldhuis JD, Ratnasabapathy R, Izzi-Engbeaya C, Lim A, Patel DA, Ghatei MA, Bloom SR, Dhillo WS. Twice-daily subcutaneous injection of kisspeptin-54 does not abolish menstrual cyclicity in healthy female volunteers. J Clin Endocrinol Metab. 2013;98(11):4464-4474.
  174. Jayasena CN, Comninos AN, Veldhuis JD, Misra S, Abbara A, Izzi-Engbeaya C, Donaldson M, Ghatei MA, Bloom SR, Dhillo WS. A single injection of kisspeptin-54 temporarily increases luteinizing hormone pulsatility in healthy women. Clin Endocrinol (Oxf). 2013;79(4):558-563.
  175. Jayasena CN, Nijher GM, Abbara A, Murphy KG, Lim A, Patel D, Mehta A, Todd C, Donaldson M, Trew GH, Ghatei MA, Bloom SR, Dhillo WS. Twice-weekly administration of kisspeptin-54 for 8 weeks stimulates release of reproductive hormones in women with hypothalamic amenorrhea. Clin Pharmacol Ther. 2010;88(6):840-847.
  176. Jayasena CN, Nijher GM, Chaudhri OB, Murphy KG, Ranger A, Lim A, Patel D, Mehta A, Todd C, Ramachandran R, Salem V, Stamp GW, Donaldson M, Ghatei MA, Bloom SR, Dhillo WS. Subcutaneous injection of kisspeptin-54 acutely stimulates gonadotropin secretion in women with hypothalamic amenorrhea, but chronic administration causes tachyphylaxis. J Clin Endocrinol Metab. 2009;94(11):4315-4323.
  177. Jayasena CN, Nijher GM, Comninos AN, Abbara A, Januszewki A, Vaal ML, Sriskandarajah L, Murphy KG, Farzad Z, Ghatei MA, Bloom SR, Dhillo WS. The effects of kisspeptin-10 on reproductive hormone release show sexual dimorphism in humans. J Clin Endocrinol Metab. 2011;96(12):E1963-1972.
  178. Abbara A, Narayanaswamy S, Izzi-Engbeaya C, Comninos AN, Clarke SA, Malik Z, Papadopoulou D, Clobentz A, Sarang Z, Bassett P, Jayasena CN, Dhillo WS. Hypothalamic Response to Kisspeptin-54 and Pituitary Response to Gonadotropin-Releasing Hormone Are Preserved in Healthy Older Men. Neuroendocrinology. 2018;106(4):401-410.
  179. George JT, Veldhuis JD, Tena-Sempere M, Millar RP, Anderson RA. Exploring the pathophysiology of hypogonadism in men with type 2 diabetes: kisspeptin-10 stimulates serum testosterone and LH secretion in men with type 2 diabetes and mild biochemical hypogonadism. Clin Endocrinol (Oxf). 2013;79(1):100-104.
  180. Jayasena CN, Abbara A, Veldhuis JD, Comninos AN, Ratnasabapathy R, De Silva A, Nijher GM, Ganiyu-Dada Z, Mehta A, Todd C, Ghatei MA, Bloom SR, Dhillo WS. Increasing LH pulsatility in women with hypothalamic amenorrhoea using intravenous infusion of Kisspeptin-54. J Clin Endocrinol Metab. 2014;99(6):E953-961.
  181. Abbara A, Eng PC, Phylactou M, Clarke SA, Richardson R, Sykes CM, Phumsatitpong C, Mills E, Modi M, Izzi-Engbeaya C, Papadopoulou D, Purugganan K, Jayasena CN, Webber L, Salim R, Owen B, Bech P, Comninos AN, McArdle CA, Voliotis M, Tsaneva-Atanasova K, Moenter S, Hanyaloglu A, Dhillo WS. Kisspeptin receptor agonist has therapeutic potential for female reproductive disorders. J Clin Invest. 2020;130(12):6739-6753.
  182. Szeliga A, Podfigurna A, Bala G, Meczekalski B. Kisspeptin and neurokinin B analogs use in gynecological endocrinology: where do we stand? J Endocrinol Invest. 2020;43(5):555-561.
  183. Abbara A, Ufer M, Voors-Pette C, Berman L, Ezzati M, Wu R, Lee TY, Ferreira JCA, Migoya E, Dhillo WS. Endocrine profile of the kisspeptin receptor agonist MVT-602 in healthy premenopausal women with and without ovarian stimulation: results from 2 randomized, placebo-controlled clinical tricals. Fertil Steril. 2024;121(1):95-106.
  184. Li XF, Kinsey-Jones JS, Cheng Y, Knox AM, Lin Y, Petrou NA, Roseweir A, Lightman SL, Milligan SR, Millar RP, O'Byrne KT. Kisspeptin signalling in the hypothalamic arcuate nucleus regulates GnRH pulse generator frequency in the rat. PLoS One. 2009;4(12):e8334.
  185. Millar RP, Roseweir AK, Tello JA, Anderson RA, George JT, Morgan K, Pawson AJ. Kisspeptin antagonists: unraveling the role of kisspeptin in reproductive physiology. Brain Res. 2010;1364:81-89.
  186. Roseweir AK, Kauffman AS, Smith JT, Guerriero KA, Morgan K, Pielecka-Fortuna J, Pineda R, Gottsch ML, Tena-Sempere M, Moenter SM, Terasawa E, Clarke IJ, Steiner RA, Millar RP. Discovery of potent kisspeptin antagonists delineate physiological mechanisms of gonadotropin regulation. J Neurosci. 2009;29(12):3920-3929.
  187. Thompson EL, Patterson M, Murphy KG, Smith KL, Dhillo WS, Todd JF, Ghatei MA, Bloom SR. Central and peripheral administration of kisspeptin-10 stimulates the hypothalamic-pituitary-gonadal axis. J Neuroendocrinol. 2004;16(10):850-858.
  188. Tovar S, Vazquez MJ, Navarro VM, Fernandez-Fernandez R, Castellano JM, Vigo E, Roa J, Casanueva FF, Aguilar E, Pinilla L, Dieguez C, Tena-Sempere M. Effects of single or repeated intravenous administration of kisspeptin upon dynamic LH secretion in conscious male rats. Endocrinology. 2006;147(6):2696-2704.
  189. Novaira HJ, Ng Y, Wolfe A, Radovick S. Kisspeptin increases GnRH mRNA expression and secretion in GnRH secreting neuronal cell lines. Mol Cell Endocrinol. 2009;311(1-2):126-134.
  190. Chan YM, Lippincott MF, Butler JP, Sidhoum VF, Li CX, Plummer L, Seminara SB. Exogenous kisspeptin administration as a probe of GnRH neuronal function in patients with idiopathic hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2014;99(12):E2762-2771.
  191. Mittelman-Smith MA, Krajewski-Hall SJ, McMullen NT, Rance NE. Ablation of KNDy Neurons Results in Hypogonadotropic Hypogonadism and Amplifies the Steroid-Induced LH Surge in Female Rats. Endocrinology. 2016;157(5):2015-2027.
  192. Gutierrez-Pascual E, Martinez-Fuentes AJ, Pinilla L, Tena-Sempere M, Malagon MM, Castano JP. Direct pituitary effects of kisspeptin: activation of gonadotrophs and somatotrophs and stimulation of luteinising hormone and growth hormone secretion. J Neuroendocrinol. 2007;19(7):521-530.
  193. Richard N, Galmiche G, Corvaisier S, Caraty A, Kottler ML. KiSS-1 and GPR54 genes are co-expressed in rat gonadotrophs and differentially regulated in vivo by oestradiol and gonadotrophin-releasing hormone. J Neuroendocrinol. 2008;20(3):381-393.
  194. Smith JT, Coolen LM, Kriegsfeld LJ, Sari IP, Jaafarzadehshirazi MR, Maltby M, Bateman K, Goodman RL, Tilbrook AJ, Ubuka T, Bentley GE, Clarke IJ, Lehman MN. Variation in kisspeptin and RFamide-related peptide (RFRP) expression and terminal connections to gonadotropin-releasing hormone neurons in the brain: a novel medium for seasonal breeding in the sheep. Endocrinology. 2008;149(11):5770-5782.
  195. Smith JT, Rao A, Pereira A, Caraty A, Millar RP, Clarke IJ. Kisspeptin is present in ovine hypophysial portal blood but does not increase during the preovulatory luteinizing hormone surge: evidence that gonadotropes are not direct targets of kisspeptin in vivo. Endocrinology. 2008;149(4):1951-1959.
  196. Witham EA, Meadows JD, Hoffmann HM, Shojaei S, Coss D, Kauffman AS, Mellon PL. Kisspeptin regulates gonadotropin genes via immediate early gene induction in pituitary gonadotropes. Mol Endocrinol. 2013;27(8):1283-1294.
  197. Pampillo M, Camuso N, Taylor JE, Szereszewski JM, Ahow MR, Zajac M, Millar RP, Bhattacharya M, Babwah AV. Regulation of GPR54 signaling by GRK2 and {beta}-arrestin. Mol Endocrinol. 2009;23(12):2060-2074.
  198. Ramaswamy S, Seminara SB, Pohl CR, DiPietro MJ, Crowley WF, Jr., Plant TM. Effect of continuous intravenous administration of human metastin 45-54 on the neuroendocrine activity of the hypothalamic-pituitary-testicular axis in the adult male rhesus monkey (Macaca mulatta). Endocrinology. 2007;148(7):3364-3370.
  199. Seminara SB, Dipietro MJ, Ramaswamy S, Crowley WF, Jr., Plant TM. Continuous human metastin 45-54 infusion desensitizes G protein-coupled receptor 54-induced gonadotropin-releasing hormone release monitored indirectly in the juvenile male Rhesus monkey (Macaca mulatta): a finding with therapeutic implications. Endocrinology. 2006;147(5):2122-2126.
  200. Thompson EL, Murphy KG, Patterson M, Bewick GA, Stamp GW, Curtis AE, Cooke JH, Jethwa PH, Todd JF, Ghatei MA, Bloom SR. Chronic subcutaneous administration of kisspeptin-54 causes testicular degeneration in adult male rats. Am J Physiol Endocrinol Metab. 2006;291(5):E1074-1082.
  201. Lippincott MFS, V.F.; Seminara, S. Continuously administered kisspeptin as a pseudo-antagonist of the kisspeptin receptor. The Endocrine Society´s 95th Annual Meeting, ENDO 2013, San Francisco, Abstract SAT 2134-2163. 2013.
  202. MacLean DB, Matsui H, Suri A, Neuwirth R, Colombel M. Sustained exposure to the investigational Kisspeptin analog, TAK-448, down-regulates testosterone into the castration range in healthy males and in patients with prostate cancer: results from two phase 1 studies. J Clin Endocrinol Metab. 2014;99(8):E1445-1453.
  203. Scott G, Ahmad I, Howard K, MacLean D, Oliva C, Warrington S, Wilbraham D, Worthington P. Double-blind, randomized, placebo-controlled study of safety, tolerability, pharmacokinetics and pharmacodynamics of TAK-683, an investigational metastin analogue in healthy men. Br J Clin Pharmacol. 2013;75(2):381-391.
  204. Christian CA, Moenter SM. The neurobiology of preovulatory and estradiol-induced gonadotropin-releasing hormone surges. Endocr Rev. 2010;31(4):544-577.
  205. Kauffman AS. Coming of age in the kisspeptin era: sex differences, development, and puberty. Mol Cell Endocrinol. 2010;324(1-2):51-63.
  206. Terasawa E, Garcia JP, Seminara SB, Keen KL. Role of Kisspeptin and Neurokinin B in Puberty in Female Non-Human Primates. Front Endocrinol (Lausanne). 2018;9:148.
  207. Shahab M, Lippincott M, Chan YM, Davies A, Merino PM, Plummer L, Mericq V, Seminara S. Discordance in the Dependence on Kisspeptin Signaling in Mini Puberty vs Adolescent Puberty: Human Genetic Evidence. J Clin Endocrinol Metab. 2018;103(4):1273-1276.
  208. Navarro VM, Castellano JM, Fernandez-Fernandez R, Barreiro ML, Roa J, Sanchez-Criado JE, Aguilar E, Dieguez C, Pinilla L, Tena-Sempere M. Developmental and hormonally regulated messenger ribonucleic acid expression of KiSS-1 and its putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-releasing activity of KiSS-1 peptide. Endocrinology. 2004;145(10):4565-4574.
  209. Keen KL, Wegner FH, Bloom SR, Ghatei MA, Terasawa E. An increase in kisspeptin-54 release occurs with the pubertal increase in luteinizing hormone-releasing hormone-1 release in the stalk-median eminence of female rhesus monkeys in vivo. Endocrinology. 2008;149(8):4151-4157.
  210. Plant TM, Ramaswamy S, Dipietro MJ. Repetitive activation of hypothalamic G protein-coupled receptor 54 with intravenous pulses of kisspeptin in the juvenile monkey (Macaca mulatta) elicits a sustained train of gonadotropin-releasing hormone discharges. Endocrinology. 2006;147(2):1007-1013.
  211. Pineda R, Garcia-Galiano D, Roseweir A, Romero M, Sanchez-Garrido MA, Ruiz-Pino F, Morgan K, Pinilla L, Millar RP, Tena-Sempere M. Critical roles of kisspeptins in female puberty and preovulatory gonadotropin surges as revealed by a novel antagonist. Endocrinology. 2010;151(2):722-730.
  212. Decourt C, Robert V, Anger K, Galibert M, Madinier JB, Liu X, Dardente H, Lomet D, Delmas AF, Caraty A, Herbison AE, Anderson GM, Aucagne V, Beltramo M. A synthetic kisspeptin analog that triggers ovulation and advances puberty. Sci Rep. 2016;6:26908.
  213. Novaira HJ, Sonko ML, Hoffman G, Koo Y, Ko C, Wolfe A, Radovick S. Disrupted kisspeptin signaling in GnRH neurons leads to hypogonadotrophic hypogonadism. Mol Endocrinol. 2014;28(2):225-238.
  214. Lippincott MF, Chan YM, Delaney A, Rivera-Morales D, Butler JP, Seminara SB. Kisspeptin Responsiveness Signals Emergence of Reproductive Endocrine Activity: Implications for Human Puberty. J Clin Endocrinol Metab. 2016;101(8):3061-3069.
  215. Chan YM, Lippincott MF, Kusa TO, Seminara SB. Divergent responses to kisspeptin in children with delayed puberty. JCI Insight. 2018;3(8).
  216. Sobrino V, Avendano MS, Perdices-Lopez C, Jimenez-Puyer M, Tena-Sempere M. Kisspeptins and the neuroendocrine control of reproduction: Recent progress and new frontiers in kisspeptin research. Front Neuroendocrinol. 2022;65:100977.
  217. Garcia JP, Guerriero KA, Keen KL, Kenealy BP, Seminara SB, Terasawa E. Kisspeptin and Neurokinin B Signaling Network Underlies the Pubertal Increase in GnRH Release in Female Rhesus Monkeys. Endocrinology. 2017;158(10):3269-3280.
  218. Midgley AR, Jr., Jaffe RB. Regulation of human gonadotropins. X. Episodic fluctuation of LH during the menstrual cycle. J Clin Endocrinol Metab. 1971;33(6):962-969.
  219. Levine JE, Bauer-Dantoin AC, Besecke LM, Conaghan LA, Legan SJ, Meredith JM, Strobl FJ, Urban JH, Vogelsong KM, Wolfe AM. Neuroendocrine regulation of the luteinizing hormone-releasing hormone pulse generator in the rat. Recent Prog Horm Res. 1991;47:97-151; discussion 151-153.
  220. Clarke IJ. Gonadotrophin-releasing hormone secretion (GnRH) in anoestrous ewes and the induction of GnRH surges by oestrogen. J Endocrinol. 1988;117(3):355-360.
  221. Pau KY, Berria M, Hess DL, Spies HG. Preovulatory gonadotropin-releasing hormone surge in ovarian-intact rhesus macaques. Endocrinology. 1993;133(4):1650-1656.
  222. Knobil E, Plant TM. The hypothalamic regulation of LH and FSH secretion in the rhesus monkey. Res Publ Assoc Res Nerv Ment Dis. 1978;56:359-372.
  223. Hall JE, Taylor AE, Martin KA, Rivier J, Schoenfeld DA, Crowley WF, Jr. Decreased release of gonadotropin-releasing hormone during the preovulatory midcycle luteinizing hormone surge in normal women. Proc Natl Acad Sci U S A. 1994;91(15):6894-6898.
  224. Bagatell CJ, Dahl KD, Bremner WJ. The direct pituitary effect of testosterone to inhibit gonadotropin secretion in men is partially mediated by aromatization to estradiol. J Androl. 1994;15(1):15-21.
  225. Emons G, Ortmann O, Thiessen S, Knuppen R. Effects of estradiol and some antiestrogens (clomiphene, tamoxifen, and hydroxytamoxifen) on luteinizing hormone secretion by rat pituitary cells in culture. Arch Gynecol. 1986;237(4):199-211.
  226. Finkelstein JS, O'Dea LS, Whitcomb RW, Crowley WF, Jr. Sex steroid control of gonadotropin secretion in the human male. II. Effects of estradiol administration in normal and gonadotropin-releasing hormone-deficient men. J Clin Endocrinol Metab. 1991;73(3):621-628.
  227. Hayes FJ, Seminara SB, Decruz S, Boepple PA, Crowley WF, Jr. Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. J Clin Endocrinol Metab. 2000;85(9):3027-3035.
  228. Kerin JF, Liu JH, Phillipou G, Yen SS. Evidence for a hypothalamic site of action of clomiphene citrate in women. J Clin Endocrinol Metab. 1985;61(2):265-268.
  229. Kesner JS, Wilson RC, Kaufman JM, Hotchkiss J, Chen Y, Yamamoto H, Pardo RR, Knobil E. Unexpected responses of the hypothalamic gonadotropin-releasing hormone "pulse generator" to physiological estradiol inputs in the absence of the ovary. Proc Natl Acad Sci U S A. 1987;84(23):8745-8749.
  230. Pitteloud N, Dwyer AA, DeCruz S, Lee H, Boepple PA, Crowley WF, Jr., Hayes FJ. Inhibition of luteinizing hormone secretion by testosterone in men requires aromatization for its pituitary but not its hypothalamic effects: evidence from the tandem study of normal and gonadotropin-releasing hormone-deficient men. J Clin Endocrinol Metab. 2008;93(3):784-791.
  231. Veldhuis JD, Evans WS, Rogol AD, Thorner MO, Stumpf P. Actions of estradiol on discrete attributes of the luteinizing hormone pulse signal in man. Studies in postmenopausal women treated with pure estradiol. J Clin Invest. 1987;79(3):769-776.
  232. Herbison AE. Multimodal influence of estrogen upon gonadotropin-releasing hormone neurons. Endocr Rev. 1998;19(3):302-330.
  233. McDevitt MA, Glidewell-Kenney C, Jimenez MA, Ahearn PC, Weiss J, Jameson JL, Levine JE. New insights into the classical and non-classical actions of estrogen: evidence from estrogen receptor knock-out and knock-in mice. Mol Cell Endocrinol. 2008;290(1-2):24-30.
  234. Rance NE, McMullen NT, Smialek JE, Price DL, Young WS, 3rd. Postmenopausal hypertrophy of neurons expressing the estrogen receptor gene in the human hypothalamus. J Clin Endocrinol Metab. 1990;71(1):79-85.
  235. Rance NE, Young WS, 3rd. Hypertrophy and increased gene expression of neurons containing neurokinin-B and substance-P messenger ribonucleic acids in the hypothalami of postmenopausal women. Endocrinology. 1991;128(5):2239-2247.
  236. Rometo AM, Rance NE. Changes in prodynorphin gene expression and neuronal morphology in the hypothalamus of postmenopausal women. J Neuroendocrinol. 2008;20(12):1376-1381.
  237. Franceschini I, Lomet D, Cateau M, Delsol G, Tillet Y, Caraty A. Kisspeptin immunoreactive cells of the ovine preoptic area and arcuate nucleus co-express estrogen receptor alpha. Neurosci Lett. 2006;401(3):225-230.
  238. Abel TW, Voytko ML, Rance NE. The effects of hormone replacement therapy on hypothalamic neuropeptide gene expression in a primate model of menopause. J Clin Endocrinol Metab. 1999;84(6):2111-2118.
  239. Eghlidi DH, Haley GE, Noriega NC, Kohama SG, Urbanski HF. Influence of age and 17beta-estradiol on kisspeptin, neurokinin B, and prodynorphin gene expression in the arcuate-median eminence of female rhesus macaques. Endocrinology. 2010;151(8):3783-3794.
  240. Goodman RL, Holaskova I, Nestor CC, Connors JM, Billings HJ, Valent M, Lehman MN, Hileman SM. Evidence that the arcuate nucleus is an important site of progesterone negative feedback in the ewe. Endocrinology. 2011;152(9):3451-3460.
  241. Jayasena CN, Abbara A, Comninos AN, Nijher GM, Christopoulos G, Narayanaswamy S, Izzi-Engbeaya C, Sridharan M, Mason AJ, Warwick J, Ashby D, Ghatei MA, Bloom SR, Carby A, Trew GH, Dhillo WS. Kisspeptin-54 triggers egg maturation in women undergoing in vitro fertilization. J Clin Invest. 2014;124(8):3667-3677.
  242. Sandoval-Guzman T, Rance NE. Central injection of senktide, an NK3 receptor agonist, or neuropeptide Y inhibits LH secretion and induces different patterns of Fos expression in the rat hypothalamus. Brain Res. 2004;1026(2):307-312.
  243. Lippincott MF, Chan YM, Rivera Morales D, Seminara SB. Continuous Kisspeptin Administration in Postmenopausal Women: Impact of Estradiol on Luteinizing Hormone Secretion. J Clin Endocrinol Metab. 2017;102(6):2091-2099.
  244. Clarkson J, d'Anglemont de Tassigny X, Moreno AS, Colledge WH, Herbison AE. Kisspeptin-GPR54 signaling is essential for preovulatory gonadotropin-releasing hormone neuron activation and the luteinizing hormone surge. J Neurosci. 2008;28(35):8691-8697.
  245. Kinoshita M, Tsukamura H, Adachi S, Matsui H, Uenoyama Y, Iwata K, Yamada S, Inoue K, Ohtaki T, Matsumoto H, Maeda K. Involvement of central metastin in the regulation of preovulatory luteinizing hormone surge and estrous cyclicity in female rats. Endocrinology. 2005;146(10):4431-4436.
  246. Watson RE, Jr., Langub MC, Jr., Engle MG, Maley BE. Estrogen-receptive neurons in the anteroventral periventricular nucleus are synaptic targets of the suprachiasmatic nucleus and peri-suprachiasmatic region. Brain Res. 1995;689(2):254-264.
  247. Kumar D, Candlish M, Periasamy V, Avcu N, Mayer C, Boehm U. Specialized subpopulations of kisspeptin neurons communicate with GnRH neurons in female mice. Endocrinology. 2015;156(1):32-38.
  248. Billings HJ, Connors JM, Altman SN, Hileman SM, Holaskova I, Lehman MN, McManus CJ, Nestor CC, Jacobs BH, Goodman RL. Neurokinin B acts via the neurokinin-3 receptor in the retrochiasmatic area to stimulate luteinizing hormone secretion in sheep. Endocrinology. 2010;151(8):3836-3846.
  249. Li Q, Millar RP, Clarke IJ, Smith JT. Evidence that Neurokinin B Controls Basal Gonadotropin-Releasing Hormone Secretion but Is Not Critical for Estrogen-Positive Feedback in Sheep. Neuroendocrinology. 2015;101(2):161-174.
  250. Terasawa E. Neuroestradiol in regulation of GnRH release. Horm Behav. 2018.
  251. Soules MR, Steiner RA, Clifton DK, Cohen NL, Aksel S, Bremner WJ. Progesterone modulation of pulsatile luteinizing hormone secretion in normal women. J Clin Endocrinol Metab. 1984;58(2):378-383.
  252. Skinner DC, Evans NP, Delaleu B, Goodman RL, Bouchard P, Caraty A. The negative feedback actions of progesterone on gonadotropin-releasing hormone secretion are transduced by the classical progesterone receptor. Proc Natl Acad Sci U S A. 1998;95(18):10978-10983.
  253. Fox SR, Harlan RE, Shivers BD, Pfaff DW. Chemical characterization of neuroendocrine targets for progesterone in the female rat brain and pituitary. Neuroendocrinology. 1990;51(3):276-283.
  254. King JC, Tai DW, Hanna IK, Pfeiffer A, Haas P, Ronsheim PM, Mitchell SC, Turcotte JC, Blaustein JD. A subgroup of LHRH neurons in guinea pigs with progestin receptors is centrally positioned within the total population of LHRH neurons. Neuroendocrinology. 1995;61(3):265-275.
  255. Skinner DC, Caraty A, Allingham R. Unmasking the progesterone receptor in the preoptic area and hypothalamus of the ewe: no colocalization with gonadotropin-releasing neurons. Endocrinology. 2001;142(2):573-579.
  256. Foradori CD, Goodman RL, Adams VL, Valent M, Lehman MN. Progesterone increases dynorphin a concentrations in cerebrospinal fluid and preprodynorphin messenger ribonucleic Acid levels in a subset of dynorphin neurons in the sheep. Endocrinology. 2005;146(4):1835-1842.
  257. Matsumoto AM, Bremner WJ. Modulation of pulsatile gonadotropin secretion by testosterone in man. J Clin Endocrinol Metab. 1984;58(4):609-614.
  258. Herbison AE, Skinner DC, Robinson JE, King IS. Androgen receptor-immunoreactive cells in ram hypothalamus: distribution and co-localization patterns with gonadotropin-releasing hormone, somatostatin and tyrosine hydroxylase. Neuroendocrinology. 1996;63(2):120-131.
  259. Smith JT, Dungan HM, Stoll EA, Gottsch ML, Braun RE, Eacker SM, Clifton DK, Steiner RA. Differential regulation of KiSS-1 mRNA expression by sex steroids in the brain of the male mouse. Endocrinology. 2005;146(7):2976-2984.
  260. Woodham C, Birch L, Prins GS. Neonatal estrogen down-regulates prostatic androgen receptor through a proteosome-mediated protein degradation pathway. Endocrinology. 2003;144(11):4841-4850.
  261. Kumar MV, Leo ME, Tindall DJ. Modulation of androgen receptor transcriptional activity by the estrogen receptor. J Androl. 1994;15(6):534-542.
  262. Navarro VM, Gottsch ML, Wu M, Garcia-Galiano D, Hobbs SJ, Bosch MA, Pinilla L, Clifton DK, Dearth A, Ronnekleiv OK, Braun RE, Palmiter RD, Tena-Sempere M, Alreja M, Steiner RA. Regulation of NKB pathways and their roles in the control of Kiss1 neurons in the arcuate nucleus of the male mouse. Endocrinology. 2011;152(11):4265-4275.
  263. Shibata M, Friedman RL, Ramaswamy S, Plant TM. Evidence that down regulation of hypothalamic KiSS-1 expression is involved in the negative feedback action of testosterone to regulate luteinising hormone secretion in the adult male rhesus monkey (Macaca mulatta). J Neuroendocrinol. 2007;19(6):432-438.
  264. Chrousos GP, Torpy DJ, Gold PW. Interactions between the hypothalamic-pituitary-adrenal axis and the female reproductive system: clinical implications. Ann Intern Med. 1998;129(3):229-240.
  265. Kalantaridou SN, Makrigiannakis A, Zoumakis E, Chrousos GP. Stress and the female reproductive system. J Reprod Immunol. 2004;62(1-2):61-68.
  266. Barron JL, Noakes TD, Levy W, Smith C, Millar RP. Hypothalamic dysfunction in overtrained athletes. J Clin Endocrinol Metab. 1985;60(4):803-806.
  267. Berga SL, Mortola JF, Girton L, Suh B, Laughlin G, Pham P, Yen SS. Neuroendocrine aberrations in women with functional hypothalamic amenorrhea. J Clin Endocrinol Metab. 1989;68(2):301-308.
  268. Boccuzzi G, Angeli A, Bisbocci D, Fonzo D, Giadano GP, Ceresa F. Effect of synthetic luteinizing hormone releasing hormone (LH-RH) on the release of gonadotropins in Cushing's disease. J Clin Endocrinol Metab. 1975;40(5):892-895.
  269. Breen KM, Oakley AE, Pytiak AV, Tilbrook AJ, Wagenmaker ER, Karsch FJ. Does cortisol acting via the type II glucocorticoid receptor mediate suppression of pulsatile luteinizing hormone secretion in response to psychosocial stress? Endocrinology. 2007;148(4):1882-1890.
  270. Luger A, Deuster PA, Kyle SB, Gallucci WT, Montgomery LC, Gold PW, Loriaux DL, Chrousos GP. Acute hypothalamic-pituitary-adrenal responses to the stress of treadmill exercise. Physiologic adaptations to physical training. N Engl J Med. 1987;316(21):1309-1315.
  271. Luton JP, Thieblot P, Valcke JC, Mahoudeau JA, Bricaire H. Reversible gonadotropin deficiency in male Cushing's disease. J Clin Endocrinol Metab. 1977;45(3):488-495.
  272. Oakley AE, Breen KM, Clarke IJ, Karsch FJ, Wagenmaker ER, Tilbrook AJ. Cortisol reduces gonadotropin-releasing hormone pulse frequency in follicular phase ewes: influence of ovarian steroids. Endocrinology. 2009;150(1):341-349.
  273. Saketos M, Sharma N, Santoro NF. Suppression of the hypothalamic-pituitary-ovarian axis in normal women by glucocorticoids. Biol Reprod. 1993;49(6):1270-1276.
  274. Breen KM, Karsch FJ. New insights regarding glucocorticoids, stress and gonadotropin suppression. Front Neuroendocrinol. 2006;27(2):233-245.
  275. Chen MD, Ordog T, O'Byrne KT, Goldsmith JR, Connaughton MA, Knobil E. The insulin hypoglycemia-induced inhibition of gonadotropin-releasing hormone pulse generator activity in the rhesus monkey: roles of vasopressin and corticotropin-releasing factor. Endocrinology. 1996;137(5):2012-2021.
  276. Kinsey-Jones JS, Li XF, Knox AM, Wilkinson ES, Zhu XL, Chaudhary AA, Milligan SR, Lightman SL, O'Byrne KT. Down-regulation of hypothalamic kisspeptin and its receptor, Kiss1r, mRNA expression is associated with stress-induced suppression of luteinising hormone secretion in the female rat. J Neuroendocrinol. 2009;21(1):20-29.
  277. Takumi K, Iijima N, Higo S, Ozawa H. Immunohistochemical analysis of the colocalization of corticotropin-releasing hormone receptor and glucocorticoid receptor in kisspeptin neurons in the hypothalamus of female rats. Neurosci Lett. 2012;531(1):40-45.
  278. Phumsatitpong C, Moenter SM. Estradiol-Dependent Stimulation and Suppression of Gonadotropin-Releasing Hormone Neuron Firing Activity by Corticotropin-Releasing Hormone in Female Mice. Endocrinology. 2018;159(1):414-425.
  279. Mitchell JC, Li XF, Breen L, Thalabard JC, O'Byrne KT. The role of the locus coeruleus in corticotropin-releasing hormone and stress-induced suppression of pulsatile luteinizing hormone secretion in the female rat. Endocrinology. 2005;146(1):323-331.
  280. Choi YJ, Habibi HR, Kil GS, Jung MM, Choi CY. Effect of cortisol on gonadotropin inhibitory hormone (GnIH) in the cinnamon clownfish, Amphiprion melanopus. Biochem Biophys Res Commun. 2017;485(2):342-348.
  281. Bohnet HG, Dahlen HG, Wuttke W, Schneider HP. Hyperprolactinemic anovulatory syndrome. J Clin Endocrinol Metab. 1976;42(1):132-143.
  282. Diaz S, Seron-Ferre M, Cardenas H, Schiappacasse V, Brandeis A, Croxatto HB. Circadian variation of basal plasma prolactin, prolactin response to suckling, and length of amenorrhea in nursing women. J Clin Endocrinol Metab. 1989;68(5):946-955.
  283. Molitch ME. Pregnancy and the hyperprolactinemic woman. N Engl J Med. 1985;312(21):1364-1370.
  284. De Rosa M, Zarrilli S, Di Sarno A, Milano N, Gaccione M, Boggia B, Lombardi G, Colao A. Hyperprolactinemia in men: clinical and biochemical features and response to treatment. Endocrine. 2003;20(1-2):75-82.
  285. Moult PJ, Rees LH, Besser GM. Pulsatile gonadotrophin secretion in hyperprolactinaemic amenorrhoea an the response to bromocriptine therapy. Clin Endocrinol (Oxf). 1982;16(2):153-162.
  286. Lecomte P, Lecomte C, Lansac J, Gallier J, Sonier CB, Simonetta C. Pregnancy after intravenous pulsatile gonadotropin-releasing hormone in a hyperprolactinaemic woman resistant to treatment with dopamine agonists. Eur J Obstet Gynecol Reprod Biol. 1997;74(2):219-221.
  287. Polson DW, Sagle M, Mason HD, Adams J, Jacobs HS, Franks S. Ovulation and normal luteal function during LHRH treatment of women with hyperprolactinaemic amenorrhoea. Clin Endocrinol (Oxf). 1986;24(5):531-537.
  288. Grattan DR, Jasoni CL, Liu X, Anderson GM, Herbison AE. Prolactin regulation of gonadotropin-releasing hormone neurons to suppress luteinizing hormone secretion in mice. Endocrinology. 2007;148(9):4344-4351.
  289. Milenkovic L, D'Angelo G, Kelly PA, Weiner RI. Inhibition of gonadotropin hormone-releasing hormone release by prolactin from GT1 neuronal cell lines through prolactin receptors. Proc Natl Acad Sci U S A. 1994;91(4):1244-1247.
  290. Kolbinger W, Beyer C, Fohr K, Reisert I, Pilgrim C. Diencephalic GABAergic neurons in vitro respond to prolactin with a rapid increase in intracellular free calcium. Neuroendocrinology. 1992;56(2):148-152.
  291. Sarkar DK, Yen SS. Hyperprolactinemia decreases the luteinizing hormone-releasing hormone concentration in pituitary portal plasma: a possible role for beta-endorphin as a mediator. Endocrinology. 1985;116(5):2080-2084.
  292. Li C, Chen P, Smith MS. Neuropeptide Y and tuberoinfundibular dopamine activities are altered during lactation: role of prolactin. Endocrinology. 1999;140(1):118-123.
  293. Moore KE. Interactions between prolactin and dopaminergic neurons. Biol Reprod. 1987;36(1):47-58.
  294. Kokay IC, Petersen SL, Grattan DR. Identification of prolactin-sensitive GABA and kisspeptin neurons in regions of the rat hypothalamus involved in the control of fertility. Endocrinology. 2011;152(2):526-535.
  295. Szawka RE, Ribeiro AB, Leite CM, Helena CV, Franci CR, Anderson GM, Hoffman GE, Anselmo-Franci JA. Kisspeptin regulates prolactin release through hypothalamic dopaminergic neurons. Endocrinology. 2010;151(7):3247-3257.
  296. Yamada S, Uenoyama Y, Kinoshita M, Iwata K, Takase K, Matsui H, Adachi S, Inoue K, Maeda KI, Tsukamura H. Inhibition of metastin (kisspeptin-54)-GPR54 signaling in the arcuate nucleus-median eminence region during lactation in rats. Endocrinology. 2007;148(5):2226-2232.
  297. Sonigo C, Bouilly J, Carre N, Tolle V, Caraty A, Tello J, Simony-Conesa FJ, Millar R, Young J, Binart N. Hyperprolactinemia-induced ovarian acyclicity is reversed by kisspeptin administration. J Clin Invest. 2012;122(10):3791-3795.
  298. Araujo-Lopes R, Crampton JR, Aquino NS, Miranda RM, Kokay IC, Reis AM, Franci CR, Grattan DR, Szawka RE. Prolactin regulates kisspeptin neurons in the arcuate nucleus to suppress LH secretion in female rats. Endocrinology. 2014;155(3):1010-1020.
  299. Brown RS, Herbison AE, Grattan DR. Prolactin regulation of kisspeptin neurones in the mouse brain and its role in the lactation-induced suppression of kisspeptin expression. J Neuroendocrinol. 2014;26(12):898-908.
  300. Millar RP, Sonigo C, Anderson RA, George J, Maione L, Brailly-Tabard S, Chanson P, Binart N, Young J. Hypothalamic-Pituitary-Ovarian Axis Reactivation by Kisspeptin-10 in Hyperprolactinemic Women With Chronic Amenorrhea. J Endocr Soc. 2017;1(11):1362-1371.
  301. Aquino NSS, Araujo-Lopes R, Henriques PC, Lopes FEF, Gusmao DO, Coimbra CC, Franci CR, Reis AM, Szawka RE. alpha-Estrogen and Progesterone Receptors Modulate Kisspeptin Effects on Prolactin: Role in Estradiol-Induced Prolactin Surge in Female Rats. Endocrinology. 2017;158(6):1812-1826.
  302. Schneider JE. Energy balance and reproduction. Physiol Behav. 2004;81(2):289-317.
  303. Yeo SH, Colledge WH. The Role of Kiss1 Neurons As Integrators of Endocrine, Metabolic, and Environmental Factors in the Hypothalamic-Pituitary-Gonadal Axis. Front Endocrinol (Lausanne). 2018;9:188.
  304. Dandona P, Dhindsa S. Update: Hypogonadotropic hypogonadism in type 2 diabetes and obesity. J Clin Endocrinol Metab. 2011;96(9):2643-2651.
  305. George JT, Millar RP, Anderson RA. Hypothesis: kisspeptin mediates male hypogonadism in obesity and type 2 diabetes. Neuroendocrinology. 2010;91(4):302-307.
  306. Bergendahl M, Aloi JA, Iranmanesh A, Mulligan TM, Veldhuis JD. Fasting suppresses pulsatile luteinizing hormone (LH) secretion and enhances orderliness of LH release in young but not older men. J Clin Endocrinol Metab. 1998;83(6):1967-1975.
  307. Bergendahl M, Veldhuis JD. Altered pulsatile gonadotropin signaling in nutritional deficiency in the male. Trends Endocrinol Metab. 1995;6(5):145-159.
  308. Weigle DS, Duell PB, Connor WE, Steiner RA, Soules MR, Kuijper JL. Effect of fasting, refeeding, and dietary fat restriction on plasma leptin levels. J Clin Endocrinol Metab. 1997;82(2):561-565.
  309. Loucks AB, Thuma JR. Luteinizing hormone pulsatility is disrupted at a threshold of energy availability in regularly menstruating women. J Clin Endocrinol Metab. 2003;88(1):297-311.
  310. Loucks AB, Verdun M, Heath EM. Low energy availability, not stress of exercise, alters LH pulsatility in exercising women. J Appl Physiol (1985). 1998;84(1):37-46.
  311. Schreihofer DA, Amico JA, Cameron JL. Reversal of fasting-induced suppression of luteinizing hormone (LH) secretion in male rhesus monkeys by intragastric nutrient infusion: evidence for rapid stimulation of LH by nutritional signals. Endocrinology. 1993;132(5):1890-1897.
  312. Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, Karalis A, Mantzoros CS. Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med. 2004;351(10):987-997.
  313. Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role of falling leptin levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J Clin Invest. 2003;111(9):1409-1421.
  314. Farooqi IS, O'Rahilly S. Leptin: a pivotal regulator of human energy homeostasis. Am J Clin Nutr. 2009;89(3):980S-984S.
  315. Rehman R, Jamil Z, Khalid A, Fatima SS. Cross talk between serum Kisspeptin-Leptin during assisted reproduction techniques. Pak J Med Sci. 2018;34(2):342-346.
  316. Backholer K, Smith JT, Rao A, Pereira A, Iqbal J, Ogawa S, Li Q, Clarke IJ. Kisspeptin cells in the ewe brain respond to leptin and communicate with neuropeptide Y and proopiomelanocortin cells. Endocrinology. 2010;151(5):2233-2243.
  317. Cunningham MJ, Clifton DK, Steiner RA. Leptin's actions on the reproductive axis: perspectives and mechanisms. Biol Reprod. 1999;60(2):216-222.
  318. Fernandez-Fernandez R, Martini AC, Navarro VM, Castellano JM, Dieguez C, Aguilar E, Pinilla L, Tena-Sempere M. Novel signals for the integration of energy balance and reproduction. Mol Cell Endocrinol. 2006;254-255:127-132.
  319. Quennell JH, Mulligan AC, Tups A, Liu X, Phipps SJ, Kemp CJ, Herbison AE, Grattan DR, Anderson GM. Leptin indirectly regulates gonadotropin-releasing hormone neuronal function. Endocrinology. 2009;150(6):2805-2812.
  320. Castellano JM, Navarro VM, Fernandez-Fernandez R, Nogueiras R, Tovar S, Roa J, Vazquez MJ, Vigo E, Casanueva FF, Aguilar E, Pinilla L, Dieguez C, Tena-Sempere M. Changes in hypothalamic KiSS-1 system and restoration of pubertal activation of the reproductive axis by kisspeptin in undernutrition. Endocrinology. 2005;146(9):3917-3925.
  321. Luque RM, Kineman RD, Tena-Sempere M. Regulation of hypothalamic expression of KiSS-1 and GPR54 genes by metabolic factors: analyses using mouse models and a cell line. Endocrinology. 2007;148(10):4601-4611.
  322. Roa J, Garcia-Galiano D, Varela L, Sanchez-Garrido MA, Pineda R, Castellano JM, Ruiz-Pino F, Romero M, Aguilar E, Lopez M, Gaytan F, Dieguez C, Pinilla L, Tena-Sempere M. The mammalian target of rapamycin as novel central regulator of puberty onset via modulation of hypothalamic Kiss1 system. Endocrinology. 2009;150(11):5016-5026.
  323. Wahab F, Ullah F, Chan YM, Seminara SB, Shahab M. Decrease in hypothalamic Kiss1 and Kiss1r expression: a potential mechanism for fasting-induced suppression of the HPG axis in the adult male rhesus monkey (Macaca mulatta). Horm Metab Res. 2011;43(2):81-85.
  324. Castellano JM, Navarro VM, Fernandez-Fernandez R, Roa J, Vigo E, Pineda R, Dieguez C, Aguilar E, Pinilla L, Tena-Sempere M. Expression of hypothalamic KiSS-1 system and rescue of defective gonadotropic responses by kisspeptin in streptozotocin-induced diabetic male rats. Diabetes. 2006;55(9):2602-2610.
  325. Elias CF. Leptin action in pubertal development: recent advances and unanswered questions. Trends Endocrinol Metab. 2012;23(1):9-15.
  326. Hill JW, Elmquist JK, Elias CF. Hypothalamic pathways linking energy balance and reproduction. Am J Physiol Endocrinol Metab. 2008;294(5):E827-832.
  327. Donato J, Jr., Cravo RM, Frazao R, Gautron L, Scott MM, Lachey J, Castro IA, Margatho LO, Lee S, Lee C, Richardson JA, Friedman J, Chua S, Jr., Coppari R, Zigman JM, Elmquist JK, Elias CF. Leptin's effect on puberty in mice is relayed by the ventral premammillary nucleus and does not require signaling in Kiss1 neurons. J Clin Invest. 2011;121(1):355-368.
  328. Schalla MA, Stengel A. Central mechanisms of kisspeptin-induced inhibition of food intake. Peptides. 2021;135:170475.
  329. Saito R, Tanaka K, Nishimura H, Nishimura K, Sonoda S, Ueno H, Motojima Y, Yoshimura M, Maruyama T, Yamamoto Y, Kusuhara K, Ueta Y. Centrally administered kisspeptin suppresses feeding via nesfatin-1 and oxytocin in male rats. Peptides. 2019;112:114-124.
  330. Frazao R, Dungan Lemko HM, da Silva RP, Ratra DV, Lee CE, Williams KW, Zigman JM, Elias CF. Estradiol modulates Kiss1 neuronal response to ghrelin. Am J Physiol Endocrinol Metab. 2014;306(6):E606-614.
  331. Kluge M, Schussler P, Schmidt D, Uhr M, Steiger A. Ghrelin suppresses secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in women. J Clin Endocrinol Metab. 2012;97(3):E448-451.
  332. Martini AC, Fernandez-Fernandez R, Tovar S, Navarro VM, Vigo E, Vazquez MJ, Davies JS, Thompson NM, Aguilar E, Pinilla L, Wells T, Dieguez C, Tena-Sempere M. Comparative analysis of the effects of ghrelin and unacylated ghrelin on luteinizing hormone secretion in male rats. Endocrinology. 2006;147(5):2374-2382.
  333. Donoso AO, Lopez FJ, Negro-Vilar A. Cross-talk between excitatory and inhibitory amino acids in the regulation of luteinizing hormone-releasing hormone secretion. Endocrinology. 1992;131(3):1559-1561.
  334. Leranth C, MacLusky NJ, Sakamoto H, Shanabrough M, Naftolin F. Glutamic acid decarboxylase-containing axons synapse on LHRH neurons in the rat medial preoptic area. Neuroendocrinology. 1985;40(6):536-539.
  335. Mijiddorj T, Kanasaki H, Sukhbaatar U, Oride A, Kyo S. DS1, a delta subunit-containing GABA(A) receptor agonist, increases gonadotropin subunit gene expression in mouse pituitary gonadotrophs. Biol Reprod. 2015;92(2):45.
  336. Moore AM, Prescott M, Marshall CJ, Yip SH, Campbell RE. Enhancement of a robust arcuate GABAergic input to gonadotropin-releasing hormone neurons in a model of polycystic ovarian syndrome. Proc Natl Acad Sci U S A. 2015;112(2):596-601.
  337. Watanabe M, Fukuda A, Nabekura J. The role of GABA in the regulation of GnRH neurons. Front Neurosci. 2014;8:387.
  338. Di Giorgio NP, Bizzozzero-Hiriart M, Libertun C, Lux-Lantos V. Unraveling the connection between GABA and kisspeptin in the control of reproduction. Reproduction. 2019;157(6):R225-R233.
  339. Kanasaki H, Tumurbaatar T, Oride A, Hara T, Okada H, Kyo S. Gamma-aminobutyric acidA receptor agonist, muscimol, increases KiSS-1 gene expression in hypothalamic cell models. Reprod Med Biol. 2017;16(4):386-391.
  340. Kurian JR, Keen KL, Guerriero KA, Terasawa E. Tonic control of kisspeptin release in prepubertal monkeys: implications to the mechanism of puberty onset. Endocrinology. 2012;153(7):3331-3336.
  341. Terasawa E, Kurian JR, Guerriero KA, Kenealy BP, Hutz ED, Keen KL. Recent discoveries on the control of gonadotrophin-releasing hormone neurones in nonhuman primates. J Neuroendocrinol. 2010;22(7):630-638.
  342. Piet R, Kalil B, McLennan T, Porteous R, Czieselsky K, Herbison AE. Dominant Neuropeptide Cotransmission in Kisspeptin-GABA Regulation of GnRH Neuron Firing Driving Ovulation. J Neurosci. 2018;38(28):6310-6322.
  343. Comninos AN, Yang L, O'Callaghan J, Mills EG, Wall MB, Demetriou L, Wing VC, Thurston L, Owen BM, Abbara A, Rabiner EA, Dhillo WS. Kisspeptin modulates gamma-aminobutyric acid levels in the human brain. Psychoneuroendocrinology. 2021;129:105244.
  344. Hatef A, Unniappan S. Gonadotropin-releasing hormone, kisspeptin, and gonadal steroids directly modulate nucleobindin-2/nesfatin-1 in murine hypothalamic gonadotropin-releasing hormone neurons and gonadotropes. Biol Reprod. 2017;96(3):635-651.
  345. Christian CA, Moenter SM. Vasoactive intestinal polypeptide can excite gonadotropin-releasing hormone neurons in a manner dependent on estradiol and gated by time of day. Endocrinology. 2008;149(6):3130-3136.
  346. Ducret E, Anderson GM, Herbison AE. RFamide-related peptide-3, a mammalian gonadotropin-inhibitory hormone ortholog, regulates gonadotropin-releasing hormone neuron firing in the mouse. Endocrinology. 2009;150(6):2799-2804.
  347. Lee EJ, Moore CT, Hosny S, Centers A, Jennes L. Expression of estrogen receptor-alpha and c-Fos in adrenergic neurons of the female rat during the steroid-induced LH surge. Brain Res. 2000;875(1-2):56-65.
  348. Miller BH, Olson SL, Levine JE, Turek FW, Horton TH, Takahashi JS. Vasopressin regulation of the proestrous luteinizing hormone surge in wild-type and Clock mutant mice. Biol Reprod. 2006;75(5):778-784.
  349. Palm IF, Van Der Beek EM, Wiegant VM, Buijs RM, Kalsbeek A. Vasopressin induces a luteinizing hormone surge in ovariectomized, estradiol-treated rats with lesions of the suprachiasmatic nucleus. Neuroscience. 1999;93(2):659-666.
  350. Leon S, Tena-Sempere M. Dissecting the Roles of Gonadotropin-Inhibitory Hormone in Mammals: Studies Using Pharmacological Tools and Genetically Modified Mouse Models. Front Endocrinol (Lausanne). 2015;6:189.
  351. Qi Y, Oldfield BJ, Clarke IJ. Projections of RFamide-related peptide-3 neurones in the ovine hypothalamus, with special reference to regions regulating energy balance and reproduction. J Neuroendocrinol. 2009;21(8):690-697.
  352. Liu X, Herbison AE. Kisspeptin Regulation of Neuronal Activity throughout the Central Nervous System. Endocrinol Metab (Seoul). 2016;31(2):193-205.
  353. Acevedo-Rodriguez A, Kauffman AS, Cherrington BD, Borges CS, Roepke TA, Laconi M. Emerging insights into Hypothalamic-pituitary-gonadal (HPG) axis regulation and interaction with stress signaling. J Neuroendocrinol. 2018.
  354. Kriegsfeld LJ, Mei DF, Bentley GE, Ubuka T, Mason AO, Inoue K, Ukena K, Tsutsui K, Silver R. Identification and characterization of a gonadotropin-inhibitory system in the brains of mammals. Proc Natl Acad Sci U S A. 2006;103(7):2410-2415.
  355. Molnar CS, Kallo I, Liposits Z, Hrabovszky E. Estradiol down-regulates RF-amide-related peptide (RFRP) expression in the mouse hypothalamus. Endocrinology. 2011;152(4):1684-1690.
  356. Uenoyama Y, Nakamura S, Hayakawa Y, Ikegami K, Watanabe Y, Deura C, Minabe S, Tomikawa J, Goto T, Ieda N, Inoue N, Sanbo M, Tamura C, Hirabayashi M, Maeda KI, Tsukamura H. Lack of pulse and surge modes and glutamatergic stimulation of luteinising hormone release in Kiss1 knockout rats. J Neuroendocrinol. 2015;27(3):187-197.

 

Calcium & Phosphate Metabolism and Related Disorders During Pregnancy and Lactation

ABSTRACT

 

Pregnancy and lactation require women to provide calcium to the fetus and neonate in amounts that may exceed their normal daily intake. Specific adaptations are invoked within each time period to meet the fetal, neonatal, and maternal calcium requirements. During pregnancy, intestinal calcium and phosphate absorption more than double, and this appears to be the main adaptation to meet the fetal demand for mineral. During lactation, intestinal calcium absorption is normal. Instead, the maternal skeleton is resorbed through the processes of osteoclast-mediated bone resorption and osteocytic osteolysis, in order to provide most of the calcium content of breast milk. In women this lactational loss of bone mass and strength is not suppressed by higher dietary intakes of calcium. After weaning, the skeleton appears to be restored to its prior bone density and strength, together with concomitant increases in bone volumes and cross-sectional diameters that may offset any effect of failure to completely restore the trabecular microarchitecture. These maternal adaptations during pregnancy and lactation also influence the presentation, diagnosis, and management of disorders of calcium, phosphorus, and bone metabolism such as primary hyperparathyroidism, hypoparathyroidism, vitamin D deficiency, and phosphate disorders. Pregnancy and lactation can also cause pseudohyperparathyroidism, a form of hypercalcemia that is mediated by parathyroid hormone-related protein, produced in the breasts or placenta during pregnancy, and by the breasts alone during lactation. Although rarely women may experience fragility fractures during pregnancy or lactation, for most women parity and lactation do not affect the long-term risks of low bone density, osteoporosis, or fracture.

 

INTRODUCTION

 

By the end of full-term gestation, the average fetus accretes about 30 g of calcium, 20 g of phosphorus, and 0.8 g of magnesium to mineralize its skeleton and maintain normal physiological processes. The suckling neonate obtains more than this amount of calcium in breast milk during six months of exclusive lactation. The adaptations through which women meet these calcium demands differ between pregnancy and lactation (Figure 1). Although providing this extra calcium to the offspring could conceivably jeopardize the ability of the mother to maintain her own calcium homeostasis and skeletal mineralization, as this review will make clear, pregnancy and lactation normally do not cause any adverse long-term consequences to the maternal skeleton. The reader is referred to several comprehensive reviews for more details and extensive reference lists for the material covered in this chapter (1-7).

Figure 1. Schematic illustration contrasting calcium homeostasis in human pregnancy and lactation, as compared to normal. The thickness of arrows indicates a relative increase or decrease with respect to the normal and non-pregnant state. Although not illustrated, the serum (total) calcium is decreased during pregnancy, while the ionized calcium remains normal during both pregnancy and lactation. Adapted from ref. (8), © 1997, The Endocrine Society.

 

MINERAL PHYSIOLOGY DURING PREGNANCY

 

Calcium provided from the maternal decidua aids in fertilization of the egg and implantation of the blastocyst; from that point onward the rate of transfer from mother to offspring increases substantially. About 80% of the calcium and phosphate present in the fetal skeleton at the end of gestation crossed the placenta during the third trimester and is mostly derived from the maternal diet during pregnancy. Intestinal calcium and phosphate absorption doubles during pregnancy, driven by 1,25-dihydroxyvitamin D (calcitriol) and other factors, and this appears to be the main adaptation through which women meet the mineral demands of pregnancy.

 

Mineral Ions

 

There are several characteristic changes in maternal serum chemistries and calciotropic hormones during pregnancy (Figure 2), which can easily be mistaken as indicating the presence of a disorder of calcium and bone metabolism, especially since it is not common for clinicians to measure calcium, phosphate, and calciotropic hormones during pregnancy (1). The serum albumin and hemoglobin fall during pregnancy due to hemodilution; the albumin remains low until parturition. In turn that fall in albumin causes the total serum calcium to decline to values that can be well below the normal range. The total calcium includes albumin-bound, bicarbonate-and-citrate-complexed, and ionized or free fractions of calcium. The ionized calcium, the physiologically important fraction, remains constant during pregnancy, which confirms that the fall in total calcium is but an artifact that can usually be ignored. However, that artifactual decline in total calcium means that the serum calcium cannot be relied upon to detect hypercalcemia or hypocalcemia. The ionized calcium should be measured or the albumin-corrected total calcium should be calculated to resolve any uncertainty about what the true serum calcium level is in a pregnant woman. Serum phosphate and magnesium concentrations remain normal during pregnancy.

 

Figure 2. Schematic depiction of longitudinal changes in calcium, phosphorus, and calciotropic hormone levels during human pregnancy. Shaded regions depict the approximate normal ranges. PTH does not decline in women with low calcium or high phytate intakes and may even rise above normal. Calcidiol (25OHD) values are not depicted; most longitudinal studies indicate that the levels are unchanged by pregnancy but may vary due to seasonal variation in sunlight exposure and changes in vitamin D intake. FGF23 values cannot be plotted due to paucity of data. Reproduced with permission from (1).

 

Parathyroid Hormone

 

Parathyroid hormone (PTH) was first measured with assays that reported high circulating levels during pregnancy. The finding of a low total serum calcium and an apparently elevated PTH led to the concept of “physiological secondary hyperparathyroidism in pregnancy.” This erroneous concept persists in some textbooks even today. Those early-generation PTH assays measured many biologically inactive fragments of PTH. When measured with 2-site “intact” assays or the more recent “bio-intact” PTH assays, PTH falls during pregnancy to the low-normal range (i.e. 0-30% of the mean non-pregnant value) during the first trimester and may increase back to the mid-normal range by term. Most of these recent studies of PTH during pregnancy have examined women from North America and Europe who also consumed calcium-replete diets. In contrast, in women from Asia and Gambia who have very low dietary calcium intakes (and often high intakes of phytate that blocks dietary calcium absorption), the PTH level does not suppress during pregnancy and in some cases it has been found to increase above normal (1).

 

Vitamin D Metabolites

 

25-hydroxyvitamin D or calcifediol (25OHD) readily crosses the rodent hemochorial placenta (9) and appears to cross hemochorial human placentas just as easily because cord blood 25OHD levels generally range from 75% to near 100% of the maternal value (1,5). A common concern is that the placenta and fetus might deplete maternal 25OHD stores, but this does not appear to be the case. Even in severely vitamin D deficient women there was no significant change in maternal 25OHD levels during pregnancy (1,4,10,11).

 

Total calcitriol levels increase two to five-fold early in pregnancy and stay elevated until parturition, whereas measured free calcitriol levels were reported to be increased only in the third trimester (12). However, when the 20-40% increase in vitamin D binding protein and the decline in serum albumin during pregnancy are considered, the calculated free calcitriol should be increased in all three trimesters (11,13-16).There are several unusual aspects about this situation. PTH is normally the main stimulator of the renal 1α-hydroxylase (CYP27B1); consequently, elevated calcitriol values are usually driven by high PTH concentrations. An exception to this is the ectopic expression of an autonomously functioning 1α-hydroxylase by such conditions as sarcoidosis and other granulomatous diseases. Another exception is pregnancy because the rise in calcitriol occurs when PTH levels are typically falling or quite low. Moreover, this increase in calcitriol occurs despite the ability of high levels of fibroblast growth factor-23 (FGF23) to suppress the synthesis and increase the catabolism of calcitriol, as shown in animal models of X-linked hypophosphatemic rickets (17-19). Evidence from additional animal models suggest that it is not PTH but other factors, such as PTH-related protein (PTHrP), estradiol, prolactin, and placental lactogen, which drive the 1α-hydroxylase to synthesize calcitriol (1).

 

The placenta expresses 1α-hydroxylase and it is often assumed that autonomous placental production of calcitriol explains why the maternal calcitriol level doubles; other sources such as maternal decidua and the fetus itself could conceivably contribute to the maternal value. However, it appears that any contributions of placenta and other extra-renal sources to the maternal calcitriol level are trivial. Animal studies indicate that the maternal renal 1α-hydroxylase is markedly upregulated during pregnancy (20,21) and that placental expression of 1α-hydroxylase is many-fold less than in the maternal kidneys (17). Clinical studies have revealed that anephric women on dialysis have very low circulating calcitriol levels before and during pregnancy (1,22), confirming that maternal kidneys must be the main source of the normal 2 to 5-fold increase in calcitriol during normal pregnancy. Rodent studies, including pregnancies in mice that lack the 1α-hydroxylase, have confirmed that there is a small contribution of fetal or placental calcitriol to the maternal circulation (1,23,24). However, it is not enough to account for the marked increase in maternal calcitriol that normally occurs during pregnancy.

 

Calcitonin

 

Serum calcitonin levels are increased during pregnancy and may derive from maternal thyroid, breast, decidua, and placenta. The importance of these extrathyroidal sites of calcitonin synthesis has been shown by serum calcitonin levels rising from undetectable to normal values in totally thyroidectomized women who become pregnant (25). Whether calcitonin plays an important role in the physiological responses to the calcium demands of pregnancy is unknown. It has been proposed to protect the maternal skeleton against excessive resorption during times of increased calcium demand; however, there are no clinical studies that have addressed this question. Study of pregnant women who lack the gene for calcitonin or the calcitonin receptor would be informative, but no such women have been identified. On the other hand, mice that lack the gene for calcitonin have normal calcium and bone metabolism during pregnancy (26,27).

 

 

PTHrP concentrations steadily increase in the maternal circulation, reaching the highest levels in the third trimester (1,11). The assays most commonly used in these studies detected PTHrP peptides encompassing amino acids 1-86, but PTHrP is a prohormone. It is cleaved into multiple N-terminal, mid-molecule, and C-terminal peptides, which differ in their biological activities and specificities. None of these peptides have been systematically measured during pregnancy. The commonly available PTHrP1-86 assays do not measure PTHrP1-34, which is likely the most abundant of the active, PTH-like, N-terminal forms of this protein. Moreover, in many clinical studies and case reports it is evident that inappropriate blood samples were used for assaying PTHrP. Special collection and handling are required because PTHrP is rapidly cleaved and degraded in serum. Blood samples should be collected in tubes containing EDTA and aprotinin (a protease inhibitor), kept chilled, and then centrifuged, separated, and frozen within 15 minutes of sample collection. Even with these rigorous standards, PTHrP has been found to begin degrading by 15 minutes after sample collection (28). Many studies did not use this method of sample collection and preparation but instead used sera that had been allowed to clot at room temperature for up to 60 minutes. This likely explains why such studies found undetectable serum concentrations of PTHrP, as compared to those that studied the plasma concentration of PTHrP during pregnancy. Individual case reports are also fraught with this problem, since standard blood collection protocols for hospital laboratories do not use the special handling described above.

 

PTHrP is produced by many tissues in the fetus and mother; consequently, it is uncertain which source(s) account for the rise in PTHrP in the maternal circulation. However, the placenta and breasts are likely the major sources of PTHrP. Whether circulating PTHrP has a role in maternal physiology during pregnancy is unclear, but its rise may stimulate the renal 1α-hydroxylase and contribute to the increase in calcitriol and, indirectly, the suppression of PTH. However, PTHrP appears less potent than PTH in stimulating the 1α-hydroxylase (29,30), which is why its contribution to the rise in calcitriol during pregnancy is uncertain. On the other hand, several case reports have clearly implicated breast- and placental-derived PTHrP as a cause of maternal hypercalcemia with elevated PTHrP and undetectable PTH, a condition called pseudohyperparathyroidism of pregnancy (see below). Since breasts and placenta were sources of excess PTHrP in these cases, those two tissues seem likely to be dominant sources of PTHrP during normal pregnancy. Moreover, since excess PTHrP impacted maternal calcium homeostasis to cause hypercalcemia in these cases, it is possible that the more modest elevations in circulating PTHrP seen during normal pregnancy also affect maternal calcium homeostasis.

 

A carboxyl-terminal form of PTHrP (so-called “osteostatin”) has been shown to inhibit osteoclastic bone resorption in vitro, and thus the notion arises that PTHrP may play a role in protecting the maternal skeleton from excessive resorption during pregnancy (31). Animal studies have shown that PTHrP has other roles during gestation such as regulating placental calcium transport in the fetus (1,32). Maternally produced PTHrP is not likely to regulate placental calcium transport since the protein should not be able to cross the placenta (1,5); instead, it is PTHrP produced within the fetus and placenta that is responsible for regulating placental calcium transport.

 

Fibroblast Growth Factor-23 (FGF23)

 

Intact FGF23 doubles its concentration in the mother’s circulation during rodent pregnancies (17-19), but whether it increases during human pregnancy is not clear. A small longitudinal study of 12 women found that intact FGF23 doubled in the third trimester over values in the first and second trimesters (33), whereas a larger longitudinal study of 81 women found no difference in intact FGF23 values at 36 weeks of gestation and 3-6 months post-weaning (34). Within 24 hours after delivery in another study, mean intact FGF23 did not differ between postpartum women and non-pregnant women (35).

 

Other Hormones

 

This section has focused on changes in static concentrations of minerals and the known calciotropic hormones; there are no studies testing hormonal reserves or response to challenges such as hypocalcemia or hypophosphatemia. Pregnancy also induces significant changes in other hormones known to affect calcium and bone metabolism, including sex steroids, prolactin, placental lactogen, oxytocin, leptin, and IGF-1. Each of these – and possibly other hormones not normally associated with mineral and bone metabolism – may have direct or indirect effects on mineral homeostasis during pregnancy. However, this aspect of the physiology of pregnancy has been largely unexplored to date.

 

Estradiol increases to about 100-fold the levels obtained during normal menstrual cycles and may be influencing bone metabolism. As noted earlier, estradiol has been postulated to be one of the stimulators of the 1α-hydroxylase, which synthesizes high levels of calcitriol during normal pregnancy, and even in the absence of PTH (1).

 

Prolactin and placental lactogen both increase during pregnancy and activate prolactin receptors. Osteoblasts express prolactin receptors, and prolactin receptor deficient mice show decreased bone formation (36). Suppressing the prolactin level with bromocriptine blunted a pregnancy-related gain in bone mineral content in rats (37). These data are consistent with the notion that prolactin or placental lactogen regulate skeletal metabolism during pregnancy. Furthermore, prolactin can indirectly affect skeletal metabolism by stimulating PTHrP synthesis and release from the breasts (38-40).

 

Circulating oxytocin levels also rise during pregnancy (41), and the oxytocin receptor is expressed by osteoclasts and osteoblasts (42). Male and female mice lacking oxytocin or its receptor have an osteoporotic phenotype with low bone formation (43). Oxytocin has been shown to stimulate osteoblast differentiation and function, stimulate osteoclast formation, but inhibit osteoclast function and skeletal resorption (43,44). Taken together, these data predict that oxytocin may regulate bone metabolism during pregnancy, but this has not been directly studied in vivo.

 

Intestinal Calcium and Phosphate Absorption

 

Intestinal absorption of calcium doubles as early as 12 weeks of human pregnancy, as shown by clinical studies that used stable isotopes of calcium, and by other calcium balance studies (1). This increase in calcium absorption appears to be the major maternal adaptation to meet the fetal need for calcium. It has been generally believed that the doubling or tripling of calcitriol levels explains the increased intestinal calcium absorption and concurrent increases in the intestinal expression of calbindin9k-D (S100G), TRPV6, Ca2+-ATPase (PMCA1), and other genes and proteins involved in calcium transport. However, intestinal calcium absorption doubles in the first trimester, well before the rise in free calcitriol levels during the third trimester. Animal studies have indicated that placental lactogen, prolactin, and other factors may stimulate intestinal calcium absorption (1) and that calcitriol or the vitamin D receptor are not required for intestinal calcium absorption to increase during pregnancy (1,23,45-48). 

 

The peak fetal demand for calcium does not occur until the third trimester, and so it is unclear why intestinal calcium absorption should be upregulated in the first trimester. It may allow the maternal skeleton to store calcium in advance of the peak demands for calcium that occur later in pregnancy and lactation; some studies in rodents have shown this to be the case with the bone mineral content rising significantly before term (17,26,47). Women have also been found to be in a positive calcium balance by mid-pregnancy (49), likely due to the effect of increased intestinal calcium absorption on skeletal mineralization.

 

Intestinal phosphate absorption also undergoes a doubling during rodent and other mammalian pregnancies (1), and presumably human pregnancy as well. However, no clinical studies have studied this.

 

Renal Handling of Calcium

 

The doubling of intestinal calcium absorption in the first trimester means that the extra calcium must be passed to the fetus, deposited in the maternal skeleton, or excreted in the urine. Renal calcium excretion is increased as early as the 12th week of gestation, and 24-hour urine values (corrected for creatinine excretion) often exceed the normal range. Conversely, fasting urine calcium values are normal or low, confirming that this hypercalciuria is a consequence of the enhanced intestinal calcium absorption (1). This is absorptive hypercalciuria and will not be reliably detected by spot or fasting urine samples that have been corrected for creatinine concentration. Absorptive hypercalciuria contributes to the increased risk of kidney stones during pregnancy. That women commonly develop hypercalciuria in pregnancy is an indication that they normally absorb more calcium than needed by the fetus, provided that their calcium intake is not low, or that the gastrointestinal absorption of calcium is not impaired by high phytate consumption or malabsorptive disorders.

 

This absorptive hypercalciuria also renders nomograms of fractional calcium excretion invalid for the diagnosis of familial hypocalciuric hypercalcemia during pregnancy (50,51).

 

Pharmacological doses of calcitonin promote renal calcium excretion, but whether the physiologically elevated levels of calcitonin during pregnancy promote renal calcium excretion is unknown.

 

Hypocalciuria during pregnancy has been associated with pre-eclampsia, pregnancy-induced hypertension, and low (equal to non-pregnant values) serum calcitriol (52-55). These changes appear largely secondary to disturbed renal function and reduced creatinine clearance, rather than being causes of the hypertension. However, calcium supplementation reduces the risk of pre-eclampsia in women within the lowest quintile of calcium intake (see section J. Low and High Calcium Intake, below), and so there is a pathophysiological link between calcium metabolism and pregnancy-induced hypertension (1).

 

Skeletal Calcium Metabolism and Bone Density/Bone Marker Changes

 

As mentioned earlier, some studies in rodents indicate that bone mineral content increases during pregnancy, and other studies have shown that histomorphometric parameters of bone turnover are increased at this time. Systematic studies of bone histomorphometry from pregnant women have not been done. However, one study of 15 women who electively terminated a pregnancy at 8-10 weeks found bone biopsy evidence of increased bone resorption, including increased resorption surface and increased numbers of resorption cavities (56). These findings were not present in biopsies obtained from 13 women at term, or in the non-pregnant controls. This study bears repeating but it does suggest that early pregnancy induces skeletal resorption.

 

Bone turnover markers – by-products of bone formation and resorption that can be measured in the serum or urine – have been systematically studied during pregnancy in multiple studies (1). In the non-pregnant adult with osteoporosis these bone markers are fraught with significant intra- and inter-individual variability which limit their utility on an individual basis. There are additional problems with the use of bone markers during pregnancy, including lack of pre-pregnancy baseline values; hemodilution; increased GFR; altered creatinine excretion; placental, uterine and fetal contributions; degradation and clearance by the placenta; and lack of diurnally timed or fasted specimens. Bone resorption has been assessed using urinary (deoxypyridinoline, pyridinoline, and hydroxyproline) and serum (C-telopeptide) markers, and the consistent finding is that bone resorption appears increased from early or mid-pregnancy (1). Conversely, bone formation has been assessed by serum markers (osteocalcin, procollagen I N-terminal propeptide, and bone specific alkaline phosphatase) that were generally not corrected for hemodilution or increased GFR. These bone formation markers are decreased in early or mid-pregnancy from pre-pregnancy or non-pregnant values and rise to normal or above before term (1). The lack of correction for hemodilution and increased GFR means that the apparent decline in bone formation markers may not indicate a true decline in bone formation; it could mask no change or even an increase in bone formation. It should be noted that total alkaline phosphatase rises early in pregnancy due to the placental fraction and is not a useful marker of bone formation during pregnancy.

 

Overall, the scant bone biopsy data and the results of bone turnover markers suggest that bone resorption is increased from as early as the 10th week of pregnancy, whereas bone formation may be suppressed (if the bone formation marker results are correct) or normal (if the bone formation markers are artifactually suppressed due to the aforementioned confounding factors) (1). Notably there is little maternal-fetal calcium transfer occurring in the first trimester, nor is there a marked increase in turnover markers during the third trimester when maternal-fetal calcium transfer is at a peak. These findings may simply underscore that resorption of the maternal skeleton is a minor contributor to calcium homeostasis during pregnancy, whereas the upregulation of intestinal calcium absorption is the main mechanism through which the fetal demand for calcium is met.

 

Another way of assessing whether the maternal skeleton contributes to calcium regulation during pregnancy is to measure bone mineral content or density. A few sequential areal bone density (aBMD) studies have been done using older techniques (single and/or dual-photon absorptiometry, i.e., SPA and DPA), and none with newer techniques (DXA or qCT) due to concerns about fetal radiation exposure. Studies of aBMD are known to be confounded by changes in body composition, weight and skeletal volumes, and all three of these factors change during normal pregnancy. The longitudinal studies used SPA or DPA and found no significant change in cortical or trabecular aBMD during pregnancy (1). Most recent studies examined 16 or fewer subjects with DXA prior to planned pregnancy (range 1-18 months prior, but not always stated) and after delivery (range 1-6 weeks postpartum) [studies reviewed in detail in (57)]. One study found no change in lumbar spine aBMD measurements obtained pre-conception and within 1-2 weeks post-delivery, whereas the other studies reported 4-5% decreases in lumbar aBMD with the postpartum measurement taken between 1-6 weeks post-delivery. A larger study from Denmark obtained DXA measurements of hip, spine, and radius at baseline (up to 8 months before pregnancy) and again within 15 days of delivery in 73 women (58). DXA of the radius was also obtained once each trimester. aBMD decreased between pre-pregnancy and post-pregnancy by 1.8% at the lumbar spine, 3.2% at the total hip, 2.4% at the whole body, 4% at the ultradistal forearm, and 1% at the total forearm, whereas it increased by 0.5% at the proximal 1/3 forearm (58). All women went on to breastfeed, which means that the final aBMD values were confounded by lactation-induced bone loss (see lactation section). These changes in aBMD were statistically significant when compared to 57 non-pregnant controls who also had serial measurements done, but the magnitudes of change were small, and would not be considered statistically significant for an individual woman.

 

Ultrasound measurements of the os calcis and fingers have been examined in other longitudinal studies, which reported a progressive decrease in indices that correlate with volumetric BMD (1,57). Whether observed changes in the os calcis accurately indicate a true or clinically meaningful decrease in volumetric BMD or imply that losses of BMD are occurring in the spine or hip during pregnancy, is not known. The reliability or relevance of data obtained from ultrasound is questionable since this technique failed to detect any change in volumetric BMD at the os calcis during lactation (59), even though substantial bone loss occurs at the spine and hip during lactation (see lactation section).

 

Overall, the existing studies have insufficient power to allow a firm conclusion as to the extent of bone loss that might occur during pregnancy, but it seems likely (especially when data from the Danish study are considered) that modest bone loss occurs, which would be difficult to discern on an individual basis. In the long term, pregnancy does not impair skeletal strength or lead to reduced bone density. Several dozen epidemiological studies of osteoporotic and osteopenic women have failed to find a significant association of parity with bone density or fracture risk (1,60), and many have shown a protective effect of parity (61-78).

 

DISORDERS OF CALCIUM AND BONE METABOLISM DURING PREGNANCY

 

Osteoporosis in Pregnancy (and Especially Lactation)

 

In much of the following, the discussion encompasses osteoporosis that may present in pregnancy, the puerperium, or during lactation, so called pregnancy and lactation-associated osteoporosis (PLO). There can be a continuum of changes in bone metabolism from pre-pregnancy, during pregnancy, in the puerperium, and into the breastfeeding and post-weaning intervals. Most (80-90%) of fractures occur in women during lactation, which indicates that the changes during lactation can be more critical than those that happen during pregnancy. For simplicity, most of the discussion occurs in this section, with a shorter discussion in the lactation section of this chapter. This is also necessary because much of the literature does not distinguish between osteoporosis of pregnancy versus lactation, and because osteoporosis presenting in lactation may have been caused in part by bone loss that occurred during pregnancy.

 

Rarely woman will present with a fragility fracture (most commonly a vertebral fracture, but appendicular fractures also occur) during the third trimester or puerperium, and especially during lactation. Low bone mineral density is usually then confirmed on a subsequent DXA (79). In most cases an aBMD value prior to pregnancy is not available because it was never indicated to be done in women who were until then thought to be healthy. Therefore, the extent of bone loss that occurred during pregnancy or lactation is unknown in most cases, and it is not possible to exclude that low bone density or skeletal fragility preceded pregnancy. In favor of a genetic predisposition is the report that among 35 women who presented with pregnancy associated osteoporosis, there was a higher than expected prevalence of fragility fractures in their mothers (80). A positive family history of osteoporosis has been found in about one-third of patients presenting with vertebral fractures in association with pregnancy or lactation (81-83). Whole genome screening has been done in other case series with pathogenic mutations found in 25-30% of women with PLO and involving such genes as COL1A1, LRP5, and WNT (79,84). Women with genetic mutations tended to have a more severe PLO, as indicated by lower aBMD or a higher number of fractures (82).

 

It is conceivable that pregnancy may induce significant skeletal losses in some women and, thereby, predispose to fracture. The normal pregnancy-induced changes in mineral metabolism may cause excessive resorption of the skeleton in selected cases, and other factors such as low dietary calcium intake and vitamin D insufficiency may contribute to skeletal losses (79). If calcium intake is very low or a malabsorptive disorder is present, skeletal resorption must occur to maintain the calcium supply to the fetus and placenta. A high rate of bone turnover is an independent risk factor for fragility fractures outside of pregnancy, and so the apparently increased bone resorption observed during pregnancy may increase fracture risk. In favor of pregnancy inducing fragility through excess skeletal losses is an observational study of 13 women with pregnancy-associated osteoporosis who were followed for up to eight years. Since the bone mineral density at the spine and hip increased significantly during follow-up in these women, the investigators concluded that significant bone loss must have occurred during the pregnancy (85). Other case series have documented spontaneous 10-20% increases in bone density in women after they fractured during pregnancy or the puerperium (1,86). Taken together, fragility fractures in pregnancy or the puerperium may result from the combination of abnormal skeletal microarchitecture or fragility preceding pregnancy, and increased bone resorption that occurred during pregnancy. In other words, the woman may have entered pregnancy with a normal skeleton that then experienced excessive resorption. Alternatively, she may have had lower bone density and bone strength prior to pregnancy, and her skeleton could not tolerate the increased weight bearing, lumbar lordosis, and physiological changes in bone metabolism that occur during pregnancy.

 

Osteoporosis in association with pregnancy or lactation is likely under-recognized and under-reported. Approximately 75% of vertebral compression fractures in older women are clinically silent and discovered only through radiological surveys; the same is likely true for reproductive age women. Back pain may signal a vertebral fracture, but it may be readily dismissed as a common symptom of normal pregnancy. Consistent with this, an online survey found that women who suffered compression fractures during pregnancy or lactation experienced a mean delay of 3 months before a diagnostic radiograph was done (87). The medical literature is biased toward reports of women who suffered a frightening cascade of multiple compression fractures in association with pregnancy (79,83), whereas how commonly a single vertebral compression fracture might occur and be detected during pregnancy is unknown. Although the literature has focused on vertebral compression fractures occurring with pregnancy, one report suggested that ankle and other lower limb fractures are more common (88).

 

Osteoporosis usually presents in association with a first pregnancy (especially during lactation) with many but not all reports suggesting that there is a low risk of recurrence in subsequent pregnancies, and no risk conferred by higher parity (79,80,85,89-92).  This may indicate that reversible factors such as nutrition were corrected after the first pregnancy, or that all structurally compromised vertebrae collapsed under the load of the first pregnancy. About 60% of patients present with lower thoracic or lumbar pain that may be quite debilitating due to vertebral collapse (85,91,92). Most cases show normal serum chemistries and calciotropic hormone levels, but in a few, secondary causes of bone loss may be identified, including low calcium intake, anorexia nervosa, celiac disease, hyperparathyroidism, osteogenesis imperfecta, inactivating mutations in LRP5, premature ovarian failure, and corticosteroid or heparin therapy (79,80,86,91-95). For example, a woman’s habitual calcium intake of only 229 mg daily was not enough to meet maternal and fetal demands for calcium, and likely contributed to a cascade of vertebral compression fractures occurring late in pregnancy and the puerperium (79). Bone biopsies have only occasionally been done in women having fractures associated with pregnancy or lactation. Most have confirmed osteoporosis and the absence of osteomalacia, while DXA as shown aBMD Z-scores are often in the low bone mass or osteoporotic ranges (85,91,92).

 

Pain from vertebral compression fractures resolves spontaneously over several weeks in most cases while the bone density has been reported to substantially improve in most women following pregnancy, including those who fractured. Fractures tend not to recur in subsequent pregnancies. Thus, although myriad medical treatments (bisphosphonates, estrogen, testosterone, calcitonin, teriparatide, denosumab) and surgical interventions (kyphoplasty, vertebroplasty, spinal fusion) have been used in individual cases of pregnancy-associated osteoporosis (79), the tendency for this condition to spontaneously improve may make pharmacological treatment unjustified except for the severest cases. At the least, it may be prudent to wait 12-18 months to determine the extent to which the aBMD recovers on its own after a pregnancy-associated vertebral fracture (79,86).

 

A recent study examined women with self-reported PLO that resulted in fractures occurring in pregnancy or, more commonly, during lactation (96). They were compared cross-sectionally to two groups of historical controls, healthy premenopausal women and women with known idiopathic osteoporosis (IOP) (96). Women with PLO had lower aBMD and reduced HR-pQCT parameters of cortical and trabecular bone than in healthy women and women with IOP (96). However, women with PLO who were assessed >12 months postpartum (“distant”) had higher aBMD than women who were assessed <12 months postpartum (“early”), which implies that recovery had occurred in the “distant” group (96). Moreover, the aBMD of women in the “distant” group was no different than that of women with IOP (96), which suggests that (at least in this cohort) many women with PLO have low bone mass and strength that precedes pregnancy.

 

A distinct condition is focal, transient osteoporosis of the hip (79). This is rare, self-limited, and probably not a manifestation of altered calciotropic hormone levels or mineral balance during pregnancy. Instead, it may be a consequence of local factors. A variety of theories have been offered to explain this condition, including femoral venous stasis due to pressure from the pregnant uterus, Sudeck’s atrophy or reflex sympathetic dystrophy (causalgia), ischemia, trauma, viral infections, marrow hypertrophy, immobilization, and fetal pressure on the obturator nerve. These patients present with unilateral or bilateral hip pain, limp and/or hip fracture in the third trimester or puerperium (79,97-99). Radiographs and DXA indicate radiolucency and reduced bone density of the symptomatic femoral head and neck, while MRI demonstrates increased water content of the femoral head and the marrow cavity; a joint effusion may also be present. The differential diagnosis of this condition includes inflammatory joint disorders, avascular necrosis of the hip, bone marrow edema, and reflex sympathetic dystrophy. It is a self-limiting condition with both symptoms and radiological appearance resolving within two to six months post-partum; conservative measures including bed rest are usually all that is required during the symptomatic phase (79). Of course, fractures of an involved femur require urgent arthroplasty or hip replacement. The condition recurs in about 40% of cases (not necessarily during pregnancy), unlike osteoporosis involving the spine, and this has prompted prophylactic hip arthroplasty to be done in a few cases where the opposite hip appears to be affected.

 

Vertebral compression fractures and transient osteoporosis of the hip are not always distinct entities; both have occurred in a few women in association with pregnancy (100-103).

 

TREATMENT CONSIDERATIONS

 

For fragility fractures occurring in association with pregnancy, treatment should include optimization of calcium and vitamin D intake, encouraging judicious weight-bearing physical activity, correction of nutritional deficiencies, and treatment of any reversible causes of bone loss or fragility. A supportive corset may provide short-term pain relief. Breastfeeding is not contraindicated but its relative safety should be discussed, since it will lead to progressive loss in aBMD and a transient further increase in fracture risk (see lactation section, below). The potential to rush in with pharmacotherapy should be tempered by the realization that aBMD normally increases 20-70% during the subsequent six to twelve months in women who fractured but received no interventions (1,79,80,85,86,104-112). Therefore, it seems prudent to delay any use of pharmacotherapy for 12-18 months until the extent of spontaneous recovery has been assessed. The extent of spontaneous recovery of lumbar spine aBMD at 12–18 months should be assessed by DXA. HR-pQCT will underestimate the extent of recovery unless the parameters are adjusted to detect and to capture the newly formed bone (osteoid and under-mineralized bone) (86).

 

Documented pharmacotherapies for PLO have included calcitonin, bisphosphonates, denosumab, strontium ranelate, and teriparatide, using the same regimens as for post-menopausal osteoporosis but with treatment durations from 6 months to as much as 10 years (1,79,83,93,113-117). These reports are observational and lacked controls to determine whether any improvements in aBMD exceeded what would have been observed with spontaneous recovery (i.e., use of calcium and vitamin D replacement only). A recent systematic review found that teriparatide increased aBMD by 8-37% and bisphosphonates by 3 to 43% (118). Notably, these ranges overlap with the bone density increases of 20-70% achieved by women in other reports who did not receive pharmacotherapy. In the few case series where pharmacologically treated women were compared to women who received calcium and vitamin D supplementation only, the final aBMD did not differ between groups (83,109,119,120). Of greater concern is that in the largest of these studies (107 women), recurrent fractures occurred twice as often in women who received teriparatide or alendronate (or both), as compared to those who received only calcium and vitamin D supplementation (109). That finding suggests that pharmacotherapy might be harmful, but better controlled studies are needed to be certain of the benefits vs. risks of pharmacotherapy in this setting.

 

Vertebroplasty and kyphoplasty have also been used to treat painful vertebral fractures post-partum, but their overall efficacy is uncertain, given that blinded randomized trials have found no superiority over sham surgery or medical approaches in older subjects (121).

 

For transient osteoporosis of the hip that has not yet resulted in a fracture, the main consideration is whether to prophylactically rod the affected femur(s), or to observe the patient with the expectation that full spontaneous recovery will occur.

 

Primary Hyperparathyroidism

 

This is an uncommon condition but there are no firm data available on its prevalence. Hypercalcemia has been found in 0.03% of routinely screened reproductive age women, while two case series indicated that 1% of all parathyroidectomies were done during pregnancy (122,123). There are at least several hundred cases in the medical literature. The diagnosis will be obscured by the normal pregnancy-induced changes that lower the total serum calcium and suppress PTH; however, finding the ionized or albumin-corrected calcium to be increased, and PTH to be detectable, should indicate primary hyperparathyroidism in most cases (note the exception of FHH in the next section).

 

Physiological changes of pregnancy described earlier increase intestinal calcium absorption and bone resorption, and cause hypercalciuria. In turn these developments can worsen primary hyperparathyroidism and may lead to more severe hypercalcemia, pancreatitis, and kidney stones. The potential for worsening of hypercalcemia is also offset in part by active transfer of calcium across the placenta into the developing fetus.

 

Primary hyperparathyroidism during pregnancy has been reported to cause a variety of symptoms that are not specific to hypercalcemia and cannot be distinguished from those occurring in normal pregnancy (nausea, vomiting, renal colic, malaise, muscle aches and pains, etc.). Conversely the literature has associated primary hyperparathyroidism with an alarming rate of adverse outcomes in the fetus and neonate, including a 10-30% rate for each of spontaneous abortion, stillbirth, and perinatal death, and 30-50% incidence of neonatal tetany (123-127). These high rates were reported in older literature; more recent case series suggest that the rates of stillbirth and neonatal death are each about 2%, while neonatal tetany occurred in 15% (124). The adverse postnatal outcomes are thought to result from suppression of the fetal and neonatal parathyroid glands; this suppression may be prolonged after birth for 3-5 months (124) and in some cases it has been permanent (124,126,128).

 

To prevent these adverse outcomes, surgical correction of primary hyperparathyroidism during the second trimester has been almost universally recommended. Several case series have found elective surgery to be well tolerated, and to dramatically reduce the rate of adverse events when compared to the earlier cases reported in the literature. In a series of 109 mothers with hyperparathyroidism during pregnancy who were treated medically (N=70) or surgically (N=39), there was a 53% incidence of neonatal complications and 16% incidence of neonatal deaths among medically treated mothers, as opposed to a 12.5% neonatal complications and 2.5% neonatal deaths in mothers who underwent parathyroidectomy (123). A systematic review of 382 cases found that neonatal deaths and infant morbidity were lower in surgically treated vs. medically treated mothers (9.1 vs. 38.9%) (129). Furthermore, among surgically treated mothers, neonatal death and infant morbidity significant reduced with surgery done in the second versus third trimesters (4.5 vs. 21.1%) (129). Choosing the second trimester allows organogenesis to be complete in the fetus and to avoid the poorer surgical outcomes and risk of preterm birth associated with surgery during the third trimester (124,127,130,131).

 

Many women in the earliest published cases had a more severe form of primary hyperparathyroidism that is not often seen today (symptomatic, with nephrocalcinosis and renal insufficiency). While mild, asymptomatic primary hyperparathyroidism during pregnancy has been followed conservatively with successful outcomes, complications continue to occur, so that, in the absence of definitive data, surgery during the second trimester remains the most common recommendation (132). An analysis of 1,057 reproductive-aged women with primary hyperparathyroidism found that the rate of C-sections was doubled but there was no difference in the incidence of spontaneous abortions; no data were available on other pregnancy outcomes or neonatal complications (133). In another study of 134 pregnancies in women with primary hyperparathyroidism compared to 431 pregnancies in normocalcemic women, there were no differences in pregnancy-related complications or spontaneous abortions, but neonatal complications were not reported (134). Other recent cases are consistent with lower rates of still birth, neonatal death, and neonatal tetany as compared to the older literature. Therefore, it is reasonable that milder cases diagnosed during the third trimester may be observed until delivery; however, rapid and severe postpartum worsening of the hypercalcemia can occur (131,135-138). This postpartum “parathyroid crisis” occurs because the placental calcium outflow has been lost, while surging PTHrP production in the breasts means an additional factor stimulating bone resorption.

 

TREATMENT CONSIDERATIONS

 

The main consideration is whether to operate electively in the second trimester or observe the patient in the hope that surgical intervention can be delayed until after delivery.

 

Of the five international consensus conferences on the management of primary hyperparathyroidism, only the most recent one commented on pregnancy (139). There are no definitive medical management guidelines for hyperparathyroidism during pregnancy apart from ensuring adequate hydration and correction of electrolyte abnormalities (132). There is some consensus that surgery is indicated for a persistent serum calcium above 2.80 mmol/L (11.1 mg/dL), or an ionized calcium above 1.4 mmol/L (5.6 mg/dL) (140,141). However, another review suggested a higher level 3.00 mmol/L (12.0 mg/dL) (142).  If surgery is undertaken, a bilateral approach is often warranted because of the lack of preoperative imaging to localize the adenoma.

 

Pharmacologic agents to treat hypercalcemia have not been adequately studied in pregnancy, and follow-up on the babies has been brief (if at all).  Calcitonin does not cross the placenta and has been used safely (132). Oral phosphate has also been used but is limited by diarrhea, hypokalemia, and risk of soft tissue calcifications. Bisphosphonates are relatively contraindicated because of their potential adverse effects on fetal endochondral bone development, although a review of 78 cases of bisphosphonate use in pregnancy found no obvious problems in most cases (112). Denosumab crosses the placenta and has been shown to cause an osteopetrotic-like phenotype in fetal cynomolgus monkeys and rats (143,144), and so it should be avoided in human pregnancy. High-dose magnesium has been proposed as a therapeutic alternative which should decreases serum PTH and calcium levels by activating the calcium sensing-receptor, but it has not been adequately studied for this purpose (145,146). The calcium receptor agonist cinacalcet, which is used to suppress PTH and calcium in nonpregnant subjects with primary or secondary hyperparathyroidism and parathyroid carcinoma, has also been tried in pregnancy (147-150). However, since the calcium receptor is expressed in the placenta and regulates fetal-placental calcium transfer (151), the possibility of adverse effects of cinacalcet on the fetus and neonate remain a concern. In 6 case reports, use of calcimimetics resulted in neonatal hypocalcemia in half of them (142). Heparin-free hemodialysis can lower the serum calcium before surgery (152). The recent consensus conference on management of primary hyperparathyroidism advised against the use of bisphosphonates and denosumab and cautioned that data on use of cinacalcet are very limited (139). Nevertheless, these agents have been used when there is a hypercalcemic crisis and surgery isn’t possible; the author is aware of cases treated pharmacologically that have not been reported in the literature.

 

In any case that was followed medically, parathyroidectomy is recommended to be done postpartum, with monitoring in place to detect a postpartum hypercalcemic crisis. Since these women are presenting young with primary hyperparathyroidism, genetic testing may be indicated to rule out inherited causes.

 

Familial Hypocalciuric Hypercalcemia (FHH)

 

Inactivating mutations in the calcium-sensing receptor cause this autosomal dominant condition which presents with hypercalcemia and hypocalciuria (153).  During pregnancy there will be persistent hypercalcemia with non-suppressed PTH, and the serum calcium may progressively rise across the trimesters. As noted above, fractional excretion of calcium is not reduced during pregnancy in this condition, because it is overridden by the physiological increase in intestinal calcium absorption that in turn causes hypercalciuria (50,51,154). Consequently, FHH presenting during pregnancy can be easily mistaken for primary hyperparathyroidism. Unfortunately, at least one pregnant woman with FHH was mistaken to have primary hyperparathyroidism because of worsening hypercalcemia and hypercalciuria and underwent a three-and-a-half gland parathyroidectomy during the second trimester. FHH was only recognized when her hypercalcemia persisted and her neonate was found to be hypercalcemic too (50).

 

Pregnancy in women with familial hypocalciuric hypocalcemia should be uneventful for the mother, but the maternal hypercalcemia has caused fetal and neonatal parathyroid suppression with subsequent tetany in both normal and hemizygous children (5,155,156). A hemizygous neonate will later develop benign hypercalcemia, but if the baby has two inactivating mutations of the calcium receptor (most commonly from both parents being hemizygous for FHH), then the neonate may suffer a life-threatening hypercalcemic crisis (5).

 

TREATMENT CONSIDERATIONS

 

Hypercalcemia is a normal state of affairs for women with FHH and it should not be treated or mistaken for primary hyperparathyroidism. Instead, the newborn should be watched for postnatal hypocalcemia and for the later development of hypercalcemia as a sign that it inherited the mutation.

 

Hypoparathyroidism

 

Hypoparathyroidism during pregnancy usually presents as a pre-existing condition that the clinician is challenged to manage. The natural history of hypoparathyroidism during pregnancy is confusing due to seemingly conflicting case reports in the literature [reviewed in (1,3,157,158)].  Early in pregnancy, some hypoparathyroid women have fewer hypocalcemic symptoms and require less supplemental calcium. This is consistent with a limited role for PTH in the pregnant woman and suggests that an increase in calcitriol and/or increased intestinal calcium absorption occurs in the absence of PTH. However, other case reports clearly indicate that some pregnant hypoparathyroid women required increased calcitriol replacement in order to avoid worsening hypocalcemia. Adding to the confusion is that in some case reports, it appears that the normal, artifactual decrease in total serum calcium during pregnancy was the parameter that led to treatment with increased calcium and calcitriol supplementation; fewer cases reported that dose increments in calcitriol and calcium were made because of maternal symptoms of hypocalcemia or tetany, or objective evidence of true hypocalcemia (low ionized or albumin-corrected calcium).

 

In a well-documented case, calcitriol was stopped and the woman required only supplemental calcium during the third trimester; she developed hypocalcemia within 48 hours of delivery, which implicates loss of placental PTHrP as contributing to her normalization during pregnancy (159). In a series of ten cases of hypoparathyroidism, the ionized calcium remained normal during pregnancy with no need for calcitriol (160).

 

Among these and other recent cases, it is clear that hypoparathyroidism may improve, stay the same, or even worsen during pregnancy (159,161-163). It is not possible to know in advance who will improve and who will worsen during pregnancy; the task is to maintain the albumin-corrected serum calcium or ionized calcium in the normal range for the duration of pregnancy. Maternal hypocalcemia due to hypoparathyroidism must be avoided because it has been associated with intrauterine fetal hyperparathyroidism and fetal death. Conversely, overtreatment must be avoided because maternal hypercalcemia is associated with the fetal and neonatal complications described above under Primary Hyperparathyroidism. Calcitriol and 1α-calcidiol are recommended due to their shorter half-lives, lower risk of toxicity, and the clinical experience with these agents.

 

Late in pregnancy, hypercalcemia may occur in hypoparathyroid women unless the calcitriol dosage and supplemental calcium are substantially reduced or discontinued. This effect appears to be mediated by the increasing levels of PTHrP in the maternal circulation in late pregnancy. Conversely, one case report of hypoparathyroidism in pregnancy found that there was a transient interval of increased requirement for calcitriol immediately after delivery and before lactation was fully underway (159). This may be the result of loss of placental sources of PTHrP followed by a surge in production of PTHrP by the lactating breast (see lactation section, below).

 

TREATMENT CONSIDERATIONS

 

The albumin-corrected serum calcium should be maintained in the mid-normal range in order to insure adequate delivery of calcium to the fetus. This short-term recommendation differs from the common recommendation for non-pregnant adults of maintaining the albumin-corrected serum calcium near or just below the lower end of normal, which reduces the renal filtered load and may slow the progression of nephrocalcinosis over the long term. As discussed above, management during pregnancy may not require any change in pre-existing doses of calcium and calcitriol or 1α-calcidiol, or it may require increases or decreases in both the calcium and the active vitamin D analog.

 

Pseudohypoparathyroidism

 

Pseudohypoparathyroidism is a genetic disorder causing resistance to PTH and manifest by hypocalcemia, hypophosphatemia, and high PTH levels. The two main subtypes include type I, which has blunted PTH-induced phosphaturia and renal production of cyclic AMP, while type II has blunting of PTH-induced phosphaturia only. They are managed similarly to hypoparathyroidism.

 

The published experience with pseudohypoparathyroidism during pregnancy is similar to that of hypoparathyroidism, with a mix of cases that improved, worsened, or had no change. Type I pseudohypoparathyroidism improved during four pregnancies as shown by fewer hypocalcemic symptoms, achievement of normocalcemia, lowering of PTH to near-normal, calcitriol increasing several-fold, urinary calcium excretion normalizing, and supplemental vitamin D, calcitriol, or analogs no longer required (164). These findings are consistent with PTH-independent increases in intestinal calcium absorption and calcitriol synthesis occurring during pregnancy that in turn improve calcium homeostasis; endogenous serum calcitriol did double mid-pregnancy in two women in whom supplemental calcitriol had been discontinued. However, in seven other pregnancies in women with types I and II pseudohypoparathyroidism there was subjective worsening of hypocalcemia-like symptoms, or the apparent need to increase the doses of calcium, calcitriol, or 1α-calcidiol (1,165-168). Another case reported that no change in calcium or calcitriol dosages were required during pregnancy (169). Lastly, a more recent series of 5 patients reported variability of improved, worsened, or no change in the condition during pregnancy (170).

 

If maternal hypocalcemia persists during pregnancy, pseudohypoparathyroidism can lead to the same adverse fetal outcomes that have been associated with maternal hypoparathyroidism, including parathyroid hyperplasia, skeletal demineralization, and fractures (171,172). The maternal calcium concentration must be maintained in the normal range to avoid these fetal outcomes.

 

TREATMENT CONSIDERATIONS

 

Maintain the albumin-corrected serum calcium in the mid-normal range. As with hypoparathyroidism, this may not require any change in pre-existing doses of calcium and calcitriol or 1α-calcidiol, or it may require increases or decreases in both the calcium and active vitamin D analog.

 

Pseudohyperparathyroidism

 

As mentioned above, pseudohyperparathyroidism is hypercalcemia that is caused by physiological release of PTHrP driving increased skeletal resorption, akin to how PTHrP also causes hypercalcemia of malignancy. In several cases the breasts were the confirmed source of PTHrP because the hypercalcemia and elevated PTHrP did not abate until a bilateral reduction mammoplasty was carried out (173-175). The condition has occurred in women who simply have large breasts (175-177). In another case the hypercalcemia, elevated PTHrP, and suppressed PTH reversed within a few hours of an urgent C-section, thereby confirming the placenta as the source (178). In all cases of pseudohyperparathyroidism, it should be anticipated that the cord blood calcium will also be increased, and that the baby is at risk for fetal and neonatal hypoparathyroidism with hypocalcemic tetany.

 

TREATMENT CONSIDERATIONS

 

The diagnosis may not be clear until after delivery, when the serum calcium rapidly normalizes (indicating placental PTHrP was the cause) or stays elevated (indicating production of PTHrP by the breasts is the cause). Prior to delivery, medical management is similar to that for primary hyperparathyroidism.

 

Vitamin D Deficiency and Insufficiency

 

There are no comprehensive studies of the effects of vitamin D deficiency or insufficiency on human pregnancy, but the available data from small clinical trials of vitamin D supplementation, observational studies, and case reports suggest that, consistent with animal studies, vitamin D insufficiency and deficiency is not associated with any worsening of maternal calcium homeostasis (this topic is reviewed in detail in (1,4,7). Maternal hypocalcemia is milder with vitamin D deficiency due to the effects of secondary hyperparathyroidism to increase skeletal resorption and renal calcium reabsorption. Consequently, hypocalcemia due to vitamin D deficiency has not been clearly associated with the same adverse fetal outcomes that maternal hypoparathyroidism causes (reviewed in detail in (5,179)). The fetal effects of vitamin D deficiency, inability to form calcitriol, and absence of the vitamin D receptor have been examined across several animal species and all have indicated that the fetus will have a normal serum calcium and fully mineralized skeleton at term (reviewed in detail in (5,179)). Neonatal hypocalcemia and rickets can occur in infants born of mothers with severe vitamin D deficiency, but it is usually in the weeks to months after birth that this presents, after intestinal calcium absorption becomes dependent on calcitriol.

 

There has been much interest in studies that have inconsistently associated third-trimester measurements of 25OHD, or estimated vitamin D intakes during pregnancy or the first year after birth, with possible extraskeletal benefits in the mother (reduced bacterial vaginosis, pre-eclampsia, pre-term delivery) or in the offspring (lower incidence of type 1 diabetes, greater skeletal mineralization, etc.). These associational studies won’t be discussed in detail (some are cited in: (1,5,180)) because they are confounded by factors which contribute to lower 25OHD levels (maternal overweight/obesity, lower socioeconomic status, poor nutrition, lack of exercise, etc.). It is necessary to test these associations in randomized clinical trials that compare higher versus lower intakes of vitamin D during pregnancy. At present the results of the associational studies are insufficient to warrant prescribing higher intakes of vitamin D during pregnancy to prevent these postulated outcomes.

 

Among many clinical trials of vitamin D supplementation that have been carried out (1), only a few have included over a 100 study participants who were vitamin D deficient at entry, while other recent studies that gained press attention did not include many vitamin D deficient subjects at all.

 

Among the trials with over 100 participants (14,181-189), the two largest were from Bangladesh and UK with over 1,000 participants (187,188). Baseline maternal 25OHD levels were lowest (20-29 nmol/L) in trials from Bangladesh, UK, Iran, and UAE, and in the 40-60 nmol/L range in the remainder. Interventions consisted of placebo/no treatment versus low dose (400 IU/day) or high dose (1,000-5,000 IU/day) vitamin D supplementation, initiated before mid-pregnancy, and maintained until delivery. For most trials the primary outcomes were simply maternal and neonatal-cord blood 25OHD and calcium. The most recent and largest study was from Bangladesh, and the primary outcome was pre-specified as infant length-for-age z-scores at 1 year of age (188). Offspring anthropometric parameters and/or bone mineral content were pre-specified only in a few of the remaining studies (184,186,187).

 

In all studies vitamin D supplementation increased maternal serum and cord blood 25OHD, but there was no overall effect on maternal or cord blood calcium, except one trial that showed a small but significant difference in maternal calcium at delivery (181). The largest achieved difference in maternal 25OHD was over 160 nmol/L (60 ng/mL) in a single study: 16 nmol/L (6.4 ng/mL) in untreated and 168 nmol/L (67 ng/mL) in vitamin D-supplemented mothers at term; however, there was no obstetrical or fetal benefit (181). In that study, the incidence of neonatal hypocalcemia was reduced in offspring of vitamin D treated mothers, reflecting the role of calcitriol to stimulate postnatal intestinal calcium absorption (181). Three trials (mean baseline 25OHD of 20-56 nmol/L) showed a significantly higher maternal PTH level in the low dose vitamin D/placebo arms compared to the high dose arms (182-184), while another trial (mean baseline 25OHD 58-60 nmol/L) did not show any effect of vitamin D supplementation on maternal PTH (14). Vitamin D supplementation also had no effect on maternal aBMD parameters in the post-partum period (14,186,190). Four trials assessed obstetrical outcomes (gestational hypertension, pre-eclampsia, gestational diabetes, infection, post-partum hemorrhage, preterm labor and others), and found no effect of vitamin D supplementation (183,187-189). However, one trial from Iran that showed a 54% risk reduction in gestational diabetes when vitamin D was taken at a dose of 50,000 IU every 2 weeks and compared to 400 IU per day (189).

 

In the large Bangladesh study, there were no significant differences in infant anthropometrics or any other fetal, neonatal or maternal outcomes (188). In one US-based study there was no benefit on mode of delivery, gestational age at delivery, and preterm birth (14), while in another there was no benefit on mode of delivery, C-section rates, adverse events, hypertension, infection, gestational diabetes, still birth, gestational age at delivery, or combinations of these outcomes (183). The UK MAVIDOS trial reported no obstetrical benefit, and no benefit to any of the primary (neonatal bone area, BMC, and aBMD within the first 10-14 days after birth) or secondary outcomes (anthropometric and body composition parameters within 48 hours of birth). However, it received much publicity for a demonstrated increase in BMC and aBMD in winter-born neonates of vitamin D-supplemented vs. placebo-treated mothers (187). Because the neonatal skeleton accretes 100 mg/day of mineral content after birth, this result may reflect improved intestinal mineral delivery over 14 days after birth, rather than a prenatal effect on skeletal mineralization (1,191,192). Curiously, autumn-born neonates of vitamin D-supplemented vs. placebo-treated mothers showed an adverse trend of similar magnitude on BMC and aBMD, which suggests possible harm from vitamin D supplementation, or chance findings due to small numbers within the sub-groups (192). These sub-group analyses of treatment by season interaction were not specified outcomes in the trial registries (ISRCTN 82927713 and EUDRACT 2007-001716-23). In the UK study that achieved the greatest difference in 25OHD levels between untreated and vitamin D-treated mothers and babies, there was a trend for more small for gestational age babies born to mothers who did not receive antenatal vitamin D supplementation (28% vs. 15%, p<0.1), but the study was not powered for this outcome (181). In studies from the UAE, and Iran there was also no benefit on obstetrical outcomes (variably, mode of delivery, C-section rates, adverse events, stillbirths, gestational age at delivery) or neonatal anthropometric measurements and bone mass measurements (182,184,186).

 

The lack of any beneficial effect on maternal, immediate fetal/neonatal and neonatal outcomes (anthropometrics and cord blood calcium), even in studies that included mothers with some of the lowest 25OHD levels (181,184,186,188), suggests that vitamin D supplementation during pregnancy confers no benefit to the neonate.  The most recent study was well-powered to demonstrate a beneficial effect on infant length and other fetal/neonatal outcomes, but did not yield any significant results, despite low vitamin D levels in the mothers at study entry (188).

 

Multiple systematic reviews have assessed the effect of vitamin D supplementation during pregnancy on maternal outcomes (193-198). The reviews differed significantly in their methodology, eligibility criteria, intervention (vitamin D alone or combined with calcium), and inclusion of trials. The Cochrane systematic review showed no significant effect of vitamin D supplementation alone on obstetrical outcomes, but there was a significant reduction in preterm birth and low birth weight (194). Conversely, the combination of vitamin D with calcium was found to reduce the risk of pre-eclampsia, but at the cost of a potential increase in preterm births (194). The other reviews yield variable and inconsistent results among them (193,195-198).

 

An update to the Cochrane systematic review compared vitamin D regimens during pregnancy (≤600 IU versus >600 IU per day; <4000 IU versus ≥ 4000 IU per day) (199). There was no significant effect on pre-eclampsia, preterm birth and low birth weight (low/very low quality of the evidence) (199). The risk of gestational diabetes was reduced with vitamin D supplementation in only one comparison (moderate quality evidence), favoring a dose >600 IU/d (RR 0.54 (0.34 to 0.86). The reviewers noted a concern that safety data were often not reported in the trials that included high doses of vitamin D (199).

 

In summary, the few large RCTs reported to date do not provide evidence for a beneficial effect of high dose vitamin D supplementation (1,000-5,000 IU/day), on maternal and neonatal outcomes. A recent large trial makes it unlikely that vitamin D supplementation in deficient women would yield any beneficial effect on infant length (188). Other studies had low power, baseline maternal serum 25OHD levels that were often not low, and lack of pre-specification of obstetrical and neonatal outcomes. A potential protective effect of vitamin D on neonatal BMC observed in the MAVIDOS trial (187)makes physiological sense because the intestines become the route of calcium delivery after birth, and the finding is consistent with the benefit observed in some RCTs of prenatal vitamin D supplementation reducing the incidence of neonatal hypocalcemia.

 

Overall, available data are insufficient from the individual clinical trials or these systematic reviews to conclude that vitamin D supplementation during pregnancy confers any proven obstetrical benefits, especially with respect to calcium and bone metabolism. However, much interest remains in determining whether vitamin D prevents adverse non-skeletal events in mother and baby. In the meantime, one should always ensure that any pregnant woman is vitamin D sufficient prior to pregnancy, or soon after her pregnancy is confirmed. This ensures that the baby is born with adequate 25OHD so that calcitriol can be upregulated postnatally to stimulate calcium and phosphate absorption in the neonatal intestines.

 

TREATMENT CONSIDERATIONS

 

Vitamin D supplementation is not harmful to the nonpregnant adult unless excessive doses are administered that cause hypervitaminosis D (typically in excess of 10,000 IU daily); however, the maximal level of maternal intake that is safe for the developing fetus has not been established. Clinical trials in pregnant women have safely administered doses ranging from 400 to 5,000 IU of vitamin D daily without obvious adverse effects to mother or offspring. All pregnant women should have their vitamin D intake optimized. This should prevent any non-skeletal outcomes that may be caused by vitamin D deficiency and will also ensure that the newborn has sufficient vitamin D stores to be able to normalize mineral homeostasis in the hours to days after birth, when it switches from being dependent upon the placenta for mineral delivery to relying on its maturing intestines to absorb that mineral from milk.

 

Genetic Vitamin D Resistance Syndromes

 

Case reports and series have provided insight into the effect of pregnancy on genetic disorders of vitamin physiology. Pregnancies have generally been unremarkable in women with vitamin D-dependent rickets type 1 (VDDR-I) which is due to absence of Cyp27b1, and in women with VDDR-II that is due to absence of functional VDRs (200-202). In one such uneventful VDR-II pregnancy, the pre-pregnancy intake of supplemental calcium (800 mg) and high-dose calcitriol were maintained until her clinicians increased the dose of calcitriol later in pregnancy “because of the knowledge that the circulating 1,25-(OH)2D concentration normally rises during pregnancy,” and not because of any change in albumin-adjusted serum calcium (201). Consequently, it’s unclear whether any change was needed. However, it is reasonable to increase the dose of calcitriol to mirror the increase that happens during normal pregnancy. In women with VDDR-I, the dose of calcitriol was unchanged in one-third of pregnancies but increased 1.5 to 2-fold in others (200).

 

TREATMENT CONSIDERATIONS

 

Maintain a normal albumin-corrected serum calcium with adjustments to oral calcium and calcitriol dosing as needed based on serial monitoring of blood chemistries.

 

24-Hydroxylase Deficiency

 

Genetic loss of the catabolic effects of 24-hydroxylase (CYP24A1) causes high calcitriol, mild hypercalcemia, and nephrolithiasis in non-pregnant adults, which may be asymptomatic (203). But during pregnancy in affected individuals, the physiological 2 to 5-fold increase in calcitriol is unopposed by catabolism, which causes an exaggerated increase in calcitriol, followed by severe and potentially life-threatening hypercalcemia. This has been confirmed by study of a mouse model of Cyp24a1 ablation that mimics the human condition (204). In affected women, hypercalcemia can be quite marked, with suppressed or undetectable PTH, and calcitriol concentrations that exceed what is expected for pregnancy (205-207). Pregnant patients may also present with nephrolithiasis or acute pancreatitis (207,208).

 

TREATMENT CONSIDERATIONS

 

Treatment of hypercalcemia is difficult because the agents that could be used are not approved for pregnancy. Increased intestinal calcium absorption is the direct cause, and so use of increased hydration and a modestly restricted calcium diet, combined with phosphate supplementation to bind dietary calcium, are relatively safe management approaches. If PTH increases above normal, then dietary calcium restriction should be lessened to prevent maternal bone resorption and fetal secondary hyperparathyroidism. Other pharmacologic therapy should be reserved for the most severe cases and used with caution. This includes oral glucocorticoids to suppress intestinal calcium absorption, loop diuretics, calcitonin, and bisphosphonates; denosumab should not be used because of teratogenic effects observed in cynomolgus monkeys and mice (143,144). Cinacalcet will not be useful because PTH will already be suppressed due to the combined effects of pregnancy and hypercalcemia.

 

More targeted treatments include ketoconazole or other azoles to inhibit calcitriol synthesis (209), or rifampin to stimulate catabolism of calcitriol via the 23-hydroxylase pathway (209,210). These drugs have been used in pregnancy to treat other conditions, and so there are data to support their relative safety. However, no case reports have yet involved use of these drugs to treat 24-hydroxylase deficiency in pregnancy.

 

Low or High Calcium Intake

 

Through the doubling of intestinal calcium absorption during pregnancy, women have the ability to adapt to wide ranges of calcium intakes and still meet the fetal demand for calcium. It is conceivable that extremely low maternal calcium intakes could impair maternal calcium homeostasis and fetal mineral accretion, but there are scant clinical data examining this possibility (211). One prospective study found that a dietary calcium intake of less than 800 mg daily during the third trimester was associated with significantly lower aBMD at 5 years post-pregnancy (212). Among women with low dietary calcium intake, there are differing results as to whether or not calcium supplementation during pregnancy improved maternal or neonatal bone density (213). There is short term evidence that bone turnover markers were reduced when 1.2 gm of supplemental calcium was given for 20 days to 31 Mexican woman at 25-30 weeks of gestation; their mean dietary calcium intake was 1 gm (214). In a double-blind study conducted in 256 pregnant women, 2 gm of calcium supplementation improved bone mineral content only in the infants of supplemented mothers who were in the lowest quintile of calcium intake (215). Among cases of fragility fractures presenting during pregnancy, some women had very low calcium intakes (<300 mg per day), and in such cases substantial maternal skeletal resorption must be invoked in order to meet the fetal calcium requirement and maintain the maternal serum calcium concentration (79).

 

Overall the physiological changes in calcium and bone metabolism that usually occur during pregnancy and lactation are likely to be sufficient for fetal bone growth and breast-milk production in women with reasonably sufficient calcium intake (216). However, the use of calcium supplementation for pregnant women with low calcium intake can be defended by the links between low calcium intake and both preeclampsia and hypertension in the offspring (211). Clinical trials and meta-analyses have also demonstrated the supplemental calcium will reduce the risk of preeclampsia in women with low dietary calcium intakes, but not in those with adequate intake (217-220).

 

High calcium intake, similar to primary hyperparathyroidism, can cause increased intestinal calcium absorption, maternal hypercalcemia, increased transplacental flow of calcium, and suppression of the fetal parathyroids. Cases of neonatal hypoparathyroidism have been reported wherein women consumed 3 to 6 grams of elemental calcium daily as antacids or antinauseants (1).

 

TREATMENT CONSIDERATIONS

 

Very low calcium intake must be avoided because it increases the risk of pre-eclampsia, maternal skeletal resorption, and inadequate mineralization of the fetal skeleton. Conversely, high calcium intake must be avoided because it increases the risk of maternal hypercalcemia and suppression of the fetal parathyroids. The Institute of Medicine advises that pregnant women require the same calcium intake as non-pregnant women, a value that ranges from 1,000 to 1,200 mg daily, depending on age (221).

 

Hypercalcemia of Malignancy

 

Hypercalcemia of malignancy is usually a terminal condition. When it has been diagnosed during pregnancy, in some cases the baby has been spared from chemotherapy, whereas in other cases the pregnancy was terminated (or ignored) so that chemotherapy could be administered in an attempt to prolong the woman’s life. Half of published case reports haven’t even mentioned the baby’s outcome. A baby born of a mother with humoral hypercalcemia of malignancy may have a high concentration of calcium in cord blood and is at high risk for fetal and neonatal hypoparathyroidism with hypocalcemic tetany.

 

FGF-23 Disorders

 

X-linked hypophosphatemic rickets (XLH) is caused by inactivating mutations in the PHEX gene, which lead to high circulating levels of FGF23. In turn this causes hypophosphatemia with rickets or osteomalacia. Pregnancies were normal in a mouse model of XLH. In particular, despite very high circulating levels of FGF23, which normally downregulate calcitriol synthesis and increase its catabolism, maternal serum calcitriol increased to the high levels normally seen during pregnancy (19,222). This rise in calcitriol should contribute to increased intestinal calcium and phosphate absorption. Several case reports documented persistent hypophosphatemia during pregnancy in women with XLH, but no adverse outcomes (223,224). Nevertheless, it is generally recommended to supplement with calcitriol and phosphate to keep the serum phosphate near normal during pregnancy.

 

Hyperphosphatemic disorders due to loss of FGF23 action have not been studied during human pregnancy, and animal data are also lacking because these conditions are lethal before sexual maturity. Renal insufficiency or failure causes hyperphosphatemia, and both animal and human data indicate that such renal disorders increase the risks of gestational hypertension, pre-eclampsia, eclampsia, and maternal mortality. However, the extent to which hyperphosphatemia contributes to these risks is unknown.

 

TREATMENT CONSIDERATIONS

 

For XLH and other FGF-23 mediated disorders that lead to hypophosphatemia, the serum phosphate should be kept near normal with the use of phosphate supplements and calcitriol if needed. Burosumab is a new anti-FGF23 antibody that corrects hypophosphatemia in XLH and tumor-induced hypophosphatemia (oncogenic osteomalacia). It should not be used in pregnancy because preclinical studies have shown that it crosses the placenta and causes toxicity in cynomolgus monkeys (placental mineralization, late fetal loss, shortened gestation, and preterm births) (225).

 

For hyperphosphatemic disorders due to inadequate FGF23 action, there are no data to guide potential treatment guidelines. However, judicious use of phosphate binders may be of value, with avoidance of any that may be harmful to the fetus.

 

MINERAL PHYSIOLOGY DURING LACTATION AND POST-WEANING

 

As lactation begins the mother is faced with another demand for calcium in order to make milk. The average daily loss of calcium into breast milk is 210 mg, although daily losses as great as 1000 mg calcium have been reported is some women nursing twins (1). Although women meet the calcium demands of pregnancy by upregulating intestinal calcium absorption and serum concentrations of calcitriol, a different adaptation occurs during lactation. A temporary resorption and demineralization of the maternal skeleton appears to be the main mechanism by which breastfeeding women meet these calcium requirements. This adaptation does not appear to require PTH or calcitriol but is regulated by the combined effects of increased circulating concentrations of PTHrP and low estradiol levels. Characteristic changes in serum minerals are calciotropic hormones are depicted in Figure 3.

 

Figure 3. Schematic depiction of longitudinal changes in calcium, phosphorus, and calciotropic hormone levels during lactation and post-weaning skeletal recovery in women. Normal adult values are indicated by the shaded areas. PTH does not decline in women with low calcium or high phytate intakes and may even rise above normal. Calcidiol (25OHD) values are not depicted; most longitudinal studies indicate that the levels are unchanged by lactation but may vary due to seasonal variation in sunlight exposure and changes in vitamin D intake. PTHrP and prolactin surge with each suckling episode, and this is represented by upward spikes. FGF23 values cannot be plotted due to lack of data. Very limited data suggest that calcitriol and PTH may increase during post-weaning, and the lines are dashed to reflect the uncertainty. Reproduced with permission from (1).

 

Mineral Ions

 

The albumin-corrected serum calcium and ionized calcium are both normal during lactation, but longitudinal studies have shown that both are increased slightly over the non-pregnant values. Serum phosphate levels are also higher and may exceed the normal range. Since reabsorption of phosphate by the kidneys appears to be increased, the increased serum phosphate levels may, therefore, reflect the combined effects of increased flux of phosphate into the blood from diet and from skeletal resorption, in the setting of decreased renal phosphate excretion.

 

Parathyroid Hormone

 

PTH, as measured by 2-site “intact” or newer “bio-intact” assays, may be undetectable or in the lower quarter of the normal range during the first several months of lactation in women from North America and Europe who consume adequate calcium. PTH rises to normal by the time of weaning, and in two case series was found to rise above normal post-weaning. In contrast, and similar to findings during pregnancy, PTH did not suppress in several studies of women from Asia and Gambia who consumed diets that were low in calcium or high in phytate. The low PTH concentrations are an indication that PTH isn’t required for mineral homeostasis during lactation, and this is confirmed by hypoparathyroid and aparthyroid women in whom mineral and skeletal homeostasis normalize while they continue to breastfeed (see Hypoparathyroidism, below). The same is true of mice that lack the gene for parathyroid hormone. They are hypocalcemic and hyperphosphatemic when non-pregnant but maintain normal serum calcium and phosphate concentrations while lactating and for a time during post-weaning (17).

 

Vitamin D Metabolites

 

A common concern has been that the suckling neonate will deplete maternal 25OHD stores, but this is not the case. 25OHD should not decline because it does not enter breast milk; conversely, although vitamin D can enter milk, it is present at very low concentrations because appreciable amounts exist in the maternal circulation for only a short postprandial interval. In observational studies and in the placebo arms of several clinical trials, there was either no change or at most a nonsignificant decline in maternal 25OHD levels during lactation, even in severely vitamin D deficient women (4). Calcitriol levels were twice normal during pregnancy but both free and bound calcitriol levels fall to normal within days of parturition and remain there in breastfeeding women (a single study found that women breastfeeding twins had higher calcitriol concentrations than women nursing singletons) (226). Animal studies show that severely vitamin D deficient rodents and mice lacking the vitamin D receptor are able to lactate and provide normal milk (4,47), thereby indicating that vitamin D and calcitriol are not required for lactation to proceed normally (at least in rodents). However, a more recent study found that mice lacking calcitriol produced milk with a lower calcium content (23).

 

Calcitonin

 

Calcitonin levels fall to normal during the first six weeks postpartum in women. Mice lacking the gene that encodes calcitonin lose twice the normal amount of bone mineral content during lactation, which indicates that physiological levels of calcitonin may protect the maternal skeleton from excessive resorption during this time period (26). Whether calcitonin plays a similar role in human physiology is unknown. Totally thyroidectomized women are not calcitonin deficient during lactation due to substantial production of calcitonin by the breasts, which in turn leads to systemic calcitonin concentrations that are the same as in women with intact thyroids (25). Consequently, study of totally thyroidectomized women is not the equivalent of studying a calcitonin-null state when they are breastfeeding.

 

PTHrP

 

Plasma PTHrP concentrations are significantly higher in lactating women than in non-pregnant controls. The source of PTHrP appears to be the breast, which secretes PTHrP into breast milk at concentrations that are 1,000 to 10,000 times the level found in the blood of patients with hypercalcemia of malignancy or in normal human controls. The circulating PTHrP concentration also increases after suckling (227,228). Additional evidence that the breasts are the source of PTHrP include that ablation of the PTHrP gene selectively from mammary tissue resulted in reduced circulating levels of PTHrP in lactating mice (229). PTHrP also has an intimate association with breast tissue: in animals it has been shown to regulate mammary development and blood flow, and the calcium and water content of milk in rodents, whereas in humans it is commonly expressed by breast cancers.

 

Furthermore, as described in more detail below, during lactation PTHrP reaches the maternal circulation from the lactating breast to cause resorption of calcium from the maternal skeleton, renal tubular reabsorption of calcium, and (indirectly) suppression of PTH. In support of this hypothesis, deletion of the PTHrP gene from mammary tissue at the onset of lactation resulted in more modest losses of bone mineral content during lactation in mice (229). In humans, PTHrP correlates with the amount of bone mineral density lost, negatively with serum PTH, and positively with the ionized calcium of lactating women (227,230,231). Lastly, clinical observations in hypoparathyroid and aparathyroid women demonstrate the physiological importance of PTHrP to regulate calcium and skeletal homeostasis during lactation (see Hypoparathyroidism, below).

 

Prolactin

 

Prolactin is persistently elevated during early lactation and spikes further upward with suckling. Later during lactation basal prolactin levels are normal but continue to spike with suckling. Prolactin is important for initiating and maintaining milk production (232), but it also alters bone metabolism by stimulating PTHrP production in lactating mammary tissue, inhibiting GnRH and ovarian function, and possibly (as noted earlier) through direct actions in osteoblasts that express the prolactin receptor.

 

Oxytocin

 

Oxytocin induces milk ejection by contracting myoepithelial cells within mammary tissue. If milk is not ejected, the pressure of milk stasis causes apoptosis of mammary cells, and lactation ceases. Oxytocin spikes in the maternal circulation within 10 minutes after the start of suckling (233). As noted earlier, the oxytocin receptor is expressed in osteoblasts and osteoclasts. But whether oxytocin plays a role in bone metabolism during lactation has proven difficult to determine because oxytocin null mice cannot lactate due to the lack of milk ejection (234).

 

Estradiol

 

In lactating women, estradiol levels fall to menopausal levels or below. This stimulates RANKL and inhibits osteoprotegerin production by osteoblasts, thereby stimulating osteoclast proliferation, function, and bone resorption. Studies in mice have shown that increasing the serum estradiol concentration to 7 times the virgin level blunts the magnitude of bone loss during lactation (235), which confirms that estradiol deficiency plays a role in the skeletal resorption that occurs during lactation.

 

FGF23

 

A single longitudinal study found that intact FGF23 approximately doubled between late pregnancy and weeks 14 and 26 of lactation (34). An increase in FGF23 may occur in response to the increased bone resorption during lactation, which leads to a higher serum phosphorus (1).

 

Other Hormones

 

Serotonin appears to be involved in regulating PTHrP and its effect to resorb the maternal skeleton (236,237). Lactation induces changes in myriad other hormones, such as luteinizing and follicle stimulating hormone, progesterone, testosterone, inhibins, and activins. Whether these play roles in regulating skeletal metabolism during lactation has not been investigated.

 

Intestinal Absorption of Calcium and Phosphate

 

Although intestinal calcium absorption was upregulated during pregnancy, it quickly decreases post-partum to the non-pregnant rate. This also corresponds to the fall in calcitriol levels to normal. This differs from rodents which maintain increased intestinal calcium absorption during lactation; their large litters sizes mandate the need to provide some of the calcium for milk production through this route.

 

Intestinal phosphate absorption has not been measured during human lactation, whereas in rodents it remains increased (1).

 

Renal Handling of Calcium and Phosphate

 

Renal excretion of calcium is typically reduced to about 50 mg per 24 hours or lower, and the glomerular filtration rate is also decreased. These findings suggest that the tubular reabsorption of calcium must be increased to conserve calcium, perhaps through the actions of PTHrP.

 

Renal tubular phosphate reabsorption is increased during lactation. Despite this, urine phosphate excretion may be increased, likely due to the large efflux of phosphate from resorbed bone, which exceeds what is needed for milk production.

 

Skeletal Calcium Metabolism and Bone Density/Bone Marker Changes

 

Histomorphometric data from lactating animals have consistently shown increased bone turnover, and losses of 35% or more of bone mineral are achieved during 2-3 weeks of normal lactation in rodents [reviewed in (1)]. There are no histomorphometric data from lactating women; instead, biochemical markers of bone formation and resorption have been assessed in numerous cross-sectional and prospective studies. Confounding factors discussed earlier for pregnancy need to be considered when assessing bone turnover markers in lactating women; in particular, opposing changes from pregnancy include that the glomerular filtration rate is reduced and the intravascular volume is now contracted. Serum and urinary (24-hr collection) markers of bone resorption are elevated 2-3-fold during lactation and are higher than the levels attained in the third trimester. Serum markers of bone formation (not adjusted for hemoconcentration or reduced GFR) are generally high during lactation and increased over the levels attained during the third trimester. The most marked increase is in the bone resorption markers, suggesting that bone turnover becomes negatively uncoupled, with bone resorption markedly exceeding bone formation, and thereby causing net bone loss. Total alkaline phosphatase falls immediately postpartum due to loss of the placental fraction but may still remain above normal due to elevation of the bone-specific fraction. Overall, these bone marker results are compatible with a significantly increased bone resorption occurring during lactation.

 

Serial measurements of aBMD during lactation (by SPA, DPA or DXA) have shown that bone mineral content falls 3 to 10.0% in women after two to six months of lactation at trabecular sites (lumbar spine, hip, femur and distal radius), with smaller losses at cortical sites and whole body (1,60). These aBMD changes are in accord with studies in rats, mice, and primates in which the skeletal resorption has been shown to occur largely at trabecular surfaces and to a lesser degree in cortical bone, and as much as 25-30% of bone mass or aBMD is lost during three weeks of lactation in normal rodents. The loss in women occurs at a peak rate of 1-3% per month, far exceeding the 1-3% per year that can occur in postmenopausal women who are considered to be losing bone rapidly. This bone resorption is an obligate consequence of lactation and cannot be prevented by increasing the calcium intake in women. Several randomized trials and other studies have shown that calcium supplementation does not significantly reduce the amount of bone lost during lactation (238-241). Not surprisingly, the lactational decrease in bone mineral density correlates with the amount of calcium lost in the breast milk (242).

 

The skeletal losses are due in part to the low estradiol levels during lactation which stimulate osteoclast number and activity. However, low estradiol is not the sole cause of the accelerated bone resorption or other changes in calcium homeostasis that occur during lactation. It is worth noting what happens to reproductive-age women who have marked estrogen deficiency induced by GnRH agonist therapy in order to treat endometriosis, fibroids, or severe acne. Six months of GnRH-induced estrogen deficiency caused 1-4% losses in trabecular (but not cortical) aBMD, increased urinary calcium excretion, and suppression of calcitriol and PTH (Figure 4) [reviewed in (1,8)]. In contrast, during lactation women are not as estrogen deficient but lose more aBMD (at both trabecular and cortical sites), have normal (as opposed to low) calcitriol levels, and have reduced (as opposed to increased) urinary calcium excretion (Figure 4). The difference between isolated GnRH-induced estrogen deficiency and lactation appears to be explained by PTHrP. It stimulates osteoclast-mediated bone resorption and stimulates renal calcium reabsorption; by so doing, it complements the effects of low estradiol during lactation. Stimulated in part by suckling and high prolactin levels, PTHrP and estrogen deficiency combine to cause marked skeletal resorption during lactation (Figure 5).

 

Figure 4. Comparison of the effects of acute estrogen deficiency vs. lactation on calcium and bone metabolism. Acute estrogen deficiency (e.g. GnRH analog therapy) increases skeletal resorption and raises the blood calcium; in turn, PTH is suppressed and renal calcium losses are increased. During lactation, the combined effects of PTHrP (secreted by the breast) and estrogen deficiency increase skeletal resorption, reduce renal calcium losses, and raise the blood calcium, but calcium is directed into breast milk. Reprinted from ref. (8), © 1997, The Endocrine Society.

Figure 5. Brain-Breast-Bone Circuit. The breast is a central regulator of skeletal demineralization during lactation. Suckling and prolactin both inhibit the hypothalamic gonadotropin-releasing hormone (GnRH) pulse center, which in turn suppresses the gonadotropins (luteinizing hormone [LH] and follicle-stimulating hormone [FSH]), leading to low levels of the ovarian sex steroids (estradiol and progesterone). PTHrP production and release from the breast is controlled by several factors, including suckling, prolactin, and the calcium receptor. PTHrP enters the bloodstream and combines with systemically low estradiol levels to markedly upregulate bone resorption. Increased bone resorption releases calcium and phosphate into the blood stream, which then reaches the breast ducts and is actively pumped into the breast milk. PTHrP also passes into milk at high concentrations, but whether swallowed PTHrP plays a role in regulating calcium physiology of the neonate is unknown. Calcitonin (CT) may inhibit skeletal responsiveness to PTHrP and low estradiol. Not depicted are that direct effects of oxytocin and prolactin on bone cells are also possible. Adapted from ref. (26) © 2006, The Endocrine Society.

 

The mechanism through which the skeleton is resorbed has been shown in rodents to involve two processes, both osteoclast-mediated bone resorption (1) and osteocytic osteolysis, in which osteocytes function like osteoclasts to resorb the bone matrix that surrounds them (243). Both of these processes are dependent upon PTHrP. Conditional deletion of the PTHrP gene from mammary tissue reduced the amount of bone resorbed during lactation, whereas conditional deletion of the PTH/PTHrP receptor from osteocytes appeared to eliminate osteocytic osteolysis (244). Moreover, osteocyte-specific deletion of the PTH/PTHrP receptor resulted in a 50% blunting of the amount of aBMD lost during lactation (244), which may indicate that osteocytic osteolysis and osteoclast-mediated bone resorption each contribute about half of the net bone loss achieved during lactation. To date no studies have examined whether osteocytic osteolysis occurs in lactating women.

 

The lactational bone density losses in women are substantially and completely reversed during six to twelve months following weaning (1,60,239). This corresponds to a gain in bone density of 0.5 to 2% per month in a woman who has weaned her infant. The mechanism for this restoration of bone density is unknown, but studies in mice have shown that it is not dependent upon calcitriol, calcitonin, PTH, or PTHrP (17,23,26,47,245,246); nor is it fully explained by restoration of estradiol levels to normal (1). The remarkable ability of the skeleton to recover is exemplified by mice lacking the gene that encodes calcitonin. They lose up to 55% of trabecular mineral content from the spine during lactation but completely restore it within 18 days after weaning (26).

 

Although aBMD appears to be completely restored after weaning in women and all animals that have been studied, more detailed examination of microarchitecture by µCT has shown variable completeness of recovery of microarchitecture by skeletal site. In rodents, the vertebrae recover completely while persistent loss of trabeculae is evident in the long bones (247). Studies in women have similarly shown that the trabecular content of the long bones also appears to be incompletely restored (1,60,239,248,249). However, in both women (77,249,250) and rodents (26,251,252) the cross-sectional diameters and volumes of the long bones may be significantly increased after post-weaning. Such structural changes potentially compensate for any reduction in strength that loss of trabecular microarchitecture might induce, because an increased cross-sectional diameter increases the ability of a hollow shaft to resist bending (cross-sectional moment of inertia) and torsional stress (polar moment of inertia). This is supported by the finding that the breaking strength of rodent bones returns to pre-pregnant values after weaning (1,245), and limited clinical studies that correlated the increased bone volumes achieved after reproductive cycles with increased bone strength (77,250). In women, the vast majority of several dozen epidemiologic studies of pre- and postmenopausal women have found no adverse effect of a history of lactation on peak bone mass, bone density, or hip fracture risk (1,7,57,60). In fact, multiple studies have suggested a protective effect of lactation on the future risk of low aBMD or fragility fractures. Consequently, although lactational bone loss can transiently increase risk of fracture (see next section), it is likely unimportant in the long run for most women, in whom the skeleton is restored to its prior mineral content and strength.

 

DISORDERS OF CALCIUM AND BONE METABOLISM DURING LACTATION

 

Osteoporosis of Lactation

 

On occasion a woman will suffer one or more fragility fractures during lactation, and osteoporotic bone density will be found by DXA (79). As with osteoporosis presenting during pregnancy, this may represent a coincidental, unrelated disease; the woman may have had low bone density and abnormal skeletal microarchitecture prior to pregnancy, such that the normal bone loss incurred by lactation could not be tolerated. Alternatively, it is likely that some cases represent an exacerbation of the normal degree of skeletal demineralization that occurs during lactation, and a continuum from the changes in bone density and bone turnover that occurred during pregnancy. For example, the skeleton may have been normal pre-pregnancy, lost mineral content during pregnancy due to low calcium intake, and experienced the expected further loss during lactation (79). It may be somewhat artificial, therefore, to separate “osteoporosis of lactation” from “osteoporosis of pregnancy.” But since lactation normally causes a significant net loss of bone whereas pregnancy does not, it seems more likely for lactation to cause a subset of women to develop low-trauma fractures. For example, excessive PTHrP release from the lactating breast into the maternal circulation could conceivably cause excessive bone resorption, osteoporosis, and fractures. PTHrP levels were high in one case of lactational osteoporosis, and remained elevated for months after weaning (253).

 

The literature can be confusing because “pregnancy-associated osteoporosis” is the term often used for bone loss and fractures that present during or after pregnancy, despite such fractures being more likely to have resulted from the bone loss during lactation. In fact, multiple case series have demonstrated that about 80-90% of the fragility fractures associated with reproductive cycles occur during lactation, with the remaining 10-20% occurring either during pregnancy or in the puerperium for women who do not breastfeed. Therefore, the term “pregnancy and lactation-associated osteoporosis” (PLO) is a more suitable one to use.

 

The earlier, longer discussion about osteoporosis of pregnancy should be reviewed for more details since everything in that section applies to osteoporosis presenting during lactation. The skeleton may be normal or abnormal prior to pregnancy, bone loss may have occurred during pregnancy, and bone loss will certainly occur during lactation. The magnitude of bone loss during lactation correlates with the volume of breast milk produced, which in turn correlates with the duration of near-exclusive or exclusive breast feeding (i.e., that most or all of the baby’s nutrition comes from breast milk).

 

TREATMENT CONSIDERATIONS

 

The diagnostic and treatment considerations described earlier for osteoporosis of pregnancy also apply to women who are lactating (79). Case series have revealed that a spontaneous 20-70% increase in bone density occurs in women who fractured while breastfeeding (1,79,80,85,86,104-112). Therefore, pharmacological therapy may be best avoided for 12 to 18 months to determine the extent of spontaneous recovery, and then decide if additional treatment is necessary (79,86). The extent of spontaneous recovery of lumbar spine aBMD at 12–18 months should be assessed by DXA. As noted in the pregnancy section, HR-pQCT will underestimate the extent of recovery at this early stage unless the parameters are adjusted to detect under-mineralized bone and osteoid.

 

The mechanism through which post-weaning recovery occurs is not established, but a theoretical concern is that anti-remodeling agents such as bisphosphonates or denosumab might blunt spontaneous recovery since they suppress bone formation. Furthermore, none of the available pharmacotherapies are indicated for use in premenopausal women, and especially not in women who continue to breastfeed. As noted in the earlier section on osteoporosis associated with pregnancy, individual case reports and series have described marked increases in bone mass in association with pharmacotherapy use after lactation, but in each of these cases the magnitude of increases were in keeping with that achieved through spontaneous recovery (86). A few reports compared to women treated with vitamin D and calcium alone, and there was no difference in the final aBMD achieved with pharmacotherapy (usually teriparatide or bisphosphonates) vs. spontaneous recovery (83,109,119,120). One large case series of 107 women found that subsequent fractures were twice as likely to occur in women who had received pharmacotherapy, which suggests that pharmacotherapy may lead to weaker bone (109). Consequently, it is unclear whether early use of pharmacotherapy after lactation-induced bone loss achieved any added benefit.

 

Primary Hyperparathyroidism

 

When surgical correction of primary hyperparathyroidism is not possible or advisable during pregnancy, it is normally carried out in the postpartum interval. A hypercalcemic crisis is possible soon after delivery due in part to loss of the placental calcium infusion, which represented a drain on the serum calcium. If a woman with untreated primary hyperparathyroidism chooses to breastfeed, the serum calcium should be monitored closely for significant worsening due to the effects of secretion of PTHrP from the breasts being added to the high concentrations of PTH already in the circulation. The potential impact of this is even more evident in women with hypoparathyroidism, as discussed below.

 

TREATMENT CONSIDERATIONS

 

The albumin-corrected serum calcium may subside after pregnancy due to intestinal calcium absorption returning to normal after the pregnancy-induced increase. Consequently, there may be no urgency for parathyroidectomy to be done unless a parathyroid crisis occurs. If future pregnancies are planned, then it will be prudent for a neck exploration to be done to correct the condition in advance of another pregnancy. Breastfeeding may require that surgery and required localization procedures be delayed.

 

Familial Hypocalciuric Hypercalcemia

 

The calcium-sensing receptor is expressed in mammary epithelial ducts, and it modulates the production of PTHrP and calcium transport into milk during lactation in mice (254,255). Inactivating calcium-sensing receptor mutations increased mammary tissue production of PTHrP but decreased the calcium content of milk (255). These opposing changes meant that there was a further increase in bone resorption during lactation as compared to normal mice, and the serum calcium also became higher because of reduced output of calcium into milk. Conversely, a calcimimetic drug (similar to cinacalcet) caused increased milk calcium content (255). These data predict that women with FHH will have more marked skeletal resorption during lactation, lower milk calcium content, higher serum calcium, and a greater loss of aBMD during lactation as compared to normal women. However, the effect of breastfeeding on mineral and skeletal homeostasis in women with FHH has not yet been described.

 

TREATMENT CONSIDERATIONS

 

Women with FHH can be expected to breastfeed normally and do not require any treatment. It would be of interest for observational studies to be done to clarify if they experience excess bone loss and produce milk with lower calcium content, as compared to women without FHH.

 

Hypoparathyroidism

 

As noted earlier, in the first day or two after parturition the requirement for supplemental calcium and calcitriol may transiently increase in hypoparathyroid women before secretion of PTHrP surges in the breast tissue (159). The onset of lactation induces an important change in skeletal metabolism because the breasts produce PTHrP at high levels, some of which escapes into the maternal circulation to stimulate bone resorption and raise the serum calcium level. In women who lack parathyroid glands, the release of PTHrP into the circulation during lactation can temporarily restore calcium and bone homeostasis to normal. Levels of calcitriol and calcium supplementation required for treatment of hypoparathyroid women fall early and markedly after the onset of lactation, and hypercalcemia can occur if the calcitriol dosage and calcium intake are not substantially reduced (256-259). This decreased need for calcium and calcitriol occurs at a time when circulating PTHrP levels are high in the maternal circulation (256,259,260). As illustrated in one case, this is consistent with PTHrP reaching the maternal circulation in amounts sufficient to allow stimulation of calcitriol synthesis, and maintenance of normal (or slightly increased) maternal serum calcium (260).

 

TREATMENT CONSIDERATIONS

 

Management of hypoparathyroidism during lactation requires monitoring the albumin-corrected calcium or ionized calcium, reducing or stopping the calcitriol and calcium as indicated, and planning to reinstitute both supplements in escalating doses as lactation wanes. However, production of PTHrP doesn’t necessarily promptly cease around the time of weaning. The author is aware of a woman with hypoparathyroidism who required no supplemental calcium or calcitriol at all for about a year after her baby had been weaned. She thought that her hypoparathyroidism had been permanently cured by breastfeeding, until the abrupt recurrence of symptomatic hypocalcemia, and the need for pre-pregnancy doses of calcium and calcitriol, signaled the end of PTHrP production by her breasts. In another woman, lactation appeared to permanently cure her hypoparathyroidism (261), likely because of persistent production of PTHrP by her breasts.

 

Pseudohypoparathyroidism

 

The management of pseudohypoparathyroidism during lactation has been less well documented. Since these patients are likely resistant to the renal actions of PTHrP, and the placental sources of calcitriol are lost at parturition, the calcitriol requirements might well increase and may require further adjustments during lactation. Conversely, these patients do not have skeletal resistance to PTH, and so it is possible that calcium and calcitriol requirements may decrease secondary to enhanced skeletal resorption caused by the combined effects of high PTH levels, PTHrP release from the breast, and lactation-induced estrogen deficiency. Thus, women with pseudohypoparathyroidism might lose more bone density than normal during lactation, but this has not been studied.

 

TREATMENT CONSIDERATIONS

 

In the absence of data, it would be best to monitor the albumin-corrected serum calcium to determine if any adjustments are needed in the doses of oral calcium and calcitriol.

 

Pseudohyperparathyroidism

 

Severe, PTHrP-mediated hypercalcemia during lactation was first noted to occur in women with large breasts, but it has also developed in women with average-sized breasts in whom milk let-down took place but the baby’s illness prevented breastfeeding (176). This represents an exaggeration of normal lactational physiology, which benefits hypoparathyroid women, but in some normal women can overwhelm the normal regulatory pathways and cause potentially severe hypercalcemia.

 

TREATMENT CONSIDERATIONS

 

Cessation of lactation should reverse the condition, aided by use of breast-binders, and bromocriptine or cabergoline to suppress prolactin. However, a reduction mammoplasty or mastectomy has proved necessary for recalcitrant hypercalcemia in some cases.

 

Vitamin D Deficiency and Insufficiency, and Genetic Vitamin D Disorders

 

The mother andata from small clinical trials, observational studies and case reports indicate that lactation proceeds normally regardless of vitamin D status, and breast milk calcium content is unaffected by vitamin D deficiency or supplementation in doses as high as 6,400 IU per day given to the mother, which achieved maternal 25OHD blood levels of 168 nmol/L (topic reviewed in detail in (1,4,5,7,179)). This is likely because maternal calcium homeostasis is dominated by skeletal resorption induced by estrogen deficiency and PTHrP, with vitamin D/calcitriol playing no substantial role in lactational mineral homeostasis. It is the neonate who will suffer the consequences of being born of a vitamin D deficient mother. This is especially true if the infant is exclusively breast fed, since both vitamin D and 25-hyroxyvitamin D are normally present at very low concentrations in breast milk.

 

The high-dose (6,400 IU) vitamin D supplementation strategy raises the maternal vitamin D concentration substantially for hours and, in turn, this increases the penetration of vitamin D into milk. Consequently, breastfed babies whose mothers consumed 6,400 IU per day achieved the same 25OHD level as babies who received a 300 IU dose of vitamin D directly (262). The potential advantage of this approach is that all of the neonate’s nutrition can then come from breast milk, rather than requiring that breastfed babies receive a vitamin D supplement. Further study is needed regarding the safety of this approach for the mothers and their babies. A Cochrane review also concluded there is no established benefit from high-dose supplementation of breastfeeding women, as compared to the normal route of giving a lower dose of vitamin D directly to a breastfed baby (263).

 

A misconception about vitamin D and milk often arises because marketed forms of cow’s and goat’s milk contain approximately 100 IU of vitamin D per standard serving, but that is a synthetic vitamin D supplement which is added to the milk after the pasteurization stage. It is not put there by the cow or goat.

 

Given that vitamin D deficiency does not affect breast milk content in humans, it is likely that genetic absence of VDR or calcitriol also does not affect milk calcium, but this has not been studied.

 

Whether vitamin D deficiency impairs the ability of the maternal skeleton to recover post-weaning has not been examined in any clinical study. However, studies in mice lacking the vitamin D receptor or Cyp27b1 to synthesize calcitriol, indicate that these mice are able to fully remineralize their skeletons after lactation (23,47).

 

TREATMENT CONSIDERATIONS

 

Breastfeeding women have the same vitamin D intake requirements as non-pregnant and pregnant women (221). Therefore, no change in dose of any oral supplements is needed. Penetrance of vitamin D into breast milk is poor, and so breastfed babies need oral vitamin D supplementation to prevent vitamin D deficiency, until such time as vitamin D-supplemented infant nutrition is taken.

 

24-Hydroxylase Deficiency

 

Hereditary absence of Cyp24a1 reduces calcitriol catabolism, which can lead to very high calcitriol concentrations and marked maternal hypercalcemia during pregnancy. But calcitriol production falls to non-pregnant levels during normal lactation, and the same should be true in women with 24-hydroxylase deficiency. Consistent with this, in one affected woman who breastfed, calcitriol was normal and hypercalcemia was milder compared to pregnancy (205).

 

TREATMENT CONSIDERATIONS

 

Breastfeeding appears unlikely to require any intervention in women with 24-hydroxylase deficiency.

 

Low and High Calcium Intakes

 

The calcium content of milk appears to be largely derived from skeletal resorption during lactation, a process that cannot be suppressed in women by consuming greater amounts of calcium (however, it can be suppressed in rodents by high calcium intakes). It shouldn’t be surprising, therefore, that low calcium intake does not impair breast milk quality, nor does it accentuate maternal bone loss (216). Even in women with very low calcium intakes, the same amount of mineral was lost during lactation from the skeleton as compared to women who had supplemented calcium intakes, and the breast milk calcium content was unaffected by calcium intake or vitamin D status (264-266).  Conversely, since randomized trials and cohort studies have shown that high calcium intakes do not affect the degree of skeletal demineralization that occurs during lactation (238-241), it is unlikely that increasing calcium supplementation well above normal would affect skeletal demineralization either.

 

There is a lingering concern that adolescent mothers with low calcium intakes may not achieve normal peak bone mass as a consequence of lactation-induced bone loss. In fact the adolescent skeleton appears to recover fully from lactation (267), and adolescent women who breastfed have higher aBMD than those who did not breastfeed or had not been pregnant as adolescents (268). However, it remains reasonable to give a calcium supplement to adolescents who lactate in order to ensure that the needs of adolescent growth are met and that peak bone mass is achieved (216,267).

 

TREATMENT CONSIDERATIONS

 

A normal calcium intake of 1,000-1,200 mg daily, as suggested by the Institute of Medicine (221), is recommended for breastfeeding women. Low and high calcium intake should be avoided. It is well established that breastfeeding women do not require increased calcium intake.

 

FGF23 Disorders

 

There is very limited information available about the effect of FGF23-related disorders on mineral homeostasis during lactation, and milk production. Lactation normally causes the serum phosphorus to rise due to the increased release of phosphate from the resorbing skeleton. Indeed in one case report, serum phosphorus increased into the normal range in a breastfeeding woman with XLH (223).  Curiously, the phosphate content of expressed milk was reduced to 50% of normal in two cases of XLH (223,224), which differs from findings in animal models of XLH in which milk phosphate content was normal (269).  Use of oral phosphate supplementation normalized the milk phosphate content in one case where this intervention was studied (223).

 

Milk phosphate content has not been studied in disorders in which FGF23 activity is reduced; however, given the findings with XLH, it is conceivable that milk phosphate content will be increased.

 

TREATMENT CONSIDERATIONS

 

The mineral content of milk is not normally analyzed. Given the limited findings from two cases of XLH, it may be prudent to recommend that oral phosphate supplementation be maintained while breastfeeding, even if the serum phosphorus has spontaneously normalized. If the baby develops hypophosphatemia, this may indicate inadequate milk phosphate content or that the baby inherited XLH. Use of burosumab has not been described in breastfeeding women, but this large protein should have low penetrance into milk and is likely to be destroyed in the infant’s gastrointestinal tract (270).

 

IMPLICATIONS

 

During pregnancy and lactation, novel regulatory systems specific to these settings complement the usual regulators of mineral homeostasis. Intestinal calcium absorption more than doubles from early in pregnancy in order to meet the fetal demand for calcium. In comparison, skeletal calcium resorption is a dominant mechanism by which calcium is supplied to the breast milk, while renal calcium conservation is also apparent. Calcium supplementation during pregnancy will result in a woman absorbing more calcium, but it is clear from clinical trials and observational studies that calcium supplements have little or no impact on the amount of bone lost during lactation.

 

The skeleton appears to recover promptly from lactation to achieve the pre-pregnancy bone mass through mechanisms that remain unclear. The transient loss of bone mass during lactation can at least temporarily compromise skeletal strength and rarely lead to fragility fractures. Furthermore, full recovery of mineral content and bone strength may not always be achieved after weaning. But the majority of women can be assured that the changes in calcium and bone metabolism during pregnancy and lactation are normal, healthy, temporary, and without adverse consequences in the long-term.

 

REFERENCES

 

  1. Kovacs CS. Maternal Mineral and Bone Metabolism During Pregnancy, Lactation, and Post-Weaning Recovery. Physiol Rev 2016; 96:449-547
  2. Wysolmerski JJ. Conversations between breast and bone: physiological bone loss during lactation as evolutionary template for osteolysis in breast cancer and pathological bone loss after menopause. BoneKEy 2007; 4:209-225
  3. Kovacs CS. Calcium and bone metabolism disorders during pregnancy and lactation. Endocrinol Metab Clin North Am 2011; 40:795-826
  4. Kovacs CS. The role of vitamin D in pregnancy and lactation: insights from animal models and clinical studies. Annu Rev Nutr 2012; 32:97-123
  5. Kovacs CS. Bone development and mineral homeostasis in the fetus and neonate: roles of the calciotropic and phosphotropic hormones. Physiol Rev 2014; 94:1143-1218
  6. Kovacs CS. Physiology of Calcium, Phosphorus, and Bone Metabolism During Pregnancy, Lactation, and Postweaning. In: Kovacs CS, Deal CL, eds. Maternal-Fetal and Neonatal Endocrinology. San Diego: Academic Press; 2020:61-73.
  7. Kovacs CS, Chakhtoura M, El-Hajj Fuleihan G. Disorders of Mineral and Bone Metabolism During Pregnancy and Lactation. In: Kovacs CS, Deal CL, eds. Maternal-Fetal and Neonatal Endocrinology. San Diego: Academic Press; 2020:329-370.
  8. Kovacs CS, Kronenberg HM. Maternal-fetal calcium and bone metabolism during pregnancy, puerperium, and lactation. Endocr Rev 1997; 18:832-872
  9. Haddad JG, Jr., Boisseau V, Avioli LV. Placental transfer of vitamin D3 and 25-hydroxycholecalciferol in the rat. J Lab Clin Med 1971; 77:908-915
  10. Hillman LS, Slatopolsky E, Haddad JG. Perinatal vitamin D metabolism. IV. Maternal and cord serum 24,25-dihydroxyvitamin D concentrations. J Clin Endocrinol Metab 1978; 47:1073-1077
  11. Ardawi MS, Nasrat HA, HS BAA. Calcium-regulating hormones and parathyroid hormone-related peptide in normal human pregnancy and postpartum: a longitudinal study. Eur J Endocrinol 1997; 137:402-409
  12. Bikle DD, Gee E, Halloran B, Haddad JG. Free 1,25-dihydroxyvitamin D levels in serum from normal subjects, pregnant subjects, and subjects with liver disease. J Clin Invest 1984; 74:1966-1971
  13. Zhang JY, Lucey AJ, Horgan R, Kenny LC, Kiely M. Impact of pregnancy on vitamin D status: a longitudinal study. Br J Nutr 2014; 112:1081-1087
  14. Hollis BW, Johnson D, Hulsey TC, Ebeling M, Wagner CL. Vitamin D supplementation during pregnancy: double-blind, randomized clinical trial of safety and effectiveness. J Bone Miner Res 2011; 26:2341-2357
  15. Markestad T, Ulstein M, Aksnes L, Aarskog D. Serum concentrations of vitamin D metabolites in vitamin D supplemented pregnant women. A longitudinal study. Acta Obstet Gynecol Scand 1986; 65:63-67
  16. Ritchie LD, Fung EB, Halloran BP, Turnlund JR, Van Loan MD, Cann CE, King JC. A longitudinal study of calcium homeostasis during human pregnancy and lactation and after resumption of menses. Am J Clin Nutr 1998; 67:693-701
  17. Kirby BJ, Ma Y, Martin HM, Buckle Favaro KL, Karaplis AC, Kovacs CS. Upregulation of calcitriol during pregnancy and skeletal recovery after lactation do not require parathyroid hormone. J Bone Miner Res 2013; 28:1987-2000
  18. Cooke-Hubley S, Kirby BJ, Suppiah S, Simmonds CS, Karaplis AC, Kovacs CS. Circulating FGF23 is regulated by PTH and calcitriol during fetal development but low FGF23 does not significantly alter fetal phosphorus metabolism. IBMS BoneKEy 2013; 10:S95
  19. Ma Y, Samaraweera M, Cooke-Hubley S, Kirby BJ, Karaplis AC, Lanske B, Kovacs CS. Neither absence nor excess of FGF23 disturbs murine fetal-placental phosphorus homeostasis or prenatal skeletal development and mineralization. Endocrinology 2014; 155:1596-1605
  20. Fenton E, Britton HG. 25-hydroxycholecalciferol 1 alpha-hydroxylase activity in the kidney of the fetal, neonatal and adult guinea pig. Biol Neonate 1980; 37:254-256
  21. Kubota M, Ohno J, Shiina Y, Suda T. Vitamin D metabolism in pregnant rabbits: differences between the maternal and fetal response to administration of large amounts of vitamin D3. Endocrinology 1982; 110:1950-1956
  22. Turner M, Barre PE, Benjamin A, Goltzman D, Gascon-Barre M. Does the maternal kidney contribute to the increased circulating 1,25-dihydroxyvitamin D concentrations during pregnancy? Miner Electrolyte Metab 1988; 14:246-252
  23. Gillies BR, Ryan BA, Tonkin BA, Poulton IJ, Ma Y, Kirby BJ, St-Arnaud R, Sims NA, Kovacs CS. Absence of Calcitriol Causes Increased Lactational Bone Loss and Lower Milk Calcium but Does Not Impair Post-lactation Bone Recovery in Cyp27b1 Null Mice. J Bone Miner Res 2018; 33:16-26
  24. Ryan BA, Alhani K, Sellars KB, Kirby BJ, St-Arnaud R, Kaufmann M, Jones G, Kovacs CS. Mineral Homeostasis in Murine Fetuses Is Sensitive to Maternal Calcitriol but Not to Absence of Fetal Calcitriol. J Bone Miner Res 2019; 34:669-680
  25. Bucht E, Telenius-Berg M, Lundell G, Sjoberg HE. Immunoextracted calcitonin in milk and plasma from totally thyroidectomized women. Evidence of monomeric calcitonin in plasma during pregnancy and lactation. Acta Endocrinol (Copenh) 1986; 113:529-535
  26. Woodrow JP, Sharpe CJ, Fudge NJ, Hoff AO, Gagel RF, Kovacs CS. Calcitonin plays a critical role in regulating skeletal mineral metabolism during lactation. Endocrinology 2006; 147:4010-4021
  27. Woodrow JP. Calcitonin modulates skeletal mineral loss during lactation through interactions in mammary tissue and directly though osteoclasts in bone [PhD thesis] [PhD]. St. John's, Newfoundland: Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland; 2009.
  28. Hutchesson AC, Hughes SV, Bowden SJ, Ratcliffe WA. In vitro stability of endogenous parathyroid hormone-related protein in blood and plasma. Ann Clin Biochem 1994; 31 ( Pt 1):35-39
  29. Horwitz MJ, Tedesco MB, Sereika SM, Hollis BW, Garcia-Ocana A, Stewart AF. Direct comparison of sustained infusion of human parathyroid hormone-related protein-(1-36). J Clin Endocrinol Metab 2003; 88:1603-1609
  30. Horwitz MJ, Tedesco MB, Sereika SM, Syed MA, Garcia-Ocana A, Bisello A, Hollis BW, Rosen CJ, Wysolmerski JJ, Dann P, Gundberg C, Stewart AF. Continuous PTH and PTHrP infusion causes suppression of bone formation and discordant effects on 1,25(OH)2 vitamin D. J Bone Miner Res 2005; 20:1792-1803
  31. Cornish J, Callon KE, Nicholson GC, Reid IR. Parathyroid hormone-related protein-(107-139) inhibits bone resorption in vivo. Endocrinology 1997; 138:1299-1304
  32. Kovacs CS, Lanske B, Hunzelman JL, Guo J, Karaplis AC, Kronenberg HM. Parathyroid hormone-related peptide (PTHrP) regulates fetal-placental calcium transport through a receptor distinct from the PTH/PTHrP receptor. Proc Natl Acad Sci U S A 1996; 93:15233-15238
  33. Zhao S, Zhou J, Chen R, Zhou W, Geng H, Huang Y, Shi S, Yuan L, Wang Z, Wang D. Decreased FGF23 inhibits placental angiogenesis via the ERK1/2-EGR-1 signaling pathway in preeclampsia. Cytokine 2024; 176:156508
  34. Nabwire F, Hamill MM, Fowler MG, Byamugisha J, Kekitiinwa A, Prentice A. Biochemical Markers of Calcium and Bone Metabolism during and after Lactation in Ugandan Women with HIV on Universal Maternal Antiretroviral Therapy. J Bone Miner Res 2023; 38:1296-1311
  35. Ohata Y, Arahori H, Namba N, Kitaoka T, Hirai H, Wada K, Nakayama M, Michigami T, Imura A, Nabeshima Y, Yamazaki Y, Ozono K. Circulating levels of soluble alpha-Klotho are markedly elevated in human umbilical cord blood. J Clin Endocrinol Metab 2011; 96:E943-947
  36. Clement-Lacroix P, Ormandy C, Lepescheux L, Ammann P, Damotte D, Goffin V, Bouchard B, Amling M, Gaillard-Kelly M, Binart N, Baron R, Kelly PA. Osteoblasts are a new target for prolactin: analysis of bone formation in prolactin receptor knockout mice. Endocrinology 1999; 140:96-105
  37. Suntornsaratoon P, Wongdee K, Goswami S, Krishnamra N, Charoenphandhu N. Bone modeling in bromocriptine-treated pregnant and lactating rats: possible osteoregulatory role of prolactin in lactation. Am J Physiol Endocrinol Metab 2010; 299:E426-436
  38. Thiede MA. The mRNA encoding a parathyroid hormone-like peptide is produced in mammary tissue in response to elevations in serum prolactin. Mol Endocrinol 1989; 3:1443-1447
  39. Ratcliffe WA, Thompson GE, Care AD, Peaker M. Production of parathyroid hormone-related protein by the mammary gland of the goat. J Endocrinol 1992; 133:87-93
  40. Stiegler C, Leb G, Kleinert R, Warnkross H, Ramschak-Schwarzer S, Lipp R, Clarici G, Krejs GJ, Dobnig H. Plasma levels of parathyroid hormone-related peptide are elevated in hyperprolactinemia and correlated to bone density status. J Bone Miner Res 1995; 10:751-759
  41. Dawood MY, Ylikorkala O, Trivedi D, Fuchs F. Oxytocin in maternal circulation and amniotic fluid during pregnancy. J Clin Endocrinol Metab 1979; 49:429-434
  42. Colucci S, Colaianni G, Mori G, Grano M, Zallone A. Human osteoclasts express oxytocin receptor. Biochem Biophys Res Commun 2002; 297:442-445
  43. Tamma R, Colaianni G, Zhu LL, DiBenedetto A, Greco G, Montemurro G, Patano N, Strippoli M, Vergari R, Mancini L, Colucci S, Grano M, Faccio R, Liu X, Li J, Usmani S, Bachar M, Bab I, Nishimori K, Young LJ, Buettner C, Iqbal J, Sun L, Zaidi M, Zallone A. Oxytocin is an anabolic bone hormone. Proc Natl Acad Sci U S A 2009; 106:7149-7154
  44. Liu X, Shimono K, Zhu LL, Li J, Peng Y, Imam A, Iqbal J, Moonga S, Colaianni G, Su C, Lu Z, Iwamoto M, Pacifici M, Zallone A, Sun L, Zaidi M. Oxytocin deficiency impairs maternal skeletal remodeling. Biochem Biophys Res Commun 2009; 388:161-166
  45. Halloran BP, DeLuca HF. Calcium transport in small intestine during pregnancy and lactation. Am J Physiol 1980; 239:E64-68
  46. Brommage R, Baxter DC, Gierke LW. Vitamin D-independent intestinal calcium and phosphorus absorption during reproduction. Am J Physiol 1990; 259:G631-638
  47. Fudge NJ, Kovacs CS. Pregnancy up-regulates intestinal calcium absorption and skeletal mineralization independently of the vitamin D receptor. Endocrinology 2010; 151:886-895
  48. Ryan BA, McGregor NE, Kirby BJ, Al-Tilissi A, Poulton IJ, Sims NA, Kovacs CS. Calcitriol-Dependent and -Independent Regulation of Intestinal Calcium Absorption, Osteoblast Function, and Skeletal Mineralization during Lactation and Recovery in Mice. J Bone Miner Res 2022; 37:2483-2497
  49. Heaney RP, Skillman TG. Calcium metabolism in normal human pregnancy. J Clin Endocrinol Metab 1971; 33:661-670
  50. Walker A, Fraile JJ, Hubbard JG. "Parathyroidectomy in pregnancy"-a single centre experience with review of evidence and proposal for treatment algorithim. Gland Surg 2014; 3:158-164
  51. Morton A. Altered calcium homeostasis during pregnancy may affect biochemical differentiation of hypercalcaemia. Intern Med J 2004; 34:655-656; author reply 656-657
  52. Pedersen EB, Johannesen P, Kristensen S, Rasmussen AB, Emmertsen K, Moller J, Lauritsen JG, Wohlert M. Calcium, parathyroid hormone and calcitonin in normal pregnancy and preeclampsia. Gynecol Obstet Invest 1984; 18:156-164
  53. Frenkel Y, Barkai G, Mashiach S, Dolev E, Zimlichman R, Weiss M. Hypocalciuria of preeclampsia is independent of parathyroid hormone level. Obstet Gynecol 1991; 77:689-691
  54. Seely EW, Wood RJ, Brown EM, Graves SW. Lower serum ionized calcium and abnormal calciotropic hormone levels in preeclampsia. J Clin Endocrinol Metab 1992; 74:1436-1440
  55. Lalau JD, Jans I, el Esper N, Bouillon R, Fournier A. Calcium metabolism, plasma parathyroid hormone, and calcitriol in transient hypertension of pregnancy. Am J Hypertens 1993; 6:522-527
  56. Purdie DW, Aaron JE, Selby PL. Bone histology and mineral homeostasis in human pregnancy. Br J Obstet Gynaecol 1988; 95:849-854
  57. Kovacs CS, El-Hajj Fuleihan G. Calcium and bone disorders during pregnancy and lactation. Endocrinol Metab Clin North Am 2006; 35:21-51, v
  58. Moller UK, Vieth Streym S, Mosekilde L, Rejnmark L. Changes in bone mineral density and body composition during pregnancy and postpartum. A controlled cohort study. Osteoporos Int 2012; 23:1213-1223
  59. To WW, Wong MW. Changes in bone mineral density of the os calcis as measured by quantitative ultrasound during pregnancy and 24 months after delivery. Aust N Z J Obstet Gynaecol 2011; 51:166-171
  60. Sowers M. Pregnancy and lactation as risk factors for subsequent bone loss and osteoporosis. J Bone Miner Res1996; 11:1052-1060
  61. Kreiger N, Kelsey JL, Holford TR, O'Connor T. An epidemiologic study of hip fracture in postmenopausal women. Am J Epidemiol 1982; 116:141-148
  62. Aloia JF, Vaswani AN, Yeh JK, Ross P, Ellis K, Cohn SH. Determinants of bone mass in postmenopausal women. Arch Intern Med 1983; 143:1700-1704
  63. Aloia JF, Cohn SH, Vaswani A, Yeh JK, Yuen K, Ellis K. Risk factors for postmenopausal osteoporosis. Am J Med1985; 78:95-100
  64. Hreshchyshyn MM, Hopkins A, Zylstra S, Anbar M. Associations of parity, breast-feeding, and birth control pills with lumbar spine and femoral neck bone densities. Am J Obstet Gynecol 1988; 159:318-322
  65. Dequeker J, Tobing L, Rutten V, Geusens P. Relative risk factors for osteoporotic fracture: a pilot study of the MEDOS questionnaire. Clin Rheumatol 1991; 10:49-53
  66. Feldblum PJ, Zhang J, Rich LE, Fortney JA, Talmage RV. Lactation history and bone mineral density among perimenopausal women. Epidemiology 1992; 3:527-531
  67. Cumming RG, Klineberg RJ. Breastfeeding and other reproductive factors and the risk of hip fractures in elderly women. Int J Epidemiol 1993; 22:684-691
  68. Berning B, van Kuijk C, Schutte HE, Kuiper JW, Drogendijk AC, Fauser BC. Determinants of lumbar bone mineral density in normal weight, non-smoking women soon after menopause. A study using clinical data and quantitative computed tomography. Bone Miner 1993; 21:129-139
  69. Cure CC, Ramirez PC, Lopez-Jaramillo P. Osteoporosis, pregnancy, and lactation. Lancet 1998; 352:1227-1228
  70. Cure-Cure C, Cure-Ramirez P, Teran E, Lopez-Jaramillo P. Bone-mass peak in multiparity and reduced risk of bone-fractures in menopause. Int J Gynaecol Obstet 2002; 76:285-291
  71. Bjornerem A, Ahmed LA, Jorgensen L, Stormer J, Joakimsen RM. Breastfeeding protects against hip fracture in postmenopausal women: the Tromso study. J Bone Miner Res 2011; 26:2843-2850
  72. Chapman DJ. Longer cumulative breastfeeding duration associated with improved bone strength. Journal of human lactation : official journal of International Lactation Consultant Association 2012; 28:18-19
  73. Canal-Macias ML, Roncero-Martin R, Moran JM, Lavado-Garcia JM, Costa-Fernandez Mdel C, Pedrera-Zamorano JD. Increased bone mineral density is associated with breastfeeding history in premenopausal Spanish women. Archives of medical science : AMS 2013; 9:703-708
  74. Heidari B, Heidari P, Nourooddini HG, Hajian-Tilaki KO. Relationship between parity and bone mass in postmenopausal women according to number of parities and age. J Reprod Med 2013; 58:389-394
  75. Schnatz PF, Marakovits KA, O'Sullivan DM. Assessment of postmenopausal women and significant risk factors for osteoporosis. Obstet Gynecol Surv 2010; 65:591-596
  76. Schnatz PF, Barker KG, Marakovits KA, O'Sullivan DM. Effects of age at first pregnancy and breast-feeding on the development of postmenopausal osteoporosis. Menopause (New York, NY) 2010; 17:1161-1166
  77. Specker B, Binkley T. High parity is associated with increased bone size and strength. Osteoporos Int 2005; 16:1969-1974
  78. Crandall CJ, Liu J, Cauley J, Newcomb PA, Manson JE, Vitolins MZ, Jacobson LT, Rykman KK, Stefanick ML. Associations of Parity, Breastfeeding, and Fractures in the Women's Health Observational Study. Obstet Gynecol2017; 130:171-180
  79. Kovacs CS, Ralston SH. Presentation and management of osteoporosis presenting in association with pregnancy or lactation. Osteoporos Int 2015; 26:2223-2241
  80. Dunne F, Walters B, Marshall T, Heath DA. Pregnancy associated osteoporosis. Clin Endocrinol (Oxf) 1993; 39:487-490
  81. Gehlen M, Lazarescu AD, Hinz C, Schwarz-Eywill M, Pfeifer M, Balasingam S, Maier A. Long-term outcome of patients with pregnancy and lactation-associated osteoporosis (PLO) with a particular focus on quality of life. Clin Rheumatol 2019; 38:3575-3583
  82. Butscheidt S, Tsourdi E, Rolvien T, Delsmann A, Sturznickel J, Barvencik F, Jakob F, Hofbauer LC, Mundlos S, Kornak U, Seefried L, Oheim R. Relevant genetic variants are common in women with pregnancy and lactation-associated osteoporosis (PLO) and predispose to more severe clinical manifestations. Bone 2021; 147:115911
  83. Laroche M, Talibart M, Cormier C, Roux C, Guggenbuhl P, Degboe Y. Pregnancy-related fractures: a retrospective study of a French cohort of 52 patients and review of the literature. Osteoporos Int 2017; 28:3135-3142
  84. Butscheidt S, Delsmann A, Rolvien T, Barvencik F, Al-Bughaili M, Mundlos S, Schinke T, Amling M, Kornak U, Oheim R. Mutational analysis uncovers monogenic bone disorders in women with pregnancy-associated osteoporosis: three novel mutations in LRP5, COL1A1, and COL1A2. Osteoporos Int 2018; 29:1643-1651
  85. Phillips AJ, Ostlere SJ, Smith R. Pregnancy-associated osteoporosis: does the skeleton recover? Osteoporos Int2000; 11:449-454
  86. Kovacs CS. Complex clinical encounter series: osteoporosis presenting during pregnancy and lactation: wait and reassess. J Bone Miner Res 2024; 39:197-201
  87. Kondapalli AV, Kamanda-Kosseh M, Williams JM, Shiau S, Bucovsky M, Colon I, Shane E, Cohen A. Clinical characteristics of pregnancy and lactation associated osteoporosis: An online survey study. Osteoporos Int 2023; 34:1477-1489
  88. Herath M, Wong P, Trinh A, Allan CA, Wallace EM, Ebeling PR, Fuller PJ, Milat F. Minimal-trauma ankle fractures predominate during pregnancy: a 17-year retrospective study. Archives of osteoporosis 2017; 12:86
  89. Dent CE, Friedman M. Pregnancy and idiopathic osteoporosis. Q J Med 1965; 34:341-357
  90. Nordin BE, Roper A. Post-pregnancy osteoporosis; a syndrome? Lancet 1955; 268:431-434
  91. Khovidhunkit W, Epstein S. Osteoporosis in pregnancy. Osteoporos Int 1996; 6:345-354
  92. Smith R, Athanasou NA, Ostlere SJ, Vipond SE. Pregnancy-associated osteoporosis. Quarterly Journal of Medicine 1995; 88:865-878
  93. Campos-Obando N, Oei L, Hoefsloot LH, Kiewiet RM, Klaver CC, Simon ME, Zillikens MC. Osteoporotic vertebral fractures during pregnancy: be aware of a potential underlying genetic cause. J Clin Endocrinol Metab 2014; 99:1107-1111
  94. Cook FJ, Mumm S, Whyte MP, Wenkert D. Pregnancy-associated osteoporosis with a heterozygous deactivating LDL receptor-related protein 5 (LRP5) mutation and a homozygous methylenetetrahydrofolate reductase (MTHFR) polymorphism. J Bone Miner Res 2014; 29:922-928
  95. Khastgir G, Studd JW, King H, Abdalla H, Jones J, Carter G, Alaghband-Zadeh J. Changes in bone density and biochemical markers of bone turnover in pregnancy-associated osteoporosis. Br J Obstet Gynaecol 1996; 103:716-718
  96. Agarwal S, El-Najjar D, Kondapalli AV, Kil N, Kamanda-Kosseh M, Bucovsky M, Colon I, Lappe JM, Stubby J, Recker RR, Guo XE, Shane E, Cohen A. HR-pQCT Reveals Marked Trabecular and Cortical Structural Deficits in Women with Pregnancy and Lactation associated Osteoporosis (PLO). J Bone Miner Res 2024; 39:zjae167
  97. Lakhanpal S, Ginsburg WW, Luthra HS, Hunder GG. Transient regional osteoporosis. A study of 56 cases and review of the literature. Ann Intern Med 1987; 106:444-450
  98. Arayssi TK, Tawbi HA, Usta IM, Hourani MH. Calcitonin in the treatment of transient osteoporosis of the hip. Semin Arthritis Rheum 2003; 32:388-397
  99. Curtiss PH, Jr., Kincaid WE. Transitory demineralization of the hip in pregnancy. A report of three cases. The Journal of bone and joint surgery American volume 1959; 41-A:1327-1333
  100. Baki ME, Uygun H, Ari B, Aydin H. Bilateral femoral neck insufficiency fractures in pregnancy. Eklem hastaliklari ve cerrahisi = Joint diseases & related surgery 2014; 25:60-62
  101. Aynaci O, Kerimoglu S, Ozturk C, Saracoglu M. Bilateral non-traumatic acetabular and femoral neck fractures due to pregnancy-associated osteoporosis. Arch Orthop Trauma Surg 2008; 128:313-316
  102. Bonacker J, Janousek M, Krober M. Pregnancy-associated osteoporosis with eight fractures in the vertebral column treated with kyphoplasty and bracing: a case report. Arch Orthop Trauma Surg 2014; 134:173-179
  103. Pallavi P, Padma S, Vanitha Anna Selvi D. Transient osteoporosis of hip and lumbar spine in pregnancy. Journal of obstetrics and gynaecology of India 2012; 62:8-9
  104. Iwamoto J, Sato Y, Uzawa M, Matsumoto H. Five-year follow-up of a woman with pregnancy and lactation-associated osteoporosis and vertebral fractures. Therapeutics and clinical risk management 2012; 8:195-199
  105. Anai T, Tomiyasu T, Arima K, Miyakawa I. Pregnancy-associated osteoporosis with elevated levels of circulating parathyroid hormone-related protein: a report of two cases. The journal of obstetrics and gynaecology research1999; 25:63-67
  106. Tran HA, Petrovsky N. Pregnancy-associated osteoporosis with hypercalcaemia. Intern Med J 2002; 32:481-485
  107. Segal E, Hochberg I, Weisman Y, Ish-Shalom S. Severe postpartum osteoporosis with increased PTHrP during lactation in a patient after total thyroidectomy and parathyroidectomy. Osteoporos Int 2011; 22:2907-2911
  108. Liel Y, Atar D, Ohana N. Pregnancy-associated osteoporosis: preliminary densitometric evidence of extremely rapid recovery of bone mineral density. South Med J 1998; 91:33-35
  109. Kyvernitakis I, Reuter TC, Hellmeyer L, Hars O, Hadji P. Subsequent fracture risk of women with pregnancy and lactation-associated osteoporosis after a median of 6 years of follow-up. Osteoporos Int 2018; 29:135-142
  110. Kyvernitakis I, Reuter TC, Hellmeyer L, Hars O, Hadji P. Correction to: Subsequent fracture risk of women with pregnancy and lactation-associated osteoporosis after a median of 6 years of follow-up. Osteoporos Int 2023; 34:2143-2144
  111. Ferrari S, Bianchi ML, Eisman JA, Foldes AJ, Adami S, Wahl DA, Stepan JJ, de Vernejoul MC, Kaufman JM. Osteoporosis in young adults: pathophysiology, diagnosis, and management. Osteoporos Int 2012; 23:2735-2748
  112. Stathopoulos IP, Liakou CG, Katsalira A, Trovas G, Lyritis GG, Papaioannou NA, Tournis S. The use of bisphosphonates in women prior to or during pregnancy and lactation. Hormones (Athens, Greece) 2011; 10:280-291
  113. Ijuin A, Yoshikata H, Asano R, Tsuburai T, Kikuchi R, Sakakibara H. Teriparatide and denosumab treatment for pregnancy and lactation-associated osteoporosis with multiple vertebral fractures: A case study. Taiwanese journal of obstetrics & gynecology 2017; 56:863-866
  114. Hong N, Kim JE, Lee SJ, Kim SH, Rhee Y. Changes in bone mineral density and bone turnover markers during treatment with teriparatide in pregnancy- and lactation-associated osteoporosis. Clin Endocrinol (Oxf) 2018; 88:652-658
  115. Grizzo FM, da Silva Martins J, Pinheiro MM, Jorgetti V, Carvalho MD, Pelloso SM. Pregnancy and Lactation-Associated Osteoporosis: Bone Histomorphometric Analysis and Response to Treatment with Zoledronic Acid. Calcif Tissue Int 2015; 97:421-425
  116. Chan B, Zacharin M. Maternal and infant outcome after pamidronate treatment of polyostotic fibrous dysplasia and osteogenesis imperfecta before conception: a report of four cases. J Clin Endocrinol Metab 2006; 91:2017-2020
  117. Tanriover MD, Oz SG, Sozen T. Ten-year follow-up in pregnancy and lactation-associated osteoporosis: sequential therapy with strontium ranelate and ibandronate. Spine J 2015; 15:1164-1165
  118. Anagnostis P, Lampropoulou-Adamidou K, Bosdou JK, Trovas G, Galanis P, Chronopoulos E, Goulis DG, Tournis S. Comparative Effectiveness of Therapeutic Interventions in Pregnancy and Lactation-Associated Osteoporosis: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab 2024; 109:879-901
  119. Orhadje E, Berg K, Hauser B, Ralston SH. Clinical Features, Incidence and Treatment Outcome in Pregnancy-Associated Osteoporosis: A Single-Centre Experience over Two Decades. Calcif Tissue Int 2023; 113:591-596
  120. Lampropoulou-Adamidou K, Trovas G, Triantafyllopoulos IK, Yavropoulou MP, Anastasilakis AD, Anagnostis P, Toulis KA, Makris K, Gazi S, Balanika A, Tournis S. Teriparatide Treatment in Patients with Pregnancy- and Lactation-Associated Osteoporosis. Calcif Tissue Int 2021; 109:554-562
  121. Rodriguez AJ, Fink HA, Mirigian L, Guanabens N, Eastell R, Akesson K, Bauer DC, Ebeling PR. Pain, Quality of Life, and Safety Outcomes of Kyphoplasty for Vertebral Compression Fractures: Report of a Task Force of the American Society for Bone and Mineral Research. J Bone Miner Res 2017; 32:1935-1944
  122. Kort KC, Schiller HJ, Numann PJ. Hyperparathyroidism and pregnancy. Am J Surg 1999; 177:66-68
  123. Kelly TR. Primary hyperparathyroidism during pregnancy. Surgery 1991; 110:1028-1033; discussion 1033-1034
  124. Shangold MM, Dor N, Welt SI, Fleischman AR, Crenshaw MC, Jr. Hyperparathyroidism and pregnancy: a review. Obstet Gynecol Surv 1982; 37:217-228
  125. Wagner G, Transhol L, Melchior JC. Hyperparathyroidism and pregnancy. Acta Endocrinol (Copenh) 1964; 47:549-564
  126. Ludwig GD. Hyperparathyroidism in relation to pregnancy. N Engl J Med 1962; 267:637-642
  127. Delmonico FL, Neer RM, Cosimi AB, Barnes AB, Russell PS. Hyperparathyroidism during pregnancy. Am J Surg1976; 131:328-337
  128. Bruce J, Strong JA. Maternal hyperparathyroidism and parathyroid deficiency in the child, with account of effect of parathyroidectomy on renal function, and of attempt to transplant part of tumor. Q J Med 1955; 24:307-319
  129. Sandler ML, Ho R, Xing MH, Gidumal S, Spitzer H, Levy JC, Chai RL. Primary Hyperparathyroidism During Pregnancy Treated With Parathyroidectomy: A Systematic Review. The Laryngoscope 2021; 131:1915-1921
  130. Kristoffersson A, Dahlgren S, Lithner F, Jarhult J. Primary hyperparathyroidism in pregnancy. Surgery 1985; 97:326-330
  131. Carella MJ, Gossain VV. Hyperparathyroidism and pregnancy: case report and review. J Gen Intern Med 1992; 7:448-453
  132. Schnatz PF, Curry SL. Primary hyperparathyroidism in pregnancy: evidence-based management. Obstet Gynecol Surv 2002; 57:365-376
  133. Abood A, Vestergaard P. Pregnancy outcomes in women with primary hyperparathyroidism. Eur J Endocrinol2014; 171:69-76
  134. Hirsch D, Kopel V, Nadler V, Levy S, Toledano Y, Tsvetov G. Pregnancy outcomes in women with primary hyperparathyroidism. J Clin Endocrinol Metab 2015; 100:2115-2122
  135. Matthias GS, Helliwell TR, Williams A. Postpartum hyperparathyroid crisis. Case report. Br J Obstet Gynaecol1987; 94:807-810
  136. Clark D, Seeds JW, Cefalo RC. Hyperparathyroid crisis and pregnancy. Am J Obstet Gynecol 1981; 140:840-842
  137. Salem R, Taylor S. Hyperparathyroidism in pregnancy. Br J Surg 1979; 66:648-650
  138. Nilsson IL, Adner N, Reihner E, Palme-Kilander C, Edstrom G, Degerblad M. Primary hyperparathyroidism in pregnancy: a diagnostic and therapeutic challenge. J Womens Health (Larchmt) 2010; 19:1117-1121
  139. Bilezikian JP, Khan AA, Silverberg SJ, Fuleihan GE, Marcocci C, Minisola S, Perrier N, Sitges-Serra A, Thakker RV, Guyatt G, Mannstadt M, Potts JT, Clarke BL, Brandi ML. Evaluation and Management of Primary Hyperparathyroidism: Summary Statement and Guidelines from the Fifth International Workshop. J Bone Miner Res 2022; 37:2293-2314
  140. Han ES, Fritton K, Bacon P, Slodzinski MK, Argani C. Preterm Parturient with Polyhydramnios and Pancreatitis: Primary Presentation of Hyperparathyroidism. Case reports in obstetrics and gynecology 2018; 2018:2091082
  141. Herrera-Martinez AD, Bahamondes-Opazo R, Palomares-Ortega R, Munoz-Jimenez C, Galvez-Moreno MA, Quesada Gomez JM. Primary hyperparathyroidism in pregnancy: a two-case report and literature review. Case reports in obstetrics and gynecology 2015; 2015:171828
  142. Ali DS, Dandurand K, Khan AA. Primary Hyperparathyroidism in Pregnancy: Literature Review of the Diagnosis and Management. Journal of clinical medicine 2021; 10
  143. Boyce RW, Varela A, Chouinard L, Bussiere JL, Chellman GJ, Ominsky MS, Pyrah IT. Infant cynomolgus monkeys exposed to denosumab in utero exhibit an osteoclast-poor osteopetrotic-like skeletal phenotype at birth and in the early postnatal period. Bone 2014; 64:314-325
  144. Okamatsu N, Sakai N, Karakawa A, Kouyama N, Sato Y, Inagaki K, Kiuchi Y, Oguchi K, Negishi-Koga T, Takami M. Biological effects of anti-RANKL antibody administration in pregnant mice and their newborns. Biochem Biophys Res Commun 2017; 491:614-621
  145. Rajala B, Abbasi RA, Hutchinson HT, Taylor T. Acute pancreatitis and primary hyperparathyroidism in pregnancy: treatment of hypercalcemia with magnesium sulfate. Obstet Gynecol 1987; 70:460-462
  146. Dahan M, Chang RJ. Pancreatitis secondary to hyperparathyroidism during pregnancy. Obstet Gynecol 2001; 98:923-925
  147. Horjus C, Groot I, Telting D, van Setten P, van Sorge A, Kovacs CS, Hermus A, de Boer H. Cinacalcet for hyperparathyroidism in pregnancy and puerperium. J Pediatr Endocrinol Metab 2009; 22:741-749
  148. Gonzalo Garcia I, Robles Fradejas M, Martin Macias MLA, Biain Ciganda A, Bustinza Beaskoetxea Z, Ruiz Perez E, Fernandez Matia G, Martinez Guisasola J. Primary hyperparathyroidism in pregnancy treated with cinacalcet: a case report. J Obstet Gynaecol 2018; 38:132-134
  149. Vera L, Oddo S, Di Iorgi N, Bentivoglio G, Giusti M. Primary hyperparathyroidism in pregnancy treated with cinacalcet: a case report and review of the literature. Journal of medical case reports 2016; 10:361
  150. Nadarasa K, Bailey M, Chahal H, Raja O, Bhat R, Gayle C, Grossman AB, Druce MR. The use of cinacalcet in pregnancy to treat a complex case of parathyroid carcinoma. Endocrinol Diabetes Metab Case Rep 2014; 2014:140056
  151. Kovacs CS, Ho-Pao CL, Hunzelman JL, Lanske B, Fox J, Seidman JG, Seidman CE, Kronenberg HM. Regulation of murine fetal-placental calcium metabolism by the calcium-sensing receptor. J Clin Invest 1998; 101:2812-2820
  152. Zeng H, Li Z, Zhang X, Wang N, Tian Y, Wang J. Anesthetic management of primary hyperparathyroidism during pregnancy: A case report. Medicine (Baltimore) 2017; 96:e9390
  153. Brown EM, Pollak M, Seidman CE, Seidman JG, Chou YH, Riccardi D, Hebert SC. Calcium-ion-sensing cell-surface receptors. N Engl J Med 1995; 333:234-240
  154. Jones AR, Hare MJ, Brown J, Yang J, Meyer C, Milat F, Allan CA. Familial Hypocalciuric Hypercalcemia in Pregnancy: Diagnostic Pitfalls. JBMR Plus 2020; 4:e10362
  155. Powell BR, Buist NR. Late presenting, prolonged hypocalcemia in an infant of a woman with hypocalciuric hypercalcemia. Clin Pediatr (Phila) 1990; 29:241-243
  156. Thomas BR, Bennett JD. Symptomatic hypocalcemia and hypoparathyroidism in two infants of mothers with hyperparathyroidism and familial benign hypercalcemia. J Perinatol 1995; 15:23-26
  157. Kovacs CS. Hypoparathyroidism during pregnancy, lactation, and fetal/neonatal development. In: Brandi ML, Brown EM, eds. Hypoparathyroidism. Milan: Springer; 2015:249-269.
  158. Khan AA, Clarke B, Rejnmark L, Brandi ML. MANAGEMENT OF ENDOCRINE DISEASE: Hypoparathyroidism in pregnancy: review and evidence-based recommendations for management. Eur J Endocrinol 2019; 180:R37-R44
  159. Sweeney LL, Malabanan AO, Rosen H. Decreased calcitriol requirement during pregnancy and lactation with a window of increased requirement immediately post partum. Endocr Pract 2010; 16:459-462
  160. Graham WP, III., Gordon CS, Loken HF, Blum A, Halden A. Effect of pregnancy and of the menstrual cycle on hypoparathyroidism. J Clin Endocrinol Metab 1964; 24:512-516
  161. Hatswell BL, Allan CA, Teng J, Wong P, Ebeling PR, Wallace EM, Fuller PJ, Milat F. Management of hypoparathyroidism in pregnancy and lactation - A report of 10 cases. Bone Rep 2015; 3:15-19
  162. Hartogsohn EAR, Khan AA, Kjaersulf LU, Sikjaer T, Hussain S, Rejnmark L. Changes in treatment needs of hypoparathyroidism during pregnancy and lactation: A case series. Clin Endocrinol (Oxf) 2020; 93:261-268
  163. Wang JJ, Wang O, Wang YB, Yang J, Song A, Jiang Y, Li M, Xia WB, Xing XP. Changes in serum calcium and treatment of hypoparathyroidism during pregnancy and lactation: A single-center case series. J Clin Endocrinol Metab 2021;
  164. Breslau NA, Zerwekh JE. Relationship of estrogen and pregnancy to calcium homeostasis in pseudohypoparathyroidism. J Clin Endocrinol Metab 1986; 62:45-51
  165. O'Donnell D, Costa J, Meyers AM. Management of pseudohypoparathyroidism in pregnancy. Case report. Br J Obstet Gynaecol 1985; 92:639-641
  166. Saito H, Saito M, Saito K, Terauchi A, Kobayashi T, Tominaga T, Hosoi E, Senoo M, Saito T. Subclinical pseudohypoparathyroidism type II: evidence for failure of physiologic adjustment in calcium metabolism during pregnancy. Am J Med Sci 1989; 297:247-250
  167. Seki K, Osada H, Yasuda T, Sekiya S. Pseudohypoparathyroidism type 1b in pregnancy. Gynecol Obstet Invest1999; 47:278-280
  168. Ochiai D, Uchino H, Ikeda T, Yakubo K, Fukuiya T. Pseudohypoparathyroidism type 1a in pregnancy. J Obstet Gynaecol 2013; 33:900
  169. Guedes Ramallo P, Boix Carreno E. Management of pseudohypoparathyroidism in pregnancy: a case report. J Obstet Gynaecol 2017; 37:519-520
  170. Wang JJ, Yang Y, Wang YB, Song A, Jiang Y, Li M, Xia WB, Liu YP, Wang O, Xing XP. Pseudohypoparathyroidism during pregnancy and the postpartum period: A case series of five patients. Frontiers in endocrinology 2022; 13:1050305
  171. Glass EJ, Barr DG. Transient neonatal hyperparathyroidism secondary to maternal pseudohypoparathyroidism. Arch Dis Child 1981; 56:565-568
  172. Vidailhet M, Monin P, Andre M, Suty Y, Marchal C, Vert P. [Neonatal hyperparathyroidism secondary to maternal hypoparathyroidism]. Arch Fr Pediatr 1980; 37:305-312
  173. Khosla S, van Heerden JA, Gharib H, Jackson IT, Danks J, Hayman JA, Martin TJ. Parathyroid hormone-related protein and hypercalcemia secondary to massive mammary hyperplasia. N Engl J Med 1990; 322:1157
  174. Jackson IT, Saleh J, van Heerden JA. Gigantic mammary hyperplasia in pregnancy associated with pseudohyperparathyroidism. Plastic and reconstructive surgery 1989; 84:806-810
  175. Jodeh W, Sparks PJ, Higgins JM, Tom A, Anilovich N, Moit H, Korff L, Hadad I, Wang X, Imel EA, Donegan DM. Parathyroid hormone-related peptide induced hypercalcemia of pregnancy due to mammary hyperplasia. JBMR Plus 2024; 8:ziae083
  176. Sato K. Hypercalcemia during pregnancy, puerperium, and lactation: review and a case report of hypercalcemic crisis after delivery due to excessive production of PTH-related protein (PTHrP) without malignancy (humoral hypercalcemia of pregnancy). Endocr J 2008; 55:959-966
  177. Marx SJ, Zusman RM, Umiker WO. Benign breast dysplasia causing hypercalcemia. J Clin Endocrinol Metab1977; 45:1049-1052
  178. Eller-Vainicher C, Ossola MW, Beck-Peccoz P, Chiodini I. PTHrP-associated hypercalcemia of pregnancy resolved after delivery: a case report. Eur J Endocrinol 2012; 166:753-756
  179. Kovacs CS, Ward LM. Disorders of Calcium, Phosphorus, and Bone Metabolism During Fetal and Neonatal Development. In: Kovacs CS, Deal CL, eds. Maternal-Fetal and Neonatal Endocrinology. San Diego: Academic Press; 2020:755-782.
  180. Rosen CJ, Adams JS, Bikle DD, Black DM, Demay MB, Manson JE, Murad MH, Kovacs CS. The nonskeletal effects of vitamin D: an Endocrine Society scientific statement. Endocr Rev 2012; 33:456-492
  181. Brooke OG, Brown IR, Bone CD, Carter ND, Cleeve HJ, Maxwell JD, Robinson VP, Winder SM. Vitamin D supplements in pregnant Asian women: effects on calcium status and fetal growth. Br Med J 1980; 280:751-754
  182. Roth DE, Al Mahmud A, Raqib R, Akhtar E, Perumal N, Pezzack B, Baqui AH. Randomized placebo-controlled trial of high-dose prenatal third-trimester vitamin D3 supplementation in Bangladesh: the AViDD trial. Nutrition journal 2013; 12:47
  183. Wagner CL, McNeil R, Hamilton SA, Winkler J, Rodriguez Cook C, Warner G, Bivens B, Davis DJ, Smith PG, Murphy M, Shary JR, Hollis BW. A randomized trial of vitamin D supplementation in 2 community health center networks in South Carolina. Am J Obstet Gynecol 2013; 208:137 e131-113
  184. Dawodu A, Saadi HF, Bekdache G, Javed Y, Altaye M, Hollis BW. Randomized controlled trial (RCT) of vitamin D supplementation in pregnancy in a population with endemic vitamin D deficiency. J Clin Endocrinol Metab 2013; 98:2337-2346
  185. Grant CC, Stewart AW, Scragg R, Milne T, Rowden J, Ekeroma A, Wall C, Mitchell EA, Crengle S, Trenholme A, Crane J, Camargo CA, Jr. Vitamin D during pregnancy and infancy and infant serum 25-hydroxyvitamin D concentration. Pediatrics 2014; 133:e143-153
  186. Vaziri F, Dabbaghmanesh MH, Samsami A, Nasiri S, Shirazi PT. Vitamin D supplementation during pregnancy on infant anthropometric measurements and bone mass of mother-infant pairs: A randomized placebo clinical trial. Early Hum Dev 2016; 103:61-68
  187. Cooper C, Harvey NC, Bishop NJ, Kennedy S, Papageorghiou AT, Schoenmakers I, Fraser R, Gandhi SV, Carr A, D'Angelo S, Crozier SR, Moon RJ, Arden NK, Dennison EM, Godfrey KM, Inskip HM, Prentice A, Mughal MZ, Eastell R, Reid DM, Javaid MK, Group MS. Maternal gestational vitamin D supplementation and offspring bone health (MAVIDOS): a multicentre, double-blind, randomised placebo-controlled trial. The Lancet Diabetes & Endocrinology 2016; 4:393-402
  188. Roth DE, Morris SK, Zlotkin S, Gernand AD, Ahmed T, Shanta SS, Papp E, Korsiak J, Shi J, Islam MM, Jahan I, Keya FK, Willan AR, Weksberg R, Mohsin M, Rahman QS, Shah PS, Murphy KE, Stimec J, Pell LG, Qamar H, Al Mahmud A. Vitamin D Supplementation in Pregnancy and Lactation and Infant Growth. N Engl J Med 2018; 379:535-546
  189. Mojibian M, Soheilykhah S, Fallah Zadeh MA, Jannati Moghadam M. The effects of vitamin D supplementation on maternal and neonatal outcome: A randomized clinical trial. Iranian journal of reproductive medicine 2015; 13:687-696
  190. Wei W, Shary JR, Garrett-Mayer E, Anderson B, Forestieri NE, Hollis BW, Wagner CL. Bone mineral density during pregnancy in women participating in a randomized controlled trial of vitamin D supplementation. Am J Clin Nutr 2017; 106:1422-1430
  191. Kovacs CS, Ward LM. Physiology of Calcium, Phosphorus, and Bone Metabolism During Fetal and Neonatal Development. In: Kovacs CS, Deal CL, eds. Maternal-Fetal and Neonatal Endocrinology. San Diego: Academic Press; 2020:573-586.
  192. Kovacs CS. Analysis of the MAVIDOS trial. The Lancet Diabetes & Endocrinology 2017; 5:328
  193. Khaing W, Vallibhakara SA, Tantrakul V, Vallibhakara O, Rattanasiri S, McEvoy M, Attia J, Thakkinstian A. Calcium and Vitamin D Supplementation for Prevention of Preeclampsia: A Systematic Review and Network Meta-Analysis. Nutrients 2017; 9
  194. De-Regil LM, Palacios C, Lombardo LK, Pena-Rosas JP. Vitamin D supplementation for women during pregnancy. Cochrane Database Syst Rev 2016; 1:CD008873
  195. Hypponen E, Cavadino A, Williams D, Fraser A, Vereczkey A, Fraser WD, Banhidy F, Lawlor D, Czeizel AE. Vitamin D and pre-eclampsia: original data, systematic review and meta-analysis. Annals of nutrition & metabolism2013; 63:331-340
  196. Thorne-Lyman A, Fawzi WW. Vitamin D during pregnancy and maternal, neonatal and infant health outcomes: a systematic review and meta-analysis. Paediatric and perinatal epidemiology 2012; 26 Suppl 1:75-90
  197. Roth DE, Leung M, Mesfin E, Qamar H, Watterworth J, Papp E. Vitamin D supplementation during pregnancy: state of the evidence from a systematic review of randomised trials. BMJ 2017; 359:j5237
  198. Harvey NC, Holroyd C, Ntani G, Javaid K, Cooper P, Moon R, Cole Z, Tinati T, Godfrey K, Dennison E, Bishop NJ, Baird J, Cooper C. Vitamin D supplementation in pregnancy: a systematic review. Health Technol Assess 2014; 18:1-190
  199. Palacios C, Kostiuk LK, Pena-Rosas JP. Vitamin D supplementation for women during pregnancy. Cochrane Database Syst Rev 2019; 7:Cd008873
  200. Edouard T, Alos N, Chabot G, Roughley P, Glorieux FH, Rauch F. Short- and long-term outcome of patients with pseudo-vitamin D deficiency rickets treated with calcitriol. J Clin Endocrinol Metab 2011; 96:82-89
  201. Marx SJ, Swart EG, Jr., Hamstra AJ, DeLuca HF. Normal intrauterine development of the fetus of a woman receiving extraordinarily high doses of 1,25-dihydroxyvitamin D3. J Clin Endocrinol Metab 1980; 51:1138-1142
  202. Malloy PJ, Tiosano D, Feldman D. Hereditary 1,25-Dihydroxyvitamin-D-Resistant Rickets. In: Feldman D, Pike JW, Adams JS, eds. Vitamin D, Third Edition. San Diego, CA: Academic Press; 2011:1197-1232.
  203. Carpenter TO. CYP24A1 loss of function: Clinical phenotype of monoallelic and biallelic mutations. J Steroid Biochem Mol Biol 2017; 173:337-340
  204. Maekawa AS, Walker EC, Bennin D, Hartery SA, Kirby BJ, St-Arnaud R, Sims NA, Kovacs CS. Maternal loss of 24-hydroxylase causes increased intestinal calcium absorption and hypercalcemia during pregnancy but reduced skeletal resorption during lactation in mice. J Bone Miner Res 2024; 39:zjae166
  205. Shah AD, Hsiao EC, O'Donnell B, Salmeen K, Nussbaum R, Krebs M, Baumgartner-Parzer S, Kaufmann M, Jones G, Bikle DD, Wang Y, Mathew AS, Shoback D, Block-Kurbisch I. Maternal Hypercalcemia Due to Failure of 1,25-Dihydroxyvitamin-D3 Catabolism in a Patient With CYP24A1 Mutations. J Clin Endocrinol Metab 2015; 100:2832-2836
  206. Dinour D, Davidovits M, Aviner S, Ganon L, Michael L, Modan-Moses D, Vered I, Bibi H, Frishberg Y, Holtzman EJ. Maternal and infantile hypercalcemia caused by vitamin-D-hydroxylase mutations and vitamin D intake. Pediatr Nephrol 2015; 30:145-152
  207. Woods GN, Saitman A, Gao H, Clarke NJ, Fitzgerald RL, Chi NW. A Young Woman With Recurrent Gestational Hypercalcemia and Acute Pancreatitis Caused by CYP24A1 Deficiency. J Bone Miner Res 2016; 31:1841-1844
  208. Kwong WT, Fehmi SM. Hypercalcemic Pancreatitis Triggered by Pregnancy With a CYP24A1 Mutation. Pancreas2016; 45:e31-32
  209. Brancatella A, Cappellani D, Kaufmann M, Semeraro A, Borsari S, Sardella C, Baldinotti F, Caligo MA, Jones G, Marcocci C, Cetani F. Long-term Efficacy and Safety of Rifampin in the Treatment of a Patient Carrying a CYP24A1 Loss-of-Function Variant. J Clin Endocrinol Metab 2022; 107:e3159-e3166
  210. Hawkes CP, Li D, Hakonarson H, Meyers KE, Thummel KE, Levine MA. CYP3A4 Induction by Rifampin: An Alternative Pathway for Vitamin D Inactivation in Patients With CYP24A1 Mutations. J Clin Endocrinol Metab 2017; 102:1440-1446
  211. Prentice A. Micronutrients and the bone mineral content of the mother, fetus and newborn. J Nutr 2003; 133:1693S-1699S
  212. O'Brien EC, Geraghty AA, Kilbane MT, McKenna MJ, McAuliffe FM. Bone resorption and dietary calcium in pregnancy-a window to future maternal bone health. Osteoporos Int 2021; 32:1803-1814
  213. Prentice A. Pregnancy and lactation. In: Glorieux FH, Petifor JM, Jüppner H, eds. Pediatric bone: biology & diseases. New York: Academic Press; 2003:249-269.
  214. Janakiraman V, Ettinger A, Mercado-Garcia A, Hu H, Hernandez-Avila M. Calcium supplements and bone resorption in pregnancy: a randomized crossover trial. Am J Prev Med 2003; 24:260-264
  215. Koo WW, Walters JC, Esterlitz J, Levine RJ, Bush AJ, Sibai B. Maternal calcium supplementation and fetal bone mineralization. Obstet Gynecol 1999; 94:577-582
  216. Prentice A. Calcium in pregnancy and lactation. Annu Rev Nutr 2000; 20:249-272
  217. Hofmeyr GJ, Atallah AN, Duley L. Calcium supplementation during pregnancy for preventing hypertensive disorders and related problems. Cochrane Database Syst Rev 2006; 3:CD001059
  218. Kumar A, Devi SG, Batra S, Singh C, Shukla DK. Calcium supplementation for the prevention of pre-eclampsia. Int J Gynaecol Obstet 2009; 104:32-36
  219. Hiller JE, Crowther CA, Moore VA, Willson K, Robinson JS. Calcium supplementation in pregnancy and its impact on blood pressure in children and women: follow up of a randomised controlled trial. Aust N Z J Obstet Gynaecol2007; 47:115-121
  220. Villar J, Abdel-Aleem H, Merialdi M, Mathai M, Ali MM, Zavaleta N, Purwar M, Hofmeyr J, Nguyen TN, Campodonico L, Landoulsi S, Carroli G, Lindheimer M. World Health Organization randomized trial of calcium supplementation among low calcium intake pregnant women. Am J Obstet Gynecol 2006; 194:639-649
  221. Ross AC, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Manson JE, Mayne ST, Rosen CJ, Shapses SA. Dietary Reference Intakes for Calcium and Vitamin D.Washington, DC: Institute of Medicine, 2011.
  222. Ohata Y, Yamazaki M, Kawai M, Tsugawa N, Tachikawa K, Koinuma T, Miyagawa K, Kimoto A, Nakayama M, Namba N, Yamamoto H, Okano T, Ozono K, Michigami T. Elevated fibroblast growth factor 23 exerts its effects on placenta and regulates vitamin D metabolism in pregnancy of Hyp mice. J Bone Miner Res 2014; 29:1627-1638
  223. Jonas AJ, Dominguez B. Low breast milk phosphorus concentration in familial hypophosphatemia. J Pediatr Gastroenterol Nutr 1989; 8:541-543
  224. Reade TM, Scriver CR. Hypophosphatemic rickets and breast milk. N Engl J Med 1979; 300:1397
  225. European Medicines Agency. Crysvita: Annex I: Summary of Product Characteristics. 2022; https://www.ema.europa.eu/en/documents/product-information/crysvita-epar-product-information_en.pdf. Accessed 22 October 2023.
  226. Greer FR, Lane J, Ho M. Elevated serum parathyroid hormone, calcitonin, and 1,25-dihydroxyvitamin D in lactating women nursing twins. Am J Clin Nutr 1984; 40:562-568
  227. Dobnig H, Kainer F, Stepan V, Winter R, Lipp R, Schaffer M, Kahr A, Nocnik S, Patterer G, Leb G. Elevated parathyroid hormone-related peptide levels after human gestation: relationship to changes in bone and mineral metabolism. J Clin Endocrinol Metab 1995; 80:3699-3707
  228. Yamamoto M, Duong LT, Fisher JE, Thiede MA, Caulfield MP, Rosenblatt M. Suckling-mediated increases in urinary phosphate and 3',5'-cyclic adenosine monophosphate excretion in lactating rats: possible systemic effects of parathyroid hormone-related protein. Endocrinology 1991; 129:2614-2622
  229. VanHouten JN, Dann P, Stewart AF, Watson CJ, Pollak M, Karaplis AC, Wysolmerski JJ. Mammary-specific deletion of parathyroid hormone-related protein preserves bone mass during lactation. J Clin Invest 2003; 112:1429-1436
  230. Kovacs CS, Chik CL. Hyperprolactinemia caused by lactation and pituitary adenomas is associated with altered serum calcium, phosphate, parathyroid hormone (PTH), and PTH-related peptide levels. J Clin Endocrinol Metab1995; 80:3036-3042
  231. Sowers MF, Hollis BW, Shapiro B, Randolph J, Janney CA, Zhang D, Schork A, Crutchfield M, Stanczyk F, Russell-Aulet M. Elevated parathyroid hormone-related peptide associated with lactation and bone density loss. JAMA 1996; 276:549-554
  232. Kelly PA, Bachelot A, Kedzia C, Hennighausen L, Ormandy CJ, Kopchick JJ, Binart N. The role of prolactin and growth hormone in mammary gland development. Mol Cell Endocrinol 2002; 197:127-131
  233. Dawood MY, Khan-Dawood FS, Wahi RS, Fuchs F. Oxytocin release and plasma anterior pituitary and gonadal hormones in women during lactation. J Clin Endocrinol Metab 1981; 52:678-683
  234. Nishimori K, Young LJ, Guo Q, Wang Z, Insel TR, Matzuk MM. Oxytocin is required for nursing but is not essential for parturition or reproductive behavior. Proc Natl Acad Sci U S A 1996; 93:11699-11704
  235. VanHouten JN, Wysolmerski JJ. Low estrogen and high parathyroid hormone-related peptide levels contribute to accelerated bone resorption and bone loss in lactating mice. Endocrinology 2003; 144:5521-5529
  236. Hernandez LL, Gregerson KA, Horseman ND. Mammary gland serotonin regulates parathyroid hormone-related protein and other bone-related signals. Am J Physiol Endocrinol Metab 2012; 302:E1009-1015
  237. Horseman ND, Hernandez LL. New concepts of breast cell communication to bone. Trends Endocrinol Metab2014; 25:34-41
  238. Kolthoff N, Eiken P, Kristensen B, Nielsen SP. Bone mineral changes during pregnancy and lactation: a longitudinal cohort study. Clin Sci (Lond) 1998; 94:405-412
  239. Polatti F, Capuzzo E, Viazzo F, Colleoni R, Klersy C. Bone mineral changes during and after lactation. Obstet Gynecol 1999; 94:52-56
  240. Kalkwarf HJ, Specker BL, Bianchi DC, Ranz J, Ho M. The effect of calcium supplementation on bone density during lactation and after weaning. N Engl J Med 1997; 337:523-528
  241. Cross NA, Hillman LS, Allen SH, Krause GF. Changes in bone mineral density and markers of bone remodeling during lactation and postweaning in women consuming high amounts of calcium. J Bone Miner Res 1995; 10:1312-1320
  242. Laskey MA, Prentice A, Hanratty LA, Jarjou LM, Dibba B, Beavan SR, Cole TJ. Bone changes after 3 mo of lactation: influence of calcium intake, breast-milk output, and vitamin D-receptor genotype. Am J Clin Nutr 1998; 67:685-692
  243. Teti A, Zallone A. Do osteocytes contribute to bone mineral homeostasis? Osteocytic osteolysis revisited. Bone2009; 44:11-16
  244. Qing H, Ardeshirpour L, Pajevic PD, Dusevich V, Jahn K, Kato S, Wysolmerski J, Bonewald LF. Demonstration of osteocytic perilacunar/canalicular remodeling in mice during lactation. J Bone Miner Res 2012; 27:1018-1029
  245. Kirby BJ, Ardeshirpour L, Woodrow JP, Wysolmerski JJ, Sims NA, Karaplis AC, Kovacs CS. Skeletal recovery after weaning does not require PTHrP. J Bone Miner Res 2011; 26:1242-1251
  246. Collins JN, Kirby BJ, Woodrow JP, Gagel RF, Rosen CJ, Sims NA, Kovacs CS. Lactating Ctcgrp nulls lose twice the normal bone mineral content due to fewer osteoblasts and more osteoclasts, whereas bone mass is fully restored after weaning in association with up-regulation of Wnt signaling and other novel genes. Endocrinology2013; 154:1400-1413
  247. Liu XS, Ardeshirpour L, VanHouten JN, Shane E, Wysolmerski JJ. Site-specific changes in bone microarchitecture, mineralization, and stiffness during lactation and after weaning in mice. J Bone Miner Res 2012; 27:865-875
  248. Bjornerem A, Ghasem-Zadeh A, Wang X, Bui M, Walker SP, Zebaze R, Seeman E. Irreversible Deterioration of Cortical and Trabecular Microstructure Associated With Breastfeeding. J Bone Miner Res 2017; 32:681-687
  249. Brembeck P, Lorentzon M, Ohlsson C, Winkvist A, Augustin H. Changes in cortical volumetric bone mineral density and thickness, and trabecular thickness in lactating women postpartum. J Clin Endocrinol Metab 2015; 100:535-543
  250. Wiklund PK, Xu L, Wang Q, Mikkola T, Lyytikainen A, Volgyi E, Munukka E, Cheng SM, Alen M, Keinanen-Kiukaanniemi S, Cheng S. Lactation is associated with greater maternal bone size and bone strength later in life. Osteoporos Int 2012; 23:1939-1945
  251. Bowman BM, Miller SC. Skeletal adaptations during mammalian reproduction. J Musculoskelet Neuronal Interact2001; 1:347-355
  252. Vajda EG, Bowman BM, Miller SC. Cancellous and cortical bone mechanical properties and tissue dynamics during pregnancy, lactation, and postlactation in the rat. Biol Reprod 2001; 65:689-695
  253. Reid IR, Wattie DJ, Evans MC, Budayr AA. Post-pregnancy osteoporosis associated with hypercalcaemia. Clin Endocrinol (Oxf) 1992; 37:298-303
  254. VanHouten J, Dann P, McGeoch G, Brown EM, Krapcho K, Neville M, Wysolmerski JJ. The calcium-sensing receptor regulates mammary gland parathyroid hormone-related protein production and calcium transport. J Clin Invest 2004; 113:598-608
  255. Ardeshirpour L, Dann P, Pollak M, Wysolmerski J, VanHouten J. The calcium-sensing receptor regulates PTHrP production and calcium transport in the lactating mammary gland. Bone 2006; 38:787-793
  256. Caplan RH, Beguin EA. Hypercalcemia in a calcitriol-treated hypoparathyroid woman during lactation. Obstet Gynecol 1990; 76:485-489
  257. Salle BL, Berthezene F, Glorieux FH, Delvin EE, Berland M, David L, Varenne JP, Putet G. Hypoparathyroidism during pregnancy: treatment with calcitriol. J Clin Endocrinol Metab 1981; 52:810-813
  258. Sadeghi-Nejad A, Wolfsdorf JI, Senior B. Hypoparathyroidism and pregnancy. Treatment with calcitriol. JAMA1980; 243:254-255
  259. Shomali ME, Ross DS. Hypercalcemia in a woman with hypoparathyroidism associated with increased parathyroid hormone-related protein during lactation. Endocr Pract 1999; 5:198-200
  260. Mather KJ, Chik CL, Corenblum B. Maintenance of serum calcium by parathyroid hormone-related peptide during lactation in a hypoparathyroid patient. J Clin Endocrinol Metab 1999; 84:424-427
  261. Rideout KL. When endocrine disorders disrupt pregnancy: perspectives of affected mothers. In: Kovacs CS, Deal CL, eds. Maternal-Fetal and Neonatal Endocrinology: Physiology, Pathophysiology, and Clinical Management. San Diego: Academic Press; 2019:xxxvii-xli.
  262. Wagner CL, Hulsey TC, Fanning D, Ebeling M, Hollis BW. High-dose vitamin D3 supplementation in a cohort of breastfeeding mothers and their infants: a 6-month follow-up pilot study. Breastfeed Med 2006; 1:59-70
  263. Tan ML, Abrams SA, Osborn DA. Vitamin D supplementation for term breastfed infants to prevent vitamin D deficiency and improve bone health. Cochrane Database Syst Rev 2020; 12:Cd013046
  264. Prentice A, Jarjou LM, Cole TJ, Stirling DM, Dibba B, Fairweather-Tait S. Calcium requirements of lactating Gambian mothers: effects of a calcium supplement on breast-milk calcium concentration, maternal bone mineral content, and urinary calcium excretion. Am J Clin Nutr 1995; 62:58-67
  265. Prentice A, Jarjou LM, Stirling DM, Buffenstein R, Fairweather-Tait S. Biochemical markers of calcium and bone metabolism during 18 months of lactation in Gambian women accustomed to a low calcium intake and in those consuming a calcium supplement. J Clin Endocrinol Metab 1998; 83:1059-1066
  266. Prentice A, Yan L, Jarjou LM, Dibba B, Laskey MA, Stirling DM, Fairweather-Tait S. Vitamin D status does not influence the breast-milk calcium concentration of lactating mothers accustomed to a low calcium intake. Acta Paediatr 1997; 86:1006-1008
  267. Bezerra FF, Mendonca LM, Lobato EC, O'Brien KO, Donangelo CM. Bone mass is recovered from lactation to postweaning in adolescent mothers with low calcium intakes. Am J Clin Nutr 2004; 80:1322-1326
  268. Chantry CJ, Auinger P, Byrd RS. Lactation among adolescent mothers and subsequent bone mineral density. Arch Pediatr Adolesc Med 2004; 158:650-656
  269. Delzer PR, Meyer RA, Jr. Normal milk composition in lactating X-linked hypophosphatemic mice despite continued hypophosphatemia. Calcif Tissue Int 1983; 35:750-754
  270. Burosumab. Drugs and Lactation Database (LactMed®). Bethesda (MD): National Institute of Child Health and Human Development; 2006.

Empty Sella

ABSTRACT

 

Empty sella is a radiological finding of a flattened pituitary in a sellar space filled with cerebrospinal fluid. It may be primary or secondary consequent to various processes causing injury and shrinkage of the pituitary gland (postpartum hemorrhage, pituitary surgery, irradiation, apoplexy, infection, head trauma, hypophysitis, etc.). The mechanisms involved in the pathogenesis of the so called “primary empty sella” may range from continuously or intermittently increased intracranial pressure due to idiopathic benign intracranial hypertension, obesity, arterial hypertension, or multiple pregnancies in female patients with accompanying insufficiency of the sellar diaphragm and changes in pituitary gland volume (hyperplasia during pregnancy, lactation, menopause etc.). Primary empty sella can be an incidental radiological finding in an asymptomatic patient with preserved pituitary function. In symptomatic patients with the so called “empty sella syndrome” (headache, visual disturbances, and hormonal dysfunction), the radiological finding of an empty sella is important in the differential diagnosis of other sellar lesions. Hypopituitarism, partial or complete, and hyperprolactinemia are not uncommon in these patients. The treatment of hypopituitarism and hyperprolactinemia is advocated in all patients with confirmatory results. In patients with the secondary empty sella, hypopituitarism is more common and more readily recognized due to damage caused by surgery, radiation therapy, or various pathological causes. Rarely, an empty sella can be associated with hormonal hypersecretion from an “invisible” micro adenoma producing prolactin, growth hormone (acromegaly), or ACTH (Cushing’s disease). A wide range of radiological findings in patients with secondary and primary empty sella coupled with clinical data (important hints from the history and data on endocrine function) are presented for further illustration of this topic.

 

HISTORY

 

The term ‘empty sella’ was first used by Bush in 1951 (1) to describe a peculiar anatomical condition, observed in 40 of 788 human cadavers, particularly females, characterized by a sella turcica with an incomplete diaphragm sellae that forms only a small peripheral rim, with a pituitary gland not absent, but flattened in such a manner as to form a thin layer of tissue at the bottom of the sella turcica. Kaufman (2), in 1968, speculated that ‘…empty sella is a distinct anatomical and radiographic entity, function of an incompleteness of the diaphragma sellae and of the cerebrospinal fluid (CSF) pressure, normal or elevated’. The role of the normal fluctuations of CSF pressures and the effect of a superimposed prolonged increase in CSF pressure were related to the anatomic changes involving the bony wall of the empty sella.

 

DEFINITION, ETIOLOGY, AND PREVALENCE OF EMPTY SELLA

 

Empty sella is defined as herniation of subarachnoid space into the sella turcica (arachnoidocele). It is a term for the radiological finding of “empty sellar space” on magnetic resonance imaging (MRI) and computerized tomography (CT) with a flattened pituitary and elongated stalk. It can be partial if less than 50% of sellar space is filled with cerebro-spinal fluid (CSF), or complete if CSF fills more than 50% of space in the sella and gland thickness is less than 2mm (3,4).

 

Regarding its etiology, it can be primary if there is no pathological process in the sellar region preceding the pituitary damage or secondary if it is consequent to a specific pathological process. Primary empty sella (PES) can be an incidental finding or may arise during imaging for headache, endocrine disorders, neurological symptoms, visual disturbances, abnormal sella turcica radiograph, and other reasons.

 

Primary empty sella (PES) can be caused by intracranial hypertension and/or insufficiency of the sellar diaphragm in subjects with no previous history of pituitary disease. Insufficiency of the sellar diaphragm, a deflection of dura matter separating the suprasellar cistern from the pituitary fossa, allows unobstructed pulsatile movements of CSF from chiasmatic cistern causing flattening of the pituitary to the sellar floor. In extreme cases bone erosion of the sellar floor and CSF leak (rhinorrhea) may occur, increasing the risk of meningitis. Partial or complete absence of the sellar diaphragm has been demonstrated in patients with PES.

 

Intracranial pressure can be intermittently increased due to obesity, sleep apnea, arterial hypertension, pregnancy, and labor. Intracranial hypertension may be idiopathic or associated with other intracranial processes such as tumors, venous thrombosis, infections, or malformations. Idiopathic intracranial hypertension (IIH) or “pseudo-tumor cerebri” is a rare condition affecting 1 in 100,000 persons. It can be due to impaired CSF absorption, increased CSF secretion, and/or increased capillary permeability (5). Impaired CSF dynamics and absorption have been found in up to 77% and 84% of patients with PES, respectively (6), The prevalence of PES is very high in patients with IIH ranging from 70-94% (4).

 

Changes in the pituitary gland volume may also be involved in the pathogenesis of empty sella syndrome including hyperplasia during pregnancy and lactation and pituitary involution after menopause accounting for the significantly higher prevalence of this condition in female patients (female to male ratio 5:1).

 

Factors involved in the pathogenesis of primary empty sella are shown in Figure 1.

 

Secondary empty sella is more common and is related to various pathological processes of the sellar region. Among many causes, pituitary tumor shrinkage occurring after medical treatment, surgery, radiotherapy, and apoplexy of a pituitary adenoma are frequent causes of secondary empty sella. Likewise, postpartum pituitary necrosis, pituitary infection, hypophysitis, and traumatic brain injury may lead to pituitary atrophy. The diagnosis of secondary empty sella is more difficult if there is no known underlying pathology involving the pituitary gland. In these cases, the sella is normal in size, and the function of the flattened pituitary gland may or may not be compromised. Such is the case in congenital causes of hypopituitarism both acquired and genetic, presenting with a hypoplastic pituitary gland and ectopic posterior lobe (7). Large intracranial tumors such as slow-growing meningiomas can also cause increased intracranial pressure and secondary empty sella in a significant number of patients. Pituitary MRI images of patients with secondary and primary empty sella are presented in the section dedicated to radiological appearance of an empty sella (Fig. 2-6). Associated risk factors for primary and secondary empty sella are shown in Table 1.

 

The reported prevalence of empty sella depends on techniques used for detection. In autopsy studies empty sella has been found in up to 5.5-12% cases (1,2), On imaging the overall incidence has been estimated at 12% (4). The prevalence of PES is very high in patients with IIH (8)

 

Table 1. Associated Risk Factors for Primary and Secondary Empty Sella

Primary Empty Sella (PES)

Secondary Empty Sella (SES)

Female sex

Multiple pregnancies

Obesity and sleep apnea

Arterial hypertension

Benign intracranial hypertension

 

Medical therapy

Pituitary surgery

Irradiation

Pituitary apoplexy

Sheehan’s syndrome

Traumatic brain injury

Congenital hypopituitarism

 

Figure 1. Factors involved in pathogenesis of primary empty sella include the upper-sellar factors, incompetence or incomplete formation of the sellar diaphragm, and pituitary factors associated with the variation in the pituitary volume. Modified from Bioscientifica Ltd., Chiloiro S et al, European Journal of Endocrinology (2017) 177, R275–R285

 

RADIOLOGICAL APPEARANCE OF EMPTY SELLA

 

Very commonly an empty sella is incidentally discovered during MRI or CT imaging or during evaluation for headache, endocrine, neurological or visual disturbances. Less commonly it is observed after additional imaging for abnormal sella turcica radiographs. Chronic intracranial hypertension can lead to sellar remodeling and enlargement with thinning of the sellar floor and rarely rhinorrhea.

 

On typical presentation CSF filling is in continuity with overlying subarachnoid spaces and the residual pituitary gland is flattened against the sellar floor of an enlarged bony sella with pituitary volume usually less than 611.21 mm3 (9). The differential diagnosis with cystic lesions and congenital pituitary abnormalities may pose a challenge. The stalk is usually thinned and located in the midline. Asymmetry is a frequent sign of the secondary empty sella. In rare cases the chiasm can herniate into the sella in cases of both primary and secondary etiology. Indirect signs of intracranial hypertension may also be present such as flattening of the posterior sclera, prominent subarachnoid spaces along the optic nerves, vertical tortuosity of the optic nerve sheath complex, and increased width of the optic nerve sheaths (10).

 

Sagittal and coronal T1-weighted (T1W) contrast enhanced images and coronal T2-weighted (T2W) images are strongly indicated for MR studies because they show CSF within the sella. On FLAIR sequences the intrasellar fluid completely suppresses and it presents without restriction in DWI sequences. After contrast T1W images show normal enhancement of residual pituitary gland and the stalk in PES in contrast to scaring and distortion in SES.

 

In patients with congenital hypopituitarism an ectopic posterior pituitary, stalk duplication or absence may be present in combination with other midline defects (agenesis of corpus callosum, supraoptic dysplasia, etc.) (7).

 

Figures 2, 3, 4, 5 represent various MRI findings of patients with secondary empty sella due to postpartum hemorrhage (Sheehan’s syndrome Fig. 2), lymphocytic hypophysitis (Fig. 3), shrinkage of a macroprolactinoma after successful treatment with cabergoline (Fig.4), and congenital hypopituitarism with empty sella (Fig.5)

 

Figure 2. Sagittal T1W images of two patients with empty sella and Sheehan’s syndrome
Left Panel (sagittal T1W image without contrast) and Right (sagittal T1W image after contrast enhancement) represent the MRI appearance of empty sella in two patients with Sheehan’s syndrome. Both patients presented with hyponatremic coma due to unrecognized panhypopituitarism and infection 3 and 20 years after delivery complicated by postpartum hemorrhage.

Figure 3. Contrast enhanced coronal and sagittal T1W images of lymphocytic hypophysitis spontaneous evolution from the presentation (panel A, B), after 4 (panel C) and 10 years (panel D) of follow-up resulting in secondary empty sella.

Figure 4. Coronal T1W images demonstrating a pituitary tumor (macroprolactinoma) shrinkage in a patient treated with the dopamine agonist cabergoline for 10 years. The patient presented with hyperprolactinemia causing galactorrhea-amenorrhea, secondary adrenal insufficiency and central hypothyroidism. Hyperprolactinemia and adrenal insufficiency completely recovered during follow up.

Figure 5. Sagittal T1W showing small pituitary gland found at the bottom of sella, thin stalk and ectopic posterior lobe in a young adult with isolated childhood-onset growth hormone deficiency (congenital).

Figure 6. Sagittal T1W images of pituitary stalk pressed against the dorsum sellae causing mild hyperprolactinemia in a patient with primary empty sella.

 

PRIMARY EMPTY SELLA (PES) AND EMPTY SELLA SYNDROME

 

Epidemiologically, this is associated with female sex (female to male ratio 5:1), multiple pregnancies, obesity, arterial hypertension, and middle age. It may present with headaches, endocrine dysfunction, and visual disturbances due to pressure on the neighboring structures.

A typical clinical picture consists of headache and obesity. Women in the reproductive age may be affected by menstrual irregularities, galactorrhea, and sterility. Man can develop gynecomastia and sexual disturbances. Primary empty sella due to the syndrome of increased intracranial pressure can also be associated with symptoms of intracranial hypertension.

 

Pathogenesis of Primary Empty Sella (PES)

 

Pregnancy may trigger the onset of PES. It is associated with pituitary hyperplasia and CSF hypertension especially in multiple pregnancies. PES has also been associated with CSF hypertension related to obesity and arterial hypertension. In the largest study with 175 patients, multiple pregnancies were reported in 58.3% women with PES, while obesity and arterial hypertension were recorded in 49.5% and 27.3% of patients (10). In patients with benign intracranial hypertension, empty sella is a common finding (8, 11).

 

Endocrine Dysfunction in PES

 

Hyperprolactinemia, usually mild (less than 50 ng/ml), is present in approximately 10% of patients (10). It is often due to increased pressure of the CSF on the pituitary stalk and diminished dopamine inhibitory effect. Prolactin dynamics in PES may be influenced by gonadal status, intracranial pressure, neurotransmitters, and stalk integrity. Rarely, pituitary microadenomas causing acromegaly and Cushing’s disease may be associated with empty sella. In the recent study by Himes et al. empty sella was associated with an increased rate of MRI negative Cushing’s disease (12). Pituitary compression causing a relative reduction in the volume of the pituitary for imaging is a plausible cause for not detecting the tumor mass with MRI (12). The presence of ES was associated with lower preoperative prolactin and nadir GH responses to OGTT in patients with acromegaly (13) Functional imaging, if available may identify occult pituitary tumors as therapeutic targets in such settings.

 

The prevalence of hypopituitarism in patients with PES is variable. In a study by Guitelman et al. it was found in 28% of patients (11). Panhypopituitarism was present in 40% of these patients, while partial or isolated hormone deficiencies were diagnosed in 60% of hypopituitary patients (11). The most prevalent pituitary deficiency was growth hormone deficiency.

 

In a pooled meta-analysis, which included four studies, the frequency of hypopituitarism was 52% (14). Multiple pituitary hormone deficiencies were present in 30%, isolated in 21% of patients with PES. Growth hormone and gonadotropins were the most common isolated insufficiencies (14). Significant correlation was reported between IGF-1 values and pituitary volume measurements (15).

 

In a retrospective single-center study of 765 patients by Ekhzaimy et al. 79% of PES was diagnosed incidentally on MRI. The majority of patients were evaluated by general practitioners with suboptimal hormonal evaluation while only 20 % were referred to endocrinologists for hormonal evaluation (16).

 

A multicenter retrospective study from Italy, detected hormonal alterations in 29% of incidental PES suggesting the need for careful initial evaluation since on follow-up hormonal deterioration was uncommon (3%) (17). Hypopituitarism was associated with male sex (17, 18) and radiological finding of complete PES (18).

 

Hormonal assessment is advocated in all patients with ES. In case of borderline results or suspected isolated or partial insufficiency stimulatory tests are recommended if clinically relevant for hormone replacement.

 

Other symptoms in patients with ES include headache and visual and neurological disturbances. In patients with intracranial hypertension these symptoms are more common.

 

Neurological and Ophthalmic Dysfunction in PES

 

Headache is present in approximately 80% (3,6). In 20% of patients it may be accompanied by visual disturbances, even papilledema in intracranial hypertension (3,6).

 

Visual disturbances including worsening of visual acuity, blurred vision, diplopia, defects of oculomotor nerve, and optical neuritis were also reported in patients with PES. In case of benign intracranial hypertension ophthalmic echography and computerized visual field with evoked potentials should be performed in consultation with ophthalmologists. Study by Yilmaz et al. evaluated retinal optic nerve fiber thickness by optic coherence tomography in asymptomatic patients with empty sella and healthy controls (19). Reduced values in asymptomatic patients provide valuable data for monitoring of these patients (19). In case of chiasmal herniation into the empty sella with acute visual deterioration, new neurosurgical techniques of chiasmal transsphenoidal elevation are available (20).

 

Neurological disturbances including dizziness, syncope, cranial nerve disorders, convulsions, and depression were reported in approximately 40% of patients with PES (3, 6). Rhinorrhea increases the risk of meningitis and surgery involving osseous remodeling techniques (21).

An association of PES with periventricular white matter hyperintensities, and enlarged perivascular spaces, common features of cerebral small vessel disease, were reported confirming the previous clinical observation and possible common underlying mechanism (22).

 

TREATMENT

 

In patients with increased idiopathic intracranial pressure osmotic diuretics or acetazolamide (Diamox) are advocated. Weight loss may be helpful in obese and overweight patients especially if accompanied by sleep apnea. Neurosurgical techniques may be indicated for rhinorrhea and some symptomatic secondary causes of empty sella syndrome with increased intracranial pressure and acute visual disturbances. If hypopituitarism is present it should be treated following the current recommendations (23) and hyperprolactinemia treated with dopamine agonists.

 

CONCLUSION

 

Primary empty sella can be heterogeneous in origin and presentations range from an asymptomatic incidental radiological finding to endocrine and neuro-ophthalmological manifestations. Female sex, multiple pregnancies, obesity, and arterial hypertension are associated risk factors as well as the syndrome of benign intracranial hypertension. Secondary empty sella is caused by various pathological processes resulting in shrinkage of the pituitary gland. Routine hormonal status assessment and regular follow-up are indicated in all patients since the prevalence of pituitary dysfunction is significant.

 

REFERENCES

 

  1. Busch W. Die Morphologie der Sella turcica und ihre Beziehungen zur Hypophyse. Virchows Archiv für Pathologische Anatomie und Physiologie und für klinische Medizin 1951; 320: 437–458.
  2. Kaufman B. The ‘empty’ sella turcica-a manifestation of the intrasellar subarachnoid space. Radiology 1968; 90: 931–941.
  3. De Marinis L, Bonadonna S, Bianchi A, Giulio M, Gustina A. Extensive clinical experience: primary empty sella. Journal of Clinical Endocrinology and Metabolism 2005:5471-5477.
  4. Chiloiro S, Giampietro A. Bianchi A, Tartaglione T, Capobianco A, Anile C, De Marinis L. Primary empty sella: a comprehensive review. European Journal of Endocrinology 2017; 177:6; R275-R285.
  5. Friedman DI, Jacobson DM. Diagnostic criteria for idiopathic intracranial hypertension. Neurology2002; 59: 1492–1495.
  6. Maira G, Anile C, Mangiola A. Primary empty sella syndrome in a series of 142 patients. Journal of Neurosurery 2005; 103: 831–836.
  7. Besci Ö ,Yaşar E, Mert Erbaş I, Yüksek Acınıklı K, Korcan Demir K, Böber E, Abacı A. Clinical course of primary empty sella in children: a single center experience. The Turkish Journal of Pediatrics 2022; 64: 900-908
  8. Bhawna S, Naveen S, Vikas S, Ashok P, Goel Clinical and Radiological Profile of 122 Cases of Idiopathic Intracranial Hypertension in a Tertiary Care Centre of India. Neurology India 2022; 70(2):704-709.
  9. Hoffmann J, Schmidt C, Kunte H, Klingebiel R, Harms L, Huppertz HJ, Lüdemann L & Wiener E. Volumetric assessment of optic nerve sheath and hypophysis in idiopathic intracranial hypertension. American Journal of Neuroradiology 2014;35: 513–518.
  10. Degnan AJ, Levy LM. Pseudotumor cerebri: brief review of clinical syndrome and imaging findings. American Journal of Neuroradiology 2012; doi 10.3174/ajnrA2.404
  11. Guitelman M, Basalvibaso NG, Vitale M, Chervin A, Katz D, Miragaya K, Herrera J, Cornalo D, Servido M, Boero L, Manavela M, Danilowicz K, Alfieri A, Stalldecker G, Glerean M, Fainstein Day P, Ballarino C, Mallea Gil MS, Rogozinski A. Primary empty sella (PES): a review of 175 cases. Pituitary 2013, 16:270-274.
  12. Himes BT, Bhargav AG, Brown DA, Kaufmann TJ, Bancos I, Van Gompel JJ. Does pituitary compression/empty sella syndrome contribute to MRI-negative Cushing's disease? A single-institution experience. Neurosurg Focus. 2020 Jun;48(6):E3. doi: 10.3171/2020.3.FOCUS2084. PMID: 32480375
  13. Urhan E, Hacioglu A, Okcesiz I, Karaca Z,  Kara CS, Unluhizarci The coexistence of newly diagnosed acromegaly with primary empty sella: More frequent than expected? Growth Horm IGF Res 2023 Feb:68:101521.doi: 10.1016/j.ghir.2022.101521. Epub 2022 Nov 11.
  14. Auer MK, MR Stieg, Crispin A, Sievers C, Stalla GK, Kopczak A. Primary empty sella syndrome and the prevalence of hormonal dysregulation. Deutsches Artzteblatt International 2018; 115: 99-105.
  15. Akkus G, Sözütok S , Odabaş F, Onan B, Evran M, Karagun B, Sert M, Tetiker T. Volume in Patients with Primary Empty Sella and Clinical Relevance to Pituitary Hormone Secretion Current Medical Imaging, 2021, 17, 1018-1024.
  16. Ekhzaimy AA, Mujammami M, Tharkar S, Alansary MA, Al Otaibi D. Clinical presentation, evaluation and case management of primary empty sella syndrome: a retrospective analysis of 10-year single-center patient data. BMC Endocr Disord. 2020 Sep 17;20(1):142. doi: 10.1186/s12902-020-00621-5. PMID: 32943019; PMCID: PMC7495892.
  17. Carosi G, Brunetti A, Mangone A, Baldelli R , Tresoldi A, Del Sindaco G, Lavezzi E , Sala E, Mungari R , Fatti LM, Galazzi E, Ferrante E, Indirli R, Biamonte E, Arosio M, Cozzi R, Lania A, Mazziotti G, Mantovani G. A Multicenter Cohort Study in Patients With Primary Empty Sella: Hormonal and Neuroradiological Features Over a Long Follow-Up. Frontiers in Endocrinology doi: 10.3389/fendo.2022.925378
  18. Lu M, Ye J, Gao F. Analysis of clinical features of primary empty sella. Ann Endocrinol (Paris) 2023 Apr;84(2):249-253.
  19. Yilmaz A, Gok M, Altas H, Yildirim T, Kaygisiz S, Isik HS. Retinal nerve fibre and ganglion cell inner plexiform layer analysis by optical coherence tomography in asymptomatic empty sella patients. Int J Neurosci. 2020 Jan;130(1):45-51. doi: 10.1080/00207454.2019.1660328. Epub 2019 Sep 13. PMID:31462116.
  20. Barzaghi LR, Donofrio CA, Panni P, Losa M, Mortini P. Treatment of empty sella associated with visual impairment: a systematic review of chiasmapexy techniques. Pituitary. 2018 Feb;21(1):98-106. doi: 10.1007/s11102-017-0842-6. Review
  21. Guinto G, Nettel B, Hernández E, Gallardo D, Aréchiga N, Mercado M. Osseous Remodeling Technique of the Sella Turcica: A New Surgical Option for Primary Empty Sella Syndrome. World Neurosurg. 2019 Jun;126:e953-e958. doi:10.1016/j.wneu.2019.02.195. Epub 2019 Mar 12. PMID: 30877013.
  22. Chen T, Li G, Wu D, Xie B , Feng Y, Xiao S, Li J, Liu Y, Yang J, Li X. Primary empty sella: The risk factors and associations with the cerebral small vessel diseases: An observational study. Clinical Neurology and Neurosurgery. Volume 203, April 2021, 106586
  23. Flesseriu M, Hashim IA, Karavitaki N, Melmed S, Murad MH, Salvatori R, Samuels MH: Hormonal replacement in hypoituitarism in adults: an endocrine society clinical practice guideline. Journal of Clinical Endocrinology and metabolism 2016; 101:3888-3921.

Adrenal Insufficiency In Children

ABSTRACT 

 

Adrenal insufficiency (AI) is an uncommon but potentially life-threatening condition related to impaired secretion of cortisol by the adrenal gland. In general, this condition can be divided into primary (adrenal failure) and central (hypothalamic/pituitary) causes. In this chapter, we categorize the causes of adrenal insufficiency and systematically review the etiologies and associations to guide the laboratory evaluation and treatment, specifically as it pertains to the pediatric patient. Early diagnosis and treatment can prevent the development of adrenal crisis – prompt recognition can be lifesaving. Understanding the manifestation of unique types of adrenal insufficiency can guide management with glucocorticoid +/- mineralocorticoids and guide further investigation for associated disorders. We discuss the treatment of adrenal insufficiency, reviewing both the acute (crisis) and chronic management.

 

INTRODUCTION

 

Adrenal insufficiency (AI) refers to impaired cortisol secretion by the adrenal gland. If untreated, AI can be life-threatening, especially when it is compounded by a physiological stress, such as an acute illness, severe trauma, or surgical procedure (1,2).

 

Cortisol secretion is regulated by the hypothalamic-pituitary-adrenal axis (HPA) (Figure 1) (1,2). In brief, Corticotropin-releasing hormone (CRH) secreted by the paraventricular nuclei of the hypothalamus stimulates adrenocorticotropic hormone (ACTH) production and release by the pituitary, leading to cortisol synthesis and secretion by the adrenal gland. Cortisol then negatively feeds back to inhibit both CRH and ACTH release. Clinical manifestations of this feedback mechanism occur frequently as illustrated in clinical practice after chronic treatment with exogenous glucocorticoids, which may suppress CRH and ACTH secretion and consequently result in adrenal atrophy and AI. In addition, a high day (light) and low night (dark) diurnal cycle of ACTH release is entrained, permitting optimal times for assessing the status of spontaneous cortisol secretion (early morning) or its optimal suppression (around midnight).

 

The fetal adrenal gland markedly differs in structure and function from that of the adult (3,4). During both early and late gestation, the adrenal cortex consists primarily of the fetal zone, which produces predominantly androgens that are critical for the sexual maturation of fetal external genitalia. Early cortisol production is indirect via placental conversion of progesterone synthesized in the fetal adrenal to cortisol, whereas, toward the end of the 2nd trimester, direct cortisol synthesis occurs in the definitive zone of the fetal adrenal (5). Aldosterone synthesis remains low until the end of gestation at which time CYP11B2 expression begins. After birth, the fetal zone undergoes atrophy via apoptosis, while the zona glomerulosa and zona fasciculata differentiate from the definitive zone to secrete aldosterone and cortisol respectively, whereas the zona reticularis starts its development postnatally and does not complete development until just before adrenarche (3,4).This process is fully described in the Endotext.org chapter, “Adrenal Cortex: Embryonic Development, Anatomy, Histology and Physiology” (6). The distinctive function of fetal zone and the transition from fetal to adult adrenal function have clinical applications in the very preterm infants who may have transient signs of relative AI after birth (7-9). Furthermore, babies who have a defect in steroidogenesis can present with both adrenal insufficiency and genital atypia (10). The most classic example in this category involves 46,XX babies with congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency who present with virilization and adrenal crisis (11). In this chapter, we discuss CAH in the context of AI. Additional information on CAH can be found in the Endotext.org chapter: Congenital Adrenal Hyperplasia (12) https://www.ncbi.nlm.nih.gov/books/NBK278953/. Mineralocorticoid deficiency will be reviewed in the context of primary adrenal insufficiency (PAI), but in-depth coverage of mineralocorticoid deficiency will be included elsewhere as a separate chapter.

 

PATHOPHYSIOLOGY & CLASSIFICATION

 

Adrenal insufficiency is classified into primary and central origin (Figure 1) (10,13,14).

 

Primary AI

 

Primary AI (PAI) refers to the destruction or dysfunction of the adrenal cortex, often resulting in combined cortisol and aldosterone deficiencies due to injury to both the zona fasciculata and zona glomerulosa (15,16). In cases of destruction, clinical presentation occurs when most of the cortex, >90%, is destroyed. Initial stages may be indolent and may often be followed by an acute stressor leading to clinical presentation and adrenal crisis. In contrast, in cases of dysfunction (i.e. steroidogenesis defects like 21-hydroxylase deficiency causing classical CAH), crisis can occur in the first few weeks of life if the steroidogenic defect is not detected promptly and in the absence of an acute stressor (17). In all cases, decreased glucocorticoid production and decreased negative feedback to the hypothalamus and pituitary result in increased production of ACTH and its pro-hormone pro-opiomelanocortin (POMC) (1). The increase in melanocyte-stimulating hormone (MSH) is responsible for the well-recognized hyperpigmentation present in PAI and often the clinical clue to evolving AI.

 

Central AI

 

Central AI involves disorders of the hypothalamic-pituitary region that impair CRH and/or ACTH secretion, and therefore, cortisol production. Aldosterone secretion in the zona glomerulosa of the adrenal cortex is primarily regulated by the renin-angiotensin system rather than ACTH and, thus, remains intact in central AI. Despite the intact mineralocorticoid secretion, hyponatremia may occur due to absence of the glucocorticoid regulated tonic suppression of ADH resulting in volume expansion, a clinical picture similar to the syndrome of inappropriate ADH secretion (SIADH) (18).  

 

SYMPTOMS

 

Acquired AI can have an insidious onset. Children may present with slowly progressive or nonspecific symptoms, such as anorexia, weight loss, morning nausea or vomiting, and fatigue, which may lead to diagnostic challenges and delayed diagnosis. One cross-sectional, retrospective adult study reported that only 15% of cases were correctly diagnosed at the initial presentation and nearly half had experienced symptoms for more than 1 year before establishing their diagnosis (19). Congenital AI usually presents early in life with signs of adrenal crisis or hypoglycemic seizures. Newborns with central AI may be completely asymptomatic until physical stress elicits an adrenal crisis.

 

In the pediatric population particularly, specific signs and symptoms should alert the clinician to the possibility of AI. In central AI, central nervous system (CNS) or midline defects may be present. Furthermore, in the neonate, cholestasis may occur due to immature bile acid synthesis and transport (20,21). Virilization and non-palpable gonads associated with the hyperandrogenemia in 46,XX individuals point to classical CAH due to 21-hydroxylase deficiency at birth (22). 46,XY babies with CAH, however, would not present with ambiguity and, thus, may escape medical attention to present with salt-wasting crises between the first and second weeks of life. The inclusion of 17-hydroxyprogesterone measurement in newborn screening programs has enabled detection of these babies prior to the onset of crisis. While the majority of individuals with classical CAH due to 21-hydroxylase deficiency are identified by newborn screening, false negative results and therefore missed diagnosis have been reported (23-26). Moreover, other certain rare enzyme deficiencies are not included in the newborn screen. Thus, a keen clinical index of suspicion remains crucial in cases of CAH who have escaped the newborn screening as experts continue to consider methods to enhance the testing accuracy (27,28). Other associations are discussed in detail under the heading of etiology and are reviewed in Table 1.

 

In acquired PAI, patients may experience salt craving (i.e., a sign of mineralocorticoid deficiency) and orthostasis and may be noted on examination to have bronze hyperpigmentation, especially in non-sun-exposed areas with prominence in the palmar creases, oral mucosa, skin folds, and areola.

 

In the case of adrenal crisis, glucocorticoid deficiency may progress to vomiting, muscle weakness, lethargy, hypoglycemia, and ultimately hemodynamic instability. Mineralocorticoid deficiency causes hyponatremia and hyperkalemia and, thus, symptoms may include headache, dizziness, abdominal pain, diarrhea, and ultimately severe dehydration and hypotension. Early detection is critical given the high morbidity and mortality associated with adrenal crisis (29).

 

ETIOLOGY

 

Both primary and central AI can arise neonatally due to congenital causes or later in childhood, adolescence and beyond due to acquired causes. In contrast to adults, genetic defects are more likely to be prevalent in infants and children. In recent years, knowledge and understanding of the genetic causes of AI have significantly increased and include every step of the hypothalamic-pituitary-adrenal axis responsible for cortisol and aldosterone synthesis and action (Figure 1 and Table 1).

 

Figure 1. Genetic defects in various steps along the hypothalamic-pituitary-adrenal (HPA) axis can result in either central or primary adrenal insufficiency depending on the location in the pathway. This figure displays the HPA axis and the location operative in the genetic defect, guiding the assessment of central or primary insufficiency. Genetic causes of central AI can include several genes involved in the development of the pituitary gland (e.g., PROP1, POU1F1 (formerly PIT1), GLI2, HESX1, LHX3, LHX4, SOX3, SOX2, OTX2), and result in multiple combined pituitary deficiencies. Central AI can result from an isolated ACTH deficiency caused by defects in the synthesis of pro-opiomelanocortin (POMC) or its cleavage. Defects of the melanocortin 2 receptor (MC2R) or its accessory protein MRAP result in ACTH-resistance (a.k.a. as Familial Glucocorticoid Deficiency). There are multiple genetic causes leading to primary AI (PAI). They range from genes that are involved in the development of the adrenal gland resulting in adrenal hypoplasia, defects in cortisol synthesis itself, or metabolic and autoimmune diseases (e.g. adrenoleukodystrophy, sphingolipidosis or autoimmune polyglandular syndrome), that result in destruction of the adrenal gland over time. Defects in steroidogenesis involve multiple genes along the pathway of cortisol synthesis and are the most frequent cause of PAI. Finally, PAI can be part of certain syndromes, such as Triple A Syndrome (AAAS) or disorders associated with oxidative stress.

 

Causes of Primary AI

 

CONGENITAL CAUSES

 

Disorders of Steroidogenesis

 

Disorders of steroidogenesis include 1) defects in the cholesterol biochemistry including Smith-Lemli-Opitz (DHCR7) (30), 2) early steroidogenesis defects such as congenital lipoid adrenal hyperplasia (STAR) and CYP11A1 mutations (31,32), and 3) defects within the adrenal gland causing CAH, the most common being CYP21A2 (21-hydroxylase deficiency) and rare forms including CYP11B1 (11β-hydroxylase deficiency), 3HSD2 (3β-hydroxylase deficiency), CYP17A1 (17α-hydroxylase deficiency), and POR (Cytochrome P450 oxidoreductase deficiency) (12) (See Fig 2 for depiction of adrenal steroidogenesis, Table 1 for further information on clinical presentations and associations and refer to the Endotext chapter on CAH for further details.)

 

Figure 2. The classical and backdoor pathways of adrenal steroidogenesis: The classical pathway is highlighted in blue, and the backdoor pathway is highlighted in orange. In the classical pathway, five enzymatic steps are necessary for cortisol production. In the first step of adrenal steroidogenesis, cholesterol enters mitochondria via a carrier protein called steroidogenic acute regulatory protein (StAR). ACTH stimulates cholesterol cleavage, the first and rate limiting step of adrenal steroidogenesis. The five enzymes required for cortisol production are cholesterol side chain cleavage enzyme (SCC), 17α-hydroxylase, 3β-hydroxysteroid dehydrogenase (3βHSD2), 21-hydroxylase, and 11β-hydroxylase. The backdoor pathway is an alternative pathway producing dihydrotestosterone. The enzymes include 5α-reductase 1, aldo keto reductases, retinol dehydrogenase RoDH, 17β-hydroxysteroid dehydrogenases, 17α-hydroxylase. (Figure from CAH Endotext Chapter).

Adrenal Hypoplasia

 

Underdevelopment of the adrenal glands may occur as an X-linked genetic disorder (DAX1/NROB1 – X-linked AHC) and results in both glucocorticoid and mineralocorticoid deficiency (33,34). X-linked AHC is associated with hypogonadism and in some cases, muscular dystrophy. Adrenal hypoplasia can be seen as part of syndromic undergrowth disorders including IMAGe and MIRAGE (35), or in association with gonadal dysgenesis and sex reversal such as NR5A1 (formerly SF1) (34) and SeRKAL syndrome (WNT4) (36) (see Table 1).

 

ACTH Resistance-Like Conditions

 

ACTH-resistance, known as Familial Glucocorticoid Deficiency (FGD), is another childhood cause of PAI. It is caused by defects of the melanocortin 2 receptor (MC2R) or its accessory protein at the adrenal gland rendering it unresponsive to ACTH action (Figure 1 and Table 1). Given the specific operative nature of ACTH resistance, concerns for mineralocorticoid insufficiency are rare (37). In FGD type 1, the MC2R receptor is dysfunctional. Patients may present with hyperpigmentation, jaundice, hypoglycemia, and early adrenal crisis or later in life with hyperpigmentation and fatigue (38). In FGD type 2, the MC2R receptor is absent. These cases are due to deficiency of the MRAP (melanocortin 2 receptor accessory protein) which assists in trafficking of the MC2R receptor to the cell surface.

 

Other disorders associated with ACTH resistance include those associated with oxidative stress (most commonly defects in nicotinamide nucleotide transhydrogenase [NNT] but also described in thioredoxin reductase 2 [TNXRD2]), Triple A Syndrome (Allgrove syndrome due to disruption of the Aladin protein [AAAS] ACTH resistance/Addison disease, alacrima (absence or deficiency of ocular tears), and achalasia of the esophagus)(37,38), and disruption of mini-chromosome maintenance 4 (MCM4)(37-39).

 

Metabolic Conditions

 

Metabolic conditions associated with PAI include mitochondrial diseases – large gene deletions such as Kearns-Sayre and Pearson as well as single gene disorders (MK-TK, MRPS7, QRSL1, NDUFAF5, GFER), lysosomal storage disorder (Wolman), sphingolipidosis (SGPL1 deficiency), and adrenoleukodystrophy(40-42). Descriptions and associations can be found in Table 1. In adrenoleukodystrophy, neurologic features can prompt identification. However, neurologic manifestations are absent in adrenal only forms of the disease. Adrenoleukodystrophy is included in the newborn screening programs in some states in the US. However, screening is not yet universal in the US and is rare in other countries. Consequently, a high index of suspicion should be maintained for all boys with unknown cause of PAI, and very long chain fatty acids (VLCFA) should be measured. Prompt diagnosis aids in therapeutic decision- making including hematopoietic stem cell transplant as well as more recent approval for gene therapy (43,44).

 

Zellweger Spectrum disorders consist of a group of peroxisomal related gene disorders that result in neurological dysfunction, hepatic dysfunction, renal cysts and PAI. Screening for PAI is recommended after the first year of life(45).  

 

AQUIRED CAUSES

Autoimmune Conditions

 

Addison’s disease (autoimmune adrenal insufficiency) is the most common cause of AI in adolescents and adults with a median age of presentation at 11 years (15). While less common in young children, it can be seen in association with other endocrine dysfunctions (poly-endocrinopathy). The best described monogenic cause is related to a defect in the AIRE gene and results in Autoimmune Polyglandular Syndrome type 1 (APS1). Presenting features often include mucocutaneous candidiasis and hypocalcemia related to hypoparathyroidism. In contrast, APS2 is typically of polygenic inheritance and is associated with autoimmune thyroiditis and diabetes mellitus with abnormalities in the DQ and DR genes. Adrenal insufficiency related to APS2 has a later age of onset of about 35 years of age, although pediatric cases do occur (15).

 

Other Causes of Primary AI

 

Other acquired causes include hemorrhagic, infectious, and infiltrative conditions. Adrenal hemorrhage should only result in adrenal insufficiency if both glands are affected extensively, as 10% of remaining function is sufficient to retain adrenal activity (46). Unilateral hemorrhage should not cause adrenal insufficiency as the single remaining gland is able to compensate. While adrenal hemorrhage occurs fairly often in traumatic birth delivery, few require treatment and of those that do, most resolve within 3-9 months (47). Waterhouse-Friderichsen syndrome is bilateral adrenal hemorrhage of infectious etiology and described with N. meningitides, streptococcus pneumoniae, and other bacterial infections (48). This adrenal insufficiency may also be reversible upon treatment of the primary infection supplemented with cortisol as needed. In contrast, tuberculous adrenal disease results in necrosis of the gland and is often irreversible unless discovered very early in the course of tuberculosis (49).

 

Several drugs increase cortisol clearance and may precipitate adrenal crisis or require a dose increase in patients with AI on daily glucocorticoid replacement. These include CYP3A4 inducers such as rifampin, mitotane, carbamazepine, and St. John’s wort. Initiation of growth hormone and T4 replacement may also increase cortisol metabolism (50). Medications used for treating excessive glucocorticoid secretion (Cushing’s disease) that directly block steroidogenesis (ketoconazole, etomidate, metyrapone, osilodrostat) and the glucocorticoid receptor (mifepristone) can induce adrenal insufficiency (51).

 

Central AI

 

CONGENITAL CAUSES

 

Combined Pituitary Hormone Deficiency

 

Many genetic causes of hypopituitarism (consisting of loss of ACTH with other anterior pituitary hormones such as GH, TSH, LH/FSH) have been elucidated including PROP1, POU1F1 (formerly PIT1), GH1, GLI2, HESX1, LHX3, LHX4, SOX3, SOX2, OTX2 and others (20,21,52). These can result in hypopituitarism on its own (non-syndromic) or in a constellation of other associated phenotypic features (syndromic), commonly involving midline defects. Despite a genetic etiology, some conditions including PROP1 and GH1 may manifest with ACTH insufficiency later in life. As such, close monitoring of the HPA axis in the setting of other pituitary deficiency is crucial (53).

 

Isolated ACTH Deficiency

 

Isolated ACTH deficiency can occur due to disruption of TBX-19 (formerly known as TPIT, required for pro-opiomelanocortin protein transcription), due to defects in pro-opiomelanocortin (POMC) or pro-hormone convertase-1 (PC-1/PCSK1) (20). The classic findings of red hair and pale skin and hyperphagic obesity in POMC mutations are related to the inability to produce pigmentation and regulate hunger in the absence of α melanocyte stimulating hormones. Another syndromic description involving ACTH deficiency includes DAVID (deficit in anterior pituitary function and variable immunodeficiency) syndrome which occurs in 2/3 of NFKB2 mutations.

 

AQUIRED CAUSES

 

Brain Lesion or Injury, Infiltrative Disease

 

Traumatic brain injury, brain tumor, infiltrative (histiocytosis, iron overload), and inflammatory (hypophysitis) disorders can result in pituitary dysfunction and impaired ACTH secretion; additional pituitary hormone deficiencies can also occur frequently with these conditions. Diagnosis of AI in these settings should therefore prompt a complete pituitary workup, including evaluation for central hypothyroidism, growth hormone deficiency, and hypogonadotropic hypogonadism (if in a pubertal child). Pituitary surgery may result in central AI post-operatively and during recovery. In contrast, in cases of brain radiation or developmental differences of the pituitary gland, AI may occur over time, and longitudinal assessment is suggested for affected individuals. Traumatic brain injury has been associated with central adrenal insufficiency in up to 14% of individuals with moderate to severe TBI and 6% of athletes with history of concussion with data coming from the adult population (54,55). Data in children have shown central AI in up to 47% of individuals at 3 months and recovery by 12 months after injury (56). 

 

Transient (Hypothalamic Suppression)

 

Glucocorticoid induced - Oral glucocorticoid administration above physiologic doses (i.e. Hydrocortisone 8-10mg/m2/day or equivalent) is associated with HPA axis suppression and subsequent adrenal atrophy after 2-4 weeks of daily use (57,58). Although there is significant interindividual variability, risk factors for AI include dose, frequency, and potency of the used glucocorticoid (59). Adrenal suppression can also be seen with inhaled or intranasal glucocorticoids, especially if their use is combined with intermittent use of oral formulations. Screening for AI has been suggested for children taking high doses of inhaled glucocorticoids (>500µg/day of Fluticasone or equivalent) for more than 6 months (60). Particular attention should be paid to the combination of glucocorticoid therapy and CYP3A4 inhibitors, such as grapefruit juice, as the latter can reduce glucocorticoid clearance, and therefore, augment their efficacy.

 

Adrenal function recovers once glucocorticoids are discontinued. Time to recovery is variable from 1 to many months, and can be affected by length of exposure, dose, frequency, and glucocorticoid potency (61-63). As such, recovery should be monitored, and stress dosing instructions provided until adrenal recovery.

 

Through a similar mechanism, successful treatment of Cushing’s syndrome and Cushing’s disease by resection of the pituitary or adrenal lesion results in a temporary adrenal insufficiency, necessitating replacement until recovery is demonstrated.

 

Other medications - Various medications have been shown to suppress the HPA axis. This is more frequently seen with long term opiate use (Opiate induced adrenal insufficiency - OIAI) due to tonic inhibition of the HPA axis – in which studies have shown 9 to 29% of individuals with adrenal suppression, potentially with high risk associated with younger age (64). There have also been cases of adrenal suppression with short term opiate use (65). Other medications responsible for adrenal suppression include somatostatin analogues, antipsychotics, and antidepressants.

 

Critical illness- such as sepsis or severe trauma, elicit a “fight-or-flight” or “stress” response that involves multiple physiological processes, including release of catecholamines and activation of the HPA axis. HPA axis activation results in a rapid rise in circulating ACTH and cortisol. The concept of “relative” AI during critical illness was introduced in the early 2000’s based on adult data of patients with septic shock who demonstrated an inadequate cortisol response to endocrine testing. The term “relative AI” was later replaced by “critical illness related corticosteroid insufficiency (CIRCI)” as these patients typically have “inappropriately” low ACTH and cortisol levels in response to stress.

 

Changes in adrenal function during critical illness are not fully understood (66). However, current research indicates that there is an initial brief HPA axis activation in response to a critical illness followed by a series of adaptive events that include a reduction in cortisol-binding globulin (CBG)/albumin leading to an increase in free cortisol, prolonged cortisol half-life due to suppressed metabolism in liver and kidneys, and tissue-specific changes in glucocorticoid receptor action (66). These peripheral adjustments increase systemic cortisol availability, and with prolonged illness, may result in central HPA suppression. From the clinical standpoint, endocrine testing (i.e. measurement of serum cortisol at baseline or after ACTH stimulation) can be challenging due to reductions in CBG, with one study noting this reduction lasting 7-8 days in adult patients (67). In terms of treatment, randomized controlled trials of stress dose hydrocortisone in critically ill adults showed inconsistent results on long term mortality (68). Hydrocortisone administration was found to have a positive effect on blood pressure, which can be related the pharmacologic effects of high dose glucocorticoids rather than treatment of CIRCI (66).

 

Pediatric data on CIRCI are limited. Current pediatric guidelines do not recommend hydrocortisone treatment for children with sepsis who are hemodynamically stable after fluid resuscitation but can be considered in those with fluid-refractory, inotrope-resistant shock (69).

 

Relative AI of the newborn - Very preterm infants may experience refractory hypotension that is unresponsive to fluid resuscitation and inotropic support but responds to treatment with glucocorticoids (7-9). No other apparent cause, like sepsis, is identified in these cases and electrolyte abnormalities indicative of a mineralocorticoid defect can be observed. It has been suggested that these infants have an attenuated cortical response to stress or relative AI. The term transient adrenocortical insufficiency of prematurity (TAP) has also been used as this phenomenon resolves within the first couple weeks of life (8). Immaturity of the HPA axis both at the level of hypothalamus, pituitary, and the adrenal gland itself have been implicated as the underlying pathophysiology. A late-onset GC -responsive circulatory collapse that occurs within the first 2 weeks of life and responds to therapy with glucocorticoids has also been described (70).

 

Formal diagnosis is challenging given studies failing to show association of low cortisol concentrations with adverse outcomes (71) while others identifying an association between high cortisol values with both morbidity (intracranial hemorrhage and cerebral palsy) and mortality (72,73).

 

Neither baseline nor stimulated values of cortisol have shown diagnostic benefit.

 

Cortisol secretion markedly increases during parturition to assist with lung maturation and transition to life after birth. There are data to suggest that very preterm infants who develop bronchopulmonary dysplasia (BPD) often have relative AI after birth. To address this concern, early low-dose hydrocortisone therapy as prophylaxis for AI was found to be beneficial for survival without BPD, although the treatment was associated with increased risk for spontaneous gastrointestinal perforation (74). 

 

TABLE 1. CAUSES OF ADRENAL INSUFFICIENCY IN CHILDREN

Congenital - Central

Combined pituitary deficiencies

Non syndromic (PROP1, POU1F1)

Variable presentations associated with single or multiple pituitary defects including ACTH deficiency. May include hypoglycemia and/or microphallus

Syndromic

·    Associated syndromes include optic cell hypoplasia / septo-optic dysplasia, microphthalmia (HESX1, SOX2, OTX2) and various CNS malformations (i.e., holoprosencephaly) with midline defects.

·    Syndromes with hypothalamic dysfunction (e.g., ROHHAD syndrome, Prader- Willi syndrome) can rarely be associated with ACTH deficiency.

Isolated ACTH deficiency

Defects in TBX19, POMC or PCSK1

·    Can present in the newborn with hypoglycemic seizures and jaundice.

·    Associated features (POMC): red hair, hyperphagia/obesity. MC4R agonists can be leveraged to treat obesity.

·    Associated features (PCSK1): malabsorptive diarrhea, obesity, and hypogonadism

Acquired - Central

Brain lesion or injury

Examples: Tumor, hemorrhage, irradiation

·    Usually associated with additional pituitary defects

·    Immune checkpoint inhibitors can be associated with hyper-autoimmunity and cause hypophysitis and hypopituitarism.

Infiltrative disease

Examples: iron overload (due to transfusions in thalassemias, hemochromatosis), sarcoidosis, Langerhans cell histiocytosis

·    Usually associated with additional pituitary defects.

·    Symptoms specific to the underlying causative disorder.

Transient (hypothalamic suppression)

Glucocorticoid therapy

·    The most frequent cause of adrenal insufficiency.

·    Adrenal function recovers with discontinuation of daily glucocorticoids.

Medications

·    Opiates, somatostatin analogues, antipsychotics and antidepressants

Treatment of Cushing

·    Permanent: hypophysectomy

·    Transient: post operative for unilateral adrenalectomy, or pituitary lesion

Critical illness -related corticosteroid insufficiency (CIRCI)

Characterized by a series of adaptions of the HPA axis in response to critical illness, which are still not clearly understood.  The beneficial effects of glucocorticoids in sepsis are likely related their anti-inflammatory and blood -pressure support effects rather than treatment of adrenal insufficiency.

Congenital - primary

ACTH resistance

Familial Glucocorticoid Deficiency

Type 1 (MC2R): Presents first weeks of life with severe hypoglycemia, prolonged jaundice

Type 2 (MRAP): Presents first few months of life with AI

Syndromic

Triple A (Allgrove) syndrome (AAAS): Associated with alacrimand achalasia of the esophagus presenting in childhood or 2nd decade of life

Disorders associated with oxidative stress (NNT, TNXRD2)

Congenital Adrenal Hypoplasia

X- linked Adrenal Hypoplasia (DAX-1/NR0B1)

·    Can present with salt losing early on or may present later in childhood

·    Additional features: hypogonadotropic hypogonadism.

·    Associated with a larger Xp21 contiguous gene deletion that can result in Duchene Muscular Dystrophy and Glycerol Kinase deficiency

·    Growth hormone insufficiency in a small subset

Syndromic

IMAGe syndrome (gain of function CDKN1C, POLE1): Additional features: IUGR, metaphyseal dysplasia, GU anomalies

MIRAGE syndrome (gain of function SAMD9): Additional features: Myelodysplasia, infections, restriction of growth, GU variations, enteropathy

NR5A1 mutation and SeRKAL syndrome (WNT4):  Associated with gonadal dysgenesis and sex reversal in 46XX individuals

Rare reports: Pena-Shokeir syndrome type 1 (DOK7, RAPSN), pseudotrisomy 13, Galloway-Mowat (WDR73), Pallister Hall (GLI3) and Meckel-Gruber(MKS1)

Disorders of steroidogenesis

Congenital Adrenal Hyperplasia (CAH)

Due to defects in 21-hydroxylase (CYP21A2)

·    The most common cause of PAI in neonates. 

·    Presents with salt wasting adrenal during 1st month of life crisis and genital virilization in 46XX individuals.

·    Biomarker: elevated 17-hydroxyprogesterone levels. Included in the state newborn screening in US

Due to defects in 11β-hydroxylase (CYP11B1), 3β-hydroxysteroid dehydrogenase type 2 (3HSD2), 17α-hydroxylase/17,20-lyase (CYP17A1), P450 oxidoreductase (POR):

·    Variable phenotype depending on the defect. Salt wasting and genital atypia can be present.

·    Biomarker: steroid precursor that accumulates above the specific enzymatic defect

Defects in cholesterol biochemistry

·    Smith-Lemli-Opitz (DHCR7). Additional features: Microcephaly, cleft palate, syndactyly, polydactyly, congenital heart, atypical genitalia with undescended testis.

·    Biomarker: elevated 7-dehydrocholesterol. AI is rare

·    Wolman disease (LIPA). Additional features: lysosomal storage disorder, hepatosplenomegaly, adrenal calcifications, failure to thrive

Early steroidogenic defect

Congenital lipoid adrenal hyperplasia (STAR), P450 Side Chain Cleavage (CYP11A1) mutations:

·    Associated with salt wasting and under-virilization of 46XY individuals.

·    Operative in adrenal and gonadal steroidogenesis

Salt wasting crisis in severe P450scc deficiency presents at 7-10 days while in STAR deficiency, the onset is more insidious, after 3-4 weeks of age

Partial enzyme defect may have less severe presentation

Metabolic disorders

Sphingosine-1-Phosphate Lyase (SGPL1) deficiency

·    Results from impaired breakdown of sphingosine 1-phosphate.

·    Additional features: adrenal calcifications, nephrotic syndrome. Ichthyosis, neurologic dysfunction.

Adrenoleukodystrophy

 

·    X- linked (ABCD1). Usually presents in childhood. Associated with progressive neurologic deterioration.

·    Zellweger Spectrum disorders (PEX). Associated with neonatal adrenoleukodystrophy, hepatomegaly, chondrodysplasia punctuate, hypotonia, seizures.

·    Biomarker: Elevated Very Long Chain Fatty Acids.

Wolman

·    LIPA mutation – lysosomal storage disorder with foamy lipid droplet accumulation, adrenal calcification and malabsorption

Mitochondrial Disorders

Variable Presenting features.

Acquired – Primary

Autoimmunity

Addison disease

·    The most common cause of acquired PAI. Presents in childhood.

·    Biomarker: 21-hydroxylase antibodies

·    Can be part of autoimmune polyglandular syndrome (APS) type I (AIRE) or type II (polygenic)

o   APS 1: other associations include: hypoparathyroidism, chronic mucocutaneous candidiasis, ectodermal dystrophy, autoimmune hepatitis, hypogonadism, pernicious anemia.

o   APS2: other associations include autoimmune thyroiditis, diabetes mellitus

 

Adrenal damage

Hemorrhage, Infectious, Infiltration (Tuberculosis, HIV, CMV)

Infectious: Tuberculosis, HIV, CMV

Infiltration: neuroblastoma

 

Transient

Relative adrenal insufficiency of the newborn

Described primarily in preterm sick infants within 2 weeks of life with signs of refractory hypotension in the absence of apparent cause and associated with electrolyte abnormalities (hyponatremia, hyperkalemia). May be related to immature adrenals in the very preterm infants.

 

 

Treatment of Cushing

·    Permanent: Bilateral adrenalectomy

·    Medication induced: ketoconazole, metyrapone, osilodrostat, etomidate, mifepristone

 

 

Medication induced

CYP3A4 inducers: rifampin, mitotane, carbamazepine and St. John’s wort

 

Mimickers of Adrenal Insufficiency

 

In the introduction to this chapter, we discussed the risk of delayed diagnosis of AI given the potential overlapping symptoms of glucocorticoid deficiency with other illnesses. In the same vein, other critical illnesses including sepsis and cardiovascular disease may be mistaken for adrenal crisis, especially in the setting of hydrocortisone responsiveness. In this case, hydrocortisone’s inotropic properties may lead to symptomatic improvement but may not be diagnostic of underlying AI.

 

Further, salt wasting adrenal crisis has findings overlapping with other salt-losing crises in infants including those due to kidney inflammation, infection, and obstruction – presenting with vomiting, hyponatremia, and hyperkalemia due to aldosterone resistance (a transient pseudo hypoaldosteronism) (75,76).

 

EPIDEMIOLOGY

 

Primary Adrenal Insufficiency (PAI)

 

PAI is a rare disease. Its incidence in children is not well established. An epidemiologic study from Finland observed a cumulative incidence of 10/100,000 at 15 years and 13/100,000 at 20 years (15). Studies from Europe that include both children and adults describe a similar prevalence (71).

 

In adults, the most common cause of PAI is autoimmune disease. In contrast, genetic defects are more prevalent in children. CAH due to 21-hydroxylase deficiency occurs in about 1/15,000 births and is the most frequent cause of PAI in children accounting for 50 to 86% of cases (15,77). As such, the age-related incidence of PAI in childhood is higher in the first year of life and decreases afterwards (15). Autoimmune disease is reported as the second most common cause, either as an isolated disease or as a manifestation of a poly endocrinopathy (i.e. APS 1 or 2) (77,78). Whereas most CAH cases are identified in the newborn period, autoimmune disease typically presents after the first couple years of life. X-linked adrenoleukodystrophy (XALD) is another cause of PAI during childhood with a prevalence in newborn males of about 1/20,000, approximately 80% of whom will develop PAI (79).

 

In recent years, the number of recognized genetic causes of PAI have significantly increased(Figure 1) (13). Genetic testing for PAI has a high diagnosis rate as illustrated by recent studies of children with PAI of unknown etiology, where genetic analysis established the diagnosis in most of the patients (80,81). The most frequent genes involved in these series were MC2R, NR0B/DAX and CYP11A1.

 

Central AI

 

Central AI is also quite rare with most epidemiologic data available in adult populations. The reported occurrence is about 14-28/100,000 individuals, with greater than 50% occurring because of a pituitary tumor (82-84). A Finnish epidemiologic study reporting single center data over a period of 30 years is one of the only studies of incidence in pediatrics. The study looked specifically at combined pituitary hormone deficiencies, without clearly distinguishing AI, and observed similar proportions in pediatric in comparison with previous adult studies, with 61% of hypopituitarism being acquired and the other 39% congenital (85). Craniopharyngiomas comprise the bulk of pediatric pituitary tumors, and similarly account for one quarter of acquired hypopituitarism, with gliomas following at just over 10% (85).  Up to 30% of patients with craniopharyngiomas will present with AI due to the lesion itself prior to surgical manipulation while 77-90% will have post-surgical AI (86,87).

 

Glucocorticoid induced AI is the most common cause of AI in both children and adults. Glucocorticoids are extensively used in clinical practice for treatment of various disorders, such as asthma, autoimmune and inflammatory diseases, and cancer (88). In US, approximately 1% of the adult population is on oral glucocorticoids and rates increase in the elderly population (89), while similar rates in children are not well established. In adults, AI has been observed around 48% with oral administration and 7.8% with inhalation (57). Rates of AI in children on inhaled glucocorticoids have been reported around 10%, but results may vary depending on the study (90,91). Adrenal crisis and related deaths have been described in children treated with inhaled glucocorticoids(92,93).

 

DIAGNOSTIC APPROACH TO A PATIENT WITH SUSPECTED AI

 

Our diagnostic and differential approach to a child with suspected AI is summarized in Figure 2. Clinical signs and symptoms of AI are non-specific and require a high index of suspicion; when present, the suspected diagnosis needs to be confirmed with appropriate laboratory evaluation. Evaluation typically starts with measurement of serum ACTH, cortisol, and electrolytes. In the case of an emergency, such as suspected adrenal crisis, laboratory evaluation can only be interpreted if obtained prior to administration of glucocorticoids as therapy can influence test results.

 

The goals of the initial laboratory assessment are to confirm the diagnosis of AI, and then, understand if AI is primary or central. Most cases of PAI, but not all, involve combined glucocorticoid and mineralocorticoid deficiencies. In the case of PAI, therefore, mineralocorticoid function must be assessed with measurements of serum electrolytes, aldosterone, and renin (Figure 2). Details on laboratory assessment and additional dynamic testing are included in the LABORATORY EVALUATION section of this chapter.

 

The next step is to pinpoint the specific etiology so that management can be individualized. Clinical presentation and symptoms can suggest an underlying etiology and guide additional laboratory assessment. For example, salt wasting adrenal crisis in a newborn should prompt measurement of serum 17-hydroxyprogesterone concentrations to rule out CAH. Vitiligo in a child with PAI suggests an autoimmune process, and the underlying etiology can be confirmed with measurement of 21-hydroxylase antibody titers. AI in a boy with neurological manifestations points to adrenoleukodystrophy and calls for measurements of VLCFA. In many instances, however, the specific etiology cannot be identified. In such cases, genetic testing may be very helpful.

Figure 3. A proposed approach in the diagnosis and differential of AI in children
GC: glucocorticoid. MR: mineralocorticoid. Na: sodium. K: potassium.

LABORATORY EVALUATION

 

Assessment of Glucocorticoid Function

 

BASELINE ASSESSMENT OF CORTISOL AND ACTH CONCENTRATIONS

 

The diagnosis of PAI can be made by measurement of morning ACTH and cortisol concentrations and by demonstrating an inappropriate high ACTH concentration for the cortisol value (Figure 2). Typically, a cortisol value of <5mcg/dL (140nmol/L) associated with an ACTH level > x2 above upper normal range indicates PAI (16). A low morning cortisol value associated with an inappropriately normal ACTH concentration may indicate central AI (94). It is suggested that an 8am cortisol values of <3 mcg/dL (82.8 nmol/ L) in the absence of an elevated ACTH concentration is indicative of central AI (94). Establishing the diagnosis of central AI can be challenging because ACTH concentrations are frequently in the low normal/normal range. The presence of additional pituitary defects increases the risk for AI and needs to be considered when evaluating a child with possible central AI.

 

Assessment of adrenal function, such as in individuals at risk for glucocorticoid-induced AI, can also start with a measurement of morning cortisol (58,95). Adult studies suggest that an 8am cortisol value of >12mcg/dL can predict normal adrenal function (96,97). It is recognized, however, that various guideline and practice recommendations point to slightly different cut-offs (58,94). Further, the suggested cut-offs need to be interpreted by taking into account the cortisol assay that is used. Of note, the listed cut-offs also do not apply for patients on oral contraceptives and/or estrogen replacement as these medications  increase CBG concentrations, and therefore, cortisol values (98).

 

Morning cortisol values may be unreliable in the newborn as the diurnal pattern of cortisol secretion may take few months to become established (99). Baseline cortisol values have been extensively studied in the sick preterm infant and were not found to be predictive of AI or adverse outcomes (71-73). Furthermore, high cortisol values during first week of life were associated with intracranial hemorrhage and cerebral palsy later in life (73).

 

DYNAMIC OR STIMULATION TESTS

 

Stimulations tests are typically performed when the diagnosis of AI needs confirmation. The ACTH (a.k.a. corticotropin or Cosyntropin or Synacthen) stimulation test is the primary test performed in children. Historically, the insulin tolerance test (ITT) has been considered the gold standard test for the diagnosis of AI, but its use has been abandoned as it is associated with severe hypoglycemia (1,16,95). The overnight metyrapone test, an alternative to ITT, is also not routinely performed in children because of potential significant adverse effects (16,95). The CRH (corticotropin releasing hormone) stimulation test has been used in the diagnosis of central AI but there is no evidence that it has greater diagnostic accuracy than ACTH testing in pediatrics and CRH is no longer commercially available (100).

 

The ACTH test involves the intramuscular or intravenous administration of cosyntropin, a synthetic fragment of ACTH with full biologic activity, along with cortisol measurements at baseline and after cosyntropin administration. Although differences in practice protocols have been described, a common protocol entails cosyntropin doses of 15mcg/kg in infants, 125mcg in children <2years and 250mcg in children older than 2 years (16). Cortisol concentrations are usually measured at 30- and 60-minutes post stimulation, although again, practice variations have been described with the use of the “short” ACTH test that includes cortisol measurement at baseline and only at 30 min after stimulation(101).

 

The standard dose (250mcg) ACTH test has been validated against the ITT; it has good diagnostic accuracy for PAI but only moderate for central AI (102,103). The low dose ACTH test that involves administration of 1mcg of cosyntropin was developed as an alternative that may offer greater sensitivity in the diagnosis of central AI. Like adult data, however, meta-analysis of pediatric studies indicates that both standard- and low- dose ACTH tests have similar diagnostic accuracy with high specificity but only moderate sensitivity for the diagnosis of central AI (102). These findings suggest that the clinical picture needs to be considered when interpreting ACTH test results. Furthermore, the low dose ACTH test bears technical challenges related to potentially erroneously low administered dose because it requires dilution and cosyntropin is known to adhere to plastic tubing used in intravenous tests (103).

 

Regardless of the standard- or low- dose ACTH test used, the diagnosis of AI is based on peak cortisol concentrations of 18mcg/dL at either 30 or 60 minutes after cosyntropin (16). The cortisol cut-off of 18mcg/dL was established using cortisol assays of previous methodology that are no longer in use, and therefore, is not applicable in today’s clinical practice. Cortisol measured by liquid chromatography with tandem mass spectrometry (LC-MS/MS) is currently considered the gold standard, yet cortisol immunoassays remain widely used in most facilities. Although there no official guidelines or consensus statements at the moment, adult data support revised cortisol cut-offs at 30 min post cosyntropin of around 15mcg/dL using LC-MS/MS and second-generation immunoassays such as Elescys II (Roche Diagnostics) and Access (Beckman Coulter) (101,104,105) . Collectively, the adult data call for revised cortisol cut-offs specific to the assay that is used to avoid overdiagnosis of AI. Limited pediatric data show similar findings as in adults (106).

 

Literature deriving primarily from critical care medicine uses a cortisol rise of >9mcg/dL during ACTH stimulation to define AI. This criterion is greatly influenced by the baseline cortisol value and has been abandoned (66,95). The ACTH test may miss the diagnosis of AI in newborns with a central defect in ACTH secretion (e.g.,  septo-optic dysplasia or congenital panhypopituitarism) if it is performed within 1-2 weeks after birth. These babies have normal fetal adrenal function driven by placental CRH stimulation, and therefore, normal response to ACTH stimulation shortly after birth(107). However, they develop adrenal atrophy and insufficiency within the first 2 months of life. Similarly, the ACTH test can be falsely negative if the testing is done during the early stages of AI due to exogenous glucocorticoids or a pituitary/CNS insult and before the atrophy of the adrenals has occurred (95).  A high index of suspicion and repeat testing may be needed in such cases. The ACTH test results can be significantly influenced by CBG concentrations. Estrogens are well known to increase CBG concentrations, and estrogen containing oral contraceptives can increase the cortisol cut-off after ACTH stimulation by approximately 10mcg/dL (98,108). Conversely, disease states such as acute illness, cirrhosis, and nephrotic syndrome lead to decreased CBG concentrations and potential overdiagnosis of AI (98). Rare genetic syndromes, such as congenital CBG deficiency or familial glucocorticoid resistance, can also affect baseline as well as stimulated cortisol measurements.

 

The standard-dose ACTH test is also performed when a disorder of steroid biosynthesis, such as CAH, is suspected. In addition to cortisol, adrenal steroids are measured at baseline and 60-minute post stimulation. Specific nomograms based on 17-hydroxyprogesterone values have been developed for the diagnosis of CAH due to 21-hydroxylase deficiency (12).

 

Assessment of Mineralocorticoid Function

 

Electrolytes abnormalities that include hyponatremia in combination with hyperkalemia and acidosis should raise suspicion for a mineralocorticoid defect. The simultaneous measurement of plasma aldosterone and renin is important for the diagnosis of a mineralocorticoid defect. An inappropriate low aldosterone concentration in the presence elevated renin levels indicates mineralocorticoid deficiency. In such instances, the treating physician needs to carefully assess the glucocorticoid function of the child to determine whether the child has combined deficiencies, such as seen in CAH, or an isolated mineralocorticoid defect, such as seen with aldosterone synthase deficiency. Conversely, markedly elevated aldosterone and renin levels in a child with hyponatremia and hyperkalemia suggest mineralocorticoid resistance, such as pseudo-hypoaldosteronism(109).

 

Aldosterone and renin concentrations are higher in the newborn and can be affected by prematurity (110-112). Newborns at term have a state of partial aldosterone resistance and hence concurrent high aldosterone values in their blood, that resolve in the first few months of life, whereas babies with severe prematurity have an initial defect in aldosterone secretion followed later by the appearance of physiologic aldosterone resistance (110-112). These physiologic changes need to be considered when assessing mineralocorticoid function in the first few months of life. Lastly, some children with PAI, like those with adrenoleukodystrophy, develop glucocorticoid deficiency initially, while they retain mineralocorticoid activity (79); ongoing monitoring with measurements of serum electrolytes, plasma aldosterone and renin concentrations are required to determine the need for starting mineralocorticoid replacement.

 

Supporting/Miscellaneous Testing

 

Once AI is documented, additional testing based on patients’ history and symptomatology can pinpoint the specific cause. For example, presence of genital atypia suggests a disorder of steroidogenesis, and elevated concentrations of specific steroid precursors can determine the enzymatic defect (e.g. 17-hydroxyprogesterone in 21-hydroxylase deficiency or 11-deoxycorticosterone in 11β-hydroxylase deficiency) (12). Elevated VLCFA can establish the diagnosis of adrenal leukodystrophy in a boy with AI and neurological manifestations. Positive antibody titers for 21-hydroxylase indicates autoimmune Addison disease. Nonetheless, the underlying cause can be frequently tentative. To this end, genetic testing has become increasingly important. Testing can be tailored according to clinical suspicion and varies from single candidate gene analysis to targeted “panels” or whole exome sequencing (WES). In general, genetic testing for AI has a high diagnostic success rate and can be very helpful in complex cases where diagnosis remains otherwise uncertain (80).

 

TREATMENT

 

Hormone Replacement

 

The goals of daily replacement therapy are to avoid symptoms of adrenal insufficiency while securing optimal growth and weight gain, and appropriate puberal progression.

 

GLUCOCORTICOID REPLACEMENT

 

The glucocorticoid of choice for children is hydrocortisone, typically given three time daily (16). The physiologic daily cortisol secretion is approximately 5-8mg/m2/day (113). In practice, typical replacement doses are 8-10mg/m2/day, except in CAH which frequently requires supraphysiologic doses to suppress adrenal androgen secretion. The dose distribution during the day usually mimics the diurnal pattern of cortisol secretion with the largest dose given upon awakening, the second dose around noon and the last dose at early evening to avoid overnight hypercortisolemia, which may lead to sleep disturbances, stunted growth, or insulin resistance. A reverse circadian hydrocortisone administration has been used in CAH to suppress the overnight rise in ACTH. This regimen has been criticized as non-physiologic. Individuals with central AI can also be treated with a twice a day regimen (94), with the first larger dose given upon awakening and the second smaller dose in late afternoon. It is our practice to place children with central AI on a twice a day glucocorticoid replacement regimen, acknowledging that this procedure is based on adult recommendations, while pertinent pediatric data or guidelines are lacking. Specific to children with glucocorticoid-induced AI, hydrocortisone replacement twice a day with the second dose administered in late afternoon can be beneficial and facilitate recovery of the adrenal axis as it avoids nighttime hypercortisolemia and suppression of ACTH secretion.

 

Challenges with daily hydrocortisone replacement include its short half-life of 60-90min and significant variability in clearance among patients, which result in alternating periods of hypo- and hyper-cortisolemia while on treatment. A modified release hydrocortisone (Plenadren by Takeda Pharmaceuticals International AG Ireland) was designed as a more physiologic and convenient alternative (114). Plenadren provides hydrocortisone released in two phases (i.e., an initial phase of immediate release followed by a phase of extended release) and is given as a once-a day dose upon awakening. It is available in Europe but not in the USA. An additional concern with hydrocortisone administration in young children involves difficulties in dose titration as hydrocortisone is not available in tablets less than 5 mg. To overcome this barrier, hydrocortisone microgranules that provide doses as low as 0.5 mg have been introduced in the market (115). Hydrocortisone microgranules have 1:1 bio equivalency with the standard immediate release hydrocortisone. Finally, it should be  kept in mind that certain drugs, like carbamazepine or phenytoin, can induce CYP3A4 in the liver and increase hepatic hydrocortisone clearance requiring higher hydrocortisone replacement doses (11).

Prednisone administered orally once to twice daily can be used as an alternative to hydrocortisone when growth is complete. Use of dexamethasone is not recommended because of difficulties with dose titration and high risk for cushingoid side effects (94).

 

MINERALOCORTICOID REPLACEMENT   

 

Mineralocorticoid replacement with fludrocortisone at doses 0.05-0.2 mg daily is recommended in patients with PAI and confirmed aldosterone deficiency (16). Higher doses (i.e., 0.1-0.2mg daily) are typically required during the first year of life as newborns have lower mineralocorticoid sensitivity. In addition, infants typically require salt supplementation with sodium chloride, at doses of 1-2 grams daily given in 3-4 divided doses. Treatment with sodium chloride is gradually reduced during the first couple of years of life and replaced with unrestricted salt supplementation with food as the child transitions to regular diet. Extra fluids with electrolytes are recommended when excessive sweating is anticipated, such as during vigorous exercise or hot weather.

 

MONITORING DURING THERAPY

 

Monitoring of Glucocorticoid Replacement

 

There is no good biomarker to assess glucocorticoid replacement. Morning ACTH concentrations are frequently elevated and attempts to normalize ACTH levels may result in overtreatment (16). Measurements of serum cortisol concentrations after hydrocortisone administration is not routinely done. Monitoring of therapy, therefore, relies primarily on clinical assessment. Morning nausea with poor appetite and weight loss can all be signs of inadequate treatment. Children should be questioned about their daily activities and energy during the day (i.e., need for napping after school), and the information should be considered when titrating medication doses. Growth velocity and weight gain are also sensitive clinical indicators. Specifically, overtreatment with glucocorticoids is associated with poor linear growth and an increase in BMI. Appropriate progression through puberty is reassuring.

 

Monitoring of Mineralocorticoid Replacement

 

As with glucocorticoids, mineralocorticoid replacement is also assessed clinically starting with questions related to salt craving and measurements of standing blood pressure and heart rate (16). Fatigue and poor growth can also be signs of inadequate mineralocorticoid replacement (95).

 

Measurements of serum potassium and plasma renin (level or activity) can be used for titration of fludrocortisone dose and sodium chloride supplementation (1,2,16,94). Goals are to maintain serum potassium concentrations in the normal range and plasma renin in the upper range of normal. Orthostatic hypotension, high renin and hyperkalemia indicate undertreatment. On the contrary, low or suppressed plasma renin along with a low serum potassium concentration are signs of mineralocorticoid overtreatment. In such cases, blood pressure needs to be assessed to ensure that the child does not experience hypertension, and fludrocortisone and sodium chloride doses adjusted. In the case of hypertension, one must also remember that hydrocortisone has mineralocorticoid activity and high replacement doses can contribute to blood pressure elevations. Thus, hydrocortisone doses need to be assessed and titrated as needed.

 

Sick Day Management and Stress Dosing   

 

Endogenous cortisol secretion increases during acute illness, anesthesia, surgery, or trauma. Hence, individuals with AI require an increase in their glucocorticoid doses during times of physiological stress to avoid adrenal crisis.

 

The stress dose regimens for children are largely empirical or consensus driven and may vary across practices. With lack of good quality evidence, many pediatric recommendations are adopted from adult literature (116). As an overarching principle, stress dose management errs on the side of overtreatment to avoid a potentially life-threatening adrenal crisis.

 

For management purposes, the stressor is frequently referred to as “moderate” or “severe”. Severe stress refers to major illness (e.g., sepsis), that requires hospitalization, major surgical procedures that require general anesthesia (e.g., abdominal surgery), or severe trauma. Moderate stress refers to an acute illness that can be managed at home or with minor surgery (e.g., a dental procedure) (table 2).

 

Table 2.  Summary of Stress Dose Management of a Child with Adrenal Insufficiency: Indications and Doses

 

Indications

Hydrocortisone dose

Comments

Adrenal Crisis

Vomiting, lethargy, hemodynamic instability

Age 0-24 months

25mg

♦      IVF resuscitation with 0.9% sodium chloride/5% glucose

♦      Continue with severe stress dose coverage.

Age 2yrs - 10 yrs

50mg

Age >10 years

100mg

IM/SQ

Severe stress

Surgery, sepsis, major trauma, lethargy, repeated vomiting

100mg/m2/day divided 6 hours IV/IM

Max 50mg every 6 hours

 

♦      Wean according to clinical status/improvement.

♦      Switch to moderate stress dosing usually when able to tolerate po.

Moderate stress

Fever >38.3oC (101oF)

Significant trauma (i.e., broken bone), Minor surgery requiring anesthesia (e.g., dental procedures), seizures, intense exercise (i.e., marathon)

30-50 mg/m2/day divided 8 hours orally

♦      Continue stress dose coverage for up to 24 hours after stress resolves.

Prednisone can be used as an alternative if hydrocortisone is not available. Dexamethasone is the least preferable option.

Additional indications: Minor cold, school exams = no need for stress dosing. Medical procedure with local anesthesia = moderate stress dose orally once prior to procedure. 

 

MANAGEMENT OF ADRENAL CRISIS

 

Adrenal crisis is characterized by hypotension and volume depletion. Hypoglycemia is frequent in children. In cases of combined glucocorticoid and mineralocorticoid defects, there is additional urinary sodium loss resulting in electrolyte abnormalities (i.e., hyponatremia, hyperkalemia, acidosis, and elevated serum urea). The cornerstones of therapy are glucocorticoid replacement and fluid resuscitation.

 

Children with suspected adrenal crisis should immediately receive a bolus dose of hydrocortisone IM or IV (Table 2). For emergencies, a dose based on age can be used, such as 25mg, 50mg and 100mg for children <2 years, 1-10 years, and older than 10 years, respectively. The child should then continue receiving stress dose coverage with parenteral hydrocortisone at doses of 100mg/m2/day given every 6 hours (maximum dose 50mg every 6 hours) until their condition improves. If hydrocortisone is not available, prednisone (20mg/m2/day) can be used as an alternative, while dexamethasone (4mg/m2/day) is the least preferable glucocorticoid option, related to its metabolic consequences as well as lack of mineralocorticoid properties. 

 

Fluid resuscitation can start with a bolus of 0.9% sodium chloride at 10 mL/kg. In the case of hypoglycemia, normal saline with 5% glucose can be used. Fluids should continue based on patient’s needs. Hyperkalemia can be severe at the onset of adrenal crisis but improves rapidly with parenteral glucocorticoid and fluid management. Additional therapies to lower serum potassium concentrations can be considered (i.e., IV insulin and glucose, IV calcium gluconate or cation exchange resins) if severe hyperkalemia persists.

 

MANAGEMENT OF SEVERE STRESS

 

It entails parenteral administration of hydrocortisone at 100mg/m2/day given every 6 hours (maximum dose 50mg every 6 hours) (116) (Table 2) Doses can be tapered rapidly and based on clinical improvement to the established daily glucocorticoid regimen. For children recovering from surgery, hydrocortisone can usually be changed to oral sick-day doses once they are stable and can tolerate oral fluids and diet. Children with PAI do not require fludrocortisone during severe stress coverage, as hydrocortisone also has mineralocorticoid activity. Fludrocortisone must be added back to their treatment plan as hydrocortisone is weaned down to approximately 50-60mg/m2/day.

 

MANAGEMENT OF MODERATE STRESS OR HOME SICK DAY RULES 

 

Families are advised to increase their child’s daily glucocorticoid dose if they experience an intercurrent illness, such as fever or diarrhea (Table 2). Hydrocortisone doses that provide stress dose coverage for such situations (i.e. moderate stress) are in the range of 30-50mg/m2/day given every 6 or 8 hours (116). A frequent instruction to patients and families is to double or triple the daily hydrocortisone dose. This approach may lead to undertreatment if the daily dose is small. Calculation of a specific moderate stress dose based on the child’s body surface area (BSA) is preferable as it provides a more precise dosing.

 

Stress dose coverage is not recommended for minor upper respiratory viral infections, increased schoolwork, emotional stress, or intense exercise of brief duration. Although there is no specific guideline, some medical providers recommend using stress doses during prolonged intense training, such as a marathon.

 

Patients and families should undergo in-depth training around stress dose management. Instructions should include indications for moderate stress glucocorticoid administration and appropriate doses and training on using hydrocortisone as an emergency injection IM in case of vomiting, severe trauma, or impending adrenal crisis. They should be provided with a medical letter or card that documents stress dose instructions and contact information of medical providers and caregivers. Children should wear a medical alert bracelet and carry emergency supplies of oral glucocorticoids and emergency injectable hydrocortisone.

 

Glucocorticoid-Induced Adrenal Insufficiency: Wean and Recovery of Adrenal Function

 

GLUCOCORTICOID TAPERING

 

 There is no evidence to support a specific approach to GC taper. For individuals on chronic glucocorticoid treatment (i.e., >3-4 weeks) at supraphysiologic doses for an underlying disease (e.g. inflammatory or immune disorder), glucocorticoid taper should be done at a rate dictated by the underlying condition in order to maintain disease remission (58).  

 

Glucocorticoid withdrawal syndrome has been described primarily in adults during tapering of supraphysiologic doses or after successful treatment of Cushing (117). The syndrome can mimic signs of adrenal insufficiency (i.e.,  fatigue, muscle aches, nausea, abdominal discomfort, weight loss, mood swings, irritability) but is not related to untreated adrenal insufficiency, as the glucocorticoid daily dose is still supraphysiologic (117).  The underlying mechanism is incompletely understood and likely involves cytokine and prostaglandin upregulation as cortisol concentrations decline. Should glucocorticoid withdrawal syndrome be suspected tapering down to physiologic doses can be done at a slower pace (58).

 

Once physiologic glucocorticoid doses have been achieved, a slower taper below physiologic doses have been suggested (58). The tapering aims to alleviate symptoms of adrenal insufficiency while allowing for HPA axis recovery. For those on long-acting glucocorticoids (e.g., dexamethasone), it is suggested to switch to hydrocortisone to ease with titration at small doses and faster recovery of the adrenal axis. Weaning below physiologic doses is done mostly empirically since there is no evidence about the best practice protocol. It is our practice to wean from physiologic replacement to off glucocorticoids in four-to- five steps by reducing the dose by 20-25% (Table 3). We follow a twice daily hydrocortisone/ prednisone regimen and cut down initially the evening dose to allow for faster HPA axis recovery. Children should remain on a stress dose plan until there is evidence of recovery of adrenal function.

 

Table 3. Glucocorticoid Induced Adrenal Insufficiency: Proposed Wean From Physiologic Replacement Doses to Off Glucocorticoids

Length of GC exposure

<4 weeks

4-12 weeks

>12 weeks

Hydrocortisone

(given twice daily)

No wean

10mg/m2/day x 4 days

10mg/m2/day x 7 days

8mg/m2/day x 4 days

8mg/m2/day x 7 days

6mg/m2/day x 4 days

6mg/m2/day x 7 days

4mg/m2/day x 4 days

4mg/m2/day x 7 days

stop

stop

Prednisone

(Given twice daily)

No wean

4mg/m2/day x 4 days

4mg/m2/day x 7 days

3mg/m2/day x 4 days

3mg/m2/day x 7 days

2mg/m2/day x 4 days

2mg/m2/day x 7 days

1mg/m2/day x 4 days

1mg/m2/day x 7 days

Stop

Stop

 

ASSESSMENT AND RECOVERY OF ADRENAL FUNCTION

 

Adrenal function recovers once supraphysiologic doses of glucocorticoids are discontinued. The time to recovery, however, is variable and dependent on length and potency of the glucocorticoid that was used (58).  Assessment of adrenal function can be done with the measurement of morning cortisol concentrations. Because hydrocortisone interferes in cortisol measurements, testing should be at least 18–24 hours after the last dose. Adult data support a morning cortisol value above 12 mcg/dL as indicative of normal adrenal function, while values less than 5 mcg/dL suggest suppression of the HPA axis. These cortisol cut-offs are dependent on the cortisol assay that is used. An ACTH stimulation test can be considered for intermediate cortisol values (i.e. 5-12mcg/dL). An alternative approach can be to continue stress dose steroid coverage and repeat a measurement of morning cortisol after few weeks and until recovery of the axis is documented.

 

USEFUL LINKS/GUIDELINES

 

  • Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016 https://pubmed.ncbi.nlm.nih.gov/26760044/
  • European Society of Endocrinology and Endocrine Society Joint Clinical Guideline: Diagnosis and Therapy of Glucocorticoid-induced Adrenal Insufficiency. J Clin Endocrinol Metab. 2024 PMC11180513.https://pubmed.ncbi.nlm.nih.gov/38724043/
  • Hormonal Replacement in Hypopituitarism in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016 https://pubmed.ncbi.nlm.nih.gov/27736313/
  • Emergency and perioperative management of adrenal insufficiency in children and young people: British Society for Paediatric Endocrinology and Diabetes consensus guidance. Arch Dis Child. 2023 https://pubmed.ncbi.nlm.nih.gov/37045585/

 

REFERENCES

 

  1. Hahner S, Ross RJ, Arlt W, et al. Adrenal insufficiency. Nat Rev Dis Primers. Mar 11 2021;7(1):19. doi:10.1038/s41572-021-00252-7
  2. Husebye ES, Pearce SH, Krone NP, Kämpe O. Adrenal insufficiency. Lancet. Feb 13 2021;397(10274):613-629. doi:10.1016/S0140-6736(21)00136-7
  3. Pignatti E, du Toit T, Flück CE. Development and function of the fetal adrenal. Rev Endocr Metab Disord. Feb 2023;24(1):5-21. doi:10.1007/s11154-022-09756-3
  4. Kim JH, Choi MH. Embryonic Development and Adult Regeneration of the Adrenal Gland. Endocrinol Metab (Seoul). Dec 2020;35(4):765-773. doi:10.3803/EnM.2020.403
  5. Johnston ZC, Bellingham M, Filis P, et al. The human fetal adrenal produces cortisol but no detectable aldosterone throughout the second trimester. BMC Med. Feb 12 2018;16(1):23. doi:10.1186/s12916-018-1009-7
  6. Nicolaides NC, Willenberg HS, Bornstein SR, Chrousos GP. Adrenal Cortex: Embryonic Development, Anatomy, Histology and Physiology. In: Feingold KR, Anawalt B, Blackman MR, et al, eds. Endotext. 2000.
  7. Holmstrom LE, Jnah AJ. Relative Adrenal Insufficiency: Crisis Averted? Neonatal Netw. Nov 01 2021;40(6):369-376. doi:10.1891/11-T-703
  8. Ng PC. Adrenocortical insufficiency and refractory hypotension in preterm infants. Arch Dis Child Fetal Neonatal Ed. Nov 2016;101(6):F571-F576. doi:10.1136/archdischild-2016-311289
  9. Gutierrez CE, De Beritto T. Relative Adrenal Insufficiency in the Preterm Infant. Neoreviews. May 01 2022;23(5):e328-e334. doi:10.1542/neo.23-5-e328
  10. Buonocore F, McGlacken-Byrne SM, Del Valle I, Achermann JC. Current Insights Into Adrenal Insufficiency in the Newborn and Young Infant. Front Pediatr. 2020;8:619041. doi:10.3389/fped.2020.619041
  11. Yau M, Rao N, Nimkarn S, Vogiatzi M. Effect of ethosuximide on cortisol metabolism in the treatment of congenital adrenal hyperplasia. J Pediatr Endocrinol Metab. May 2014;27(5-6):549-54. doi:10.1515/jpem-2013-0147
  12. Yau M, Khattab A, Yuen T, New M. Congenital Adrenal Hyperplasia. In: Feingold KR, Anawalt B, Blackman MR, et al, eds. Endotext. 2000.
  13. Buonocore F, Achermann JC. Primary adrenal insufficiency: New genetic causes and their long-term consequences. Clin Endocrinol (Oxf). Jan 2020;92(1):11-20. doi:10.1111/cen.14109
  14. Hasenmajer V, Ferrigno R, Minnetti M, et al. Rare forms of genetic paediatric adrenal insufficiency: Excluding congenital adrenal hyperplasia. Rev Endocr Metab Disord. Apr 2023;24(2):345-363. doi:10.1007/s11154-023-09784-7
  15. Borchers J, Pukkala E, Mäkitie O, Laakso S. Epidemiology and Causes of Primary Adrenal Insufficiency in Children: A Population-Based Study. J Clin Endocrinol Metab. Oct 18 2023;108(11):2879-2885. doi:10.1210/clinem/dgad283
  16. Bornstein SR, Allolio B, Arlt W, et al. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. Feb 2016;101(2):364-89. J Clin Endocrinol Metab. doi:10.1210/jc.2015-1710
  17. Sharma R, Seth A. Congenital adrenal hyperplasia: issues in diagnosis and treatment in children. Indian J Pediatr. Feb 2014;81(2):178-85. doi:10.1007/s12098-013-1280-8
  18. Martel-Duguech L, Poirier J, Bourdeau I, Lacroix A. Diagnosis and management of secondary adrenal crisis. Rev Endocr Metab Disord. Jun 2024;25(3):619-637. doi:10.1007/s11154-024-09877-x
  19. Bleicken B, Hahner S, Ventz M, Quinkler M. Delayed diagnosis of adrenal insufficiency is common: a cross-sectional study in 216 patients. Am J Med Sci. Jun 2010;339(6):525-31. doi:10.1097/MAJ.0b013e3181db6b7a
  20. Patti G, Guzzeti C, Di Iorgi N, et al. Central adrenal insufficiency in children and adolescents. Best Pract Res Clin Endocrinol Metab. Aug 2018;32(4):425-444. doi:10.1016/j.beem.2018.03.012
  21. Alatzoglou KS, Dattani MT. Genetic forms of hypopituitarism and their manifestation in the neonatal period. Early Hum Dev. Nov 2009;85(11):705-12. doi:10.1016/j.earlhumdev.2009.08.057
  22. Yau M, Gujral J, New MI. Congenital Adrenal Hyperplasia: Diagnosis and Emergency Treatment. In: Feingold KR, Anawalt B, Blackman MR, et al, eds. Endotext. 2000.
  23. Sarafoglou K, Banks K, Gaviglio A, Hietala A, McCann M, Thomas W. Comparison of one-tier and two-tier newborn screening metrics for congenital adrenal hyperplasia. Pediatrics. Nov 2012;130(5):e1261-8. doi:10.1542/peds.2012-1219
  24. Saroufim R, Nebesio TD, Eugster EA. Characteristics of patients with classic congenital adrenal hyperplasia missed on the newborn screen. Horm Res Paediatr. Nov 22 2023;doi:10.1159/000535405
  25. Chan CL, McFann K, Taylor L, Wright D, Zeitler PS, Barker JM. Congenital adrenal hyperplasia and the second newborn screen. J Pediatr. Jul 2013;163(1):109-13 e1. doi:10.1016/j.jpeds.2013.01.002
  26. Varness TS, Allen DB, Hoffman GL. Newborn screening for congenital adrenal hyperplasia has reduced sensitivity in girls. J Pediatr. Oct 2005;147(4):493-8. doi:10.1016/j.jpeds.2005.04.035
  27. Sarafoglou K, Gaviglio A, Wolf C, et al. Can incorporating molecular testing improve the accuracy of newborn screening for congenital adrenal hyperplasia? J Clin Endocrinol Metab. May 3 2024;doi:10.1210/clinem/dgae297
  28. Lasarev MR, Bialk ER, Allen DB, Held PK. Application of Principal Component Analysis to Newborn Screening for Congenital Adrenal Hyperplasia. J Clin Endocrinol Metab. Aug 1 2020;105(8)doi:10.1210/clinem/dgaa371
  29. Alexandraki KI, Grossman A. Diagnosis and Management of Adrenal Insufficiency. In: Feingold KR, Anawalt B, Blackman MR, et al, eds. Endotext. 2000.
  30. Bianconi SE, Conley SK, Keil MF, et al. Adrenal function in Smith-Lemli-Opitz syndrome. Am J Med Genet A. Nov 2011;155A(11):2732-8. doi:10.1002/ajmg.a.34271
  31. Bose HS, Sugawara T, Strauss JF, 3rd, Miller WL, International Congenital Lipoid Adrenal Hyperplasia C. The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. N Engl J Med. Dec 19 1996;335(25):1870-8. doi:10.1056/NEJM199612193352503
  32. Miller WL. Disorders in the initial steps of steroid hormone synthesis. J Steroid Biochem Mol Biol. Jan 2017;165(Pt A):18-37. doi:10.1016/j.jsbmb.2016.03.009
  33. Muscatelli F, Strom TM, Walker AP, et al. Mutations in the DAX-1 gene give rise to both X-linked adrenal hypoplasia congenita and hypogonadotropic hypogonadism. Nature. Dec 15 1994;372(6507):672-6. doi:10.1038/372672a0
  34. Suntharalingham JP, Buonocore F, Duncan AJ, Achermann JC. DAX-1 (NR0B1) and steroidogenic factor-1 (SF-1, NR5A1) in human disease. Best Pract Res Clin Endocrinol Metab. Aug 2015;29(4):607-19. doi:10.1016/j.beem.2015.07.004
  35. Cabrera-Salcedo C, Kumar P, Hwa V, Dauber A. IMAGe and Related Undergrowth Syndromes: The Complex Spectrum of Gain-of-Function CDKN1C Mutations. Pediatr Endocrinol Rev. Mar 2017;14(3):289-297. doi:10.17458/per.vol14.2017.SKHD.imageandrelatedundergrowth
  36. Mandel H, Shemer R, Borochowitz ZU, et al. SERKAL syndrome: an autosomal-recessive disorder caused by a loss-of-function mutation in WNT4. Am J Hum Genet. Jan 2008;82(1):39-47. doi:10.1016/j.ajhg.2007.08.005
  37. Clark AJ, Weber A. Adrenocorticotropin insensitivity syndromes. Endocr Rev. Dec 1998;19(6):828-43. doi:10.1210/edrv.19.6.0351
  38. Elias LL, Clark AL. The molecular basis of adrenocorticotrophin resistance syndrome. Prog Mol Biol Transl Sci. 2009;88:155-71. doi:10.1016/S1877-1173(09)88005-8
  39. Meimaridou E, Hughes CR, Kowalczyk J, Chan LF, Clark AJ, Metherell LA. ACTH resistance: genes and mechanisms. Endocr Dev. 2013;24:57-66. doi:10.1159/000342504
  40. Fitoussi G, Negre-Salvayre A, Pieraggi MT, Salvayre R. New pathogenetic hypothesis for Wolman disease: possible role of oxidized low-density lipoproteins in adrenal necrosis and calcification. Biochem J. Jul 1 1994;301 ( Pt 1)(Pt 1):267-73. doi:10.1042/bj3010267
  41. Turk BR, Theda C, Fatemi A, Moser AB. X-linked adrenoleukodystrophy: Pathology, pathophysiology, diagnostic testing, newborn screening and therapies. Int J Dev Neurosci. Feb 2020;80(1):52-72. doi:10.1002/jdn.10003
  42. Hannah-Shmouni F, Stratakis CA. An overview of inborn errors of metabolism manifesting with primary adrenal insufficiency. Rev Endocr Metab Disord. Mar 2018;19(1):53-67. doi:10.1007/s11154-018-9447-2
  43. Prinzi J, Pasquali M, Hobert JA, et al. Diagnosing X-Linked Adrenoleukodystrophy after Implementation of Newborn Screening: A Reference Laboratory Perspective. Int J Neonatal Screen. Nov 2 2023;9(4)doi:10.3390/ijns9040064
  44. Singh S, Ojodu J, Kemper AR, Lam WKK, Grosse SD. Implementation of Newborn Screening for Conditions in the United States First Recommended during 2010-2018. Int J Neonatal Screen. Apr 6 2023;9(2)doi:10.3390/ijns9020020
  45. Braverman NE, Raymond GV, Rizzo WB, et al. Peroxisome biogenesis disorders in the Zellweger spectrum: An overview of current diagnosis, clinical manifestations, and treatment guidelines. Mol Genet Metab. Mar 2016;117(3):313-21. doi:10.1016/j.ymgme.2015.12.009
  46. Ruffini E, De Petris L, Zorzi G, Paoletti P, Mambelli G, Carlucci A. Two cases of neonatal adrenal hemorrhage presenting with persistent jaundice. Pediatr Med Chir. Nov-Dec 2013;35(6):285-7. doi:10.4081/pmc.2013.26
  47. Demirel N, Bas AY, Zenciroglu A, Tasci-Yildiz Y. Adrenal bleeding in neonates: report of 37 cases. Turk J Pediatr. Jan-Feb 2011;53(1):43-7.
  48. Mizuno S, Yokoyama K, Nukada T, Hara S. Waterhouse-Friderichsen Syndrome and Central Diabetes Insipidus Due to Escherichia coli Meningitis. JCEM Case Rep. Jan 2023;1(1):luac031. doi:10.1210/jcemcr/luac031
  49. Sharma S, Joshi R, Kalelkar R, Agrawal P. Tuberculous Adrenal Abscess Presenting as Adrenal Insufficiency in a 4-Year-Old Boy. J Trop Pediatr. Jun 1 2019;65(3):301-304. doi:10.1093/tropej/fmy046
  50. Fonseca V, Brown R, Hochhauser D, Ginsburg J, Havard CW. Acute adrenal crisis precipitated by thyroxine. Br Med J (Clin Res Ed). May 3 1986;292(6529):1185-6. doi:10.1136/bmj.292.6529.1185
  51. Fleseriu M, Petersenn S. Medical therapy for Cushing's disease: adrenal steroidogenesis inhibitors and glucocorticoid receptor blockers. Pituitary. Apr 2015;18(2):245-52. doi:10.1007/s11102-014-0627-0
  52. Kelberman D, Rizzoti K, Lovell-Badge R, Robinson IC, Dattani MT. Genetic regulation of pituitary gland development in human and mouse. Endocr Rev. Dec 2009;30(7):790-829. doi:10.1210/er.2009-0008
  53. Cerbone M, Dattani MT. Progression from isolated growth hormone deficiency to combined pituitary hormone deficiency. Growth Horm IGF Res. Dec 2017;37:19-25. doi:10.1016/j.ghir.2017.10.005
  54. Rai A, Rajasekar G, Sahoo J, Kumari MM, Anbazhagan S. Anterior Pituitary Dysfunction After Traumatic Brain Injury: A Prospective Study. World Neurosurg. Jul 31 2024;doi:10.1016/j.wneu.2024.07.186
  55. Claessen LOE, Kristjansdottir H, Jonsdottir MK, Lund SH, Kristensen ISU, Sigurjonsdottir HA. Screening for possible hypopituitarism following mild traumatic brain injury: The first all-female study. Who do we need to evaluate further? NeuroRehabilitation. 2023;52(2):259-271. doi:10.3233/NRE-220194
  56. Casano-Sancho P. Pituitary dysfunction after traumatic brain injury: are there definitive data in children? Arch Dis Child. Jun 2017;102(6):572-577. doi:10.1136/archdischild-2016-311609
  57. Broersen LH, Pereira AM, Jørgensen JO, Dekkers OM. Adrenal Insufficiency in Corticosteroids Use: Systematic Review and Meta-Analysis. J Clin Endocrinol Metab. Jun 2015;100(6):2171-80. doi:10.1210/jc.2015-1218
  58. Beuschlein F, Else T, Bancos I, et al. European Society of Endocrinology and Endocrine Society Joint Clinical Guideline: Diagnosis and Therapy of Glucocorticoid-induced Adrenal Insufficiency. J Clin Endocrinol Metab. Jun 17 2024;109(7):1657-1683. doi:10.1210/clinem/dgae250
  59. Borresen SW, Klose M, Glintborg D, Watt T, Andersen MS, Feldt-Rasmussen U. Approach to the Patient With Glucocorticoid-induced Adrenal Insufficiency. J Clin Endocrinol Metab. Jun 16 2022;107(7):2065-2076. doi:10.1210/clinem/dgac151
  60. Issa-El-Khoury K, Kim H, Chan ES, Vander Leek T, Noya F. CSACI position statement: systemic effect of inhaled corticosteroids on adrenal suppression in the management of pediatric asthma. Allergy Asthma Clin Immunol. 2015;11(1):9. doi:10.1186/s13223-015-0075-z
  61. Einaudi S, Bertorello N, Masera N, et al. Adrenal axis function after high-dose steroid therapy for childhood acute lymphoblastic leukemia. Pediatr Blood Cancer. Mar 2008;50(3):537-41. doi:10.1002/pbc.21339
  62. Rix M, Birkebaek NH, Rosthoj S, Clausen N. Clinical impact of corticosteroid-induced adrenal suppression during treatment for acute lymphoblastic leukemia in children: a prospective observational study using the low-dose adrenocorticotropin test. J Pediatr. Nov 2005;147(5):645-50. doi:10.1016/j.jpeds.2005.06.006
  63. Ahmet A, Rowan-Legg A, Pancer L. Adrenal suppression from exogenous glucocorticoids: Recognizing risk factors and preventing morbidity. Paediatr Child Health. Jul 2021;26(4):242-254. doi:10.1093/pch/pxab015
  64. Donegan D, Bancos I. Opioid-Induced Adrenal Insufficiency. Mayo Clin Proc. Jul 2018;93(7):937-944. doi:10.1016/j.mayocp.2018.04.010
  65. Policola C, Stokes V, Karavitaki N, Grossman A. Adrenal insufficiency in acute oral opiate therapy. Endocrinol Diabetes Metab Case Rep. 2014;2014:130071. doi:10.1530/EDM-13-0071
  66. Langouche L, Téblick A, Gunst J, Van den Berghe G. The Hypothalamus-pituitary-adrenocortical Response to Critical Illness: A Concept in Need of Revision. Endocr Rev. Nov 09 2023;44(6):1096-1106. doi:10.1210/endrev/bnad021
  67. Beishuizen A, Thijs LG, Vermes I. Patterns of corticosteroid-binding globulin and the free cortisol index during septic shock and multitrauma. Intensive Care Med. Oct 2001;27(10):1584-91. doi:10.1007/s001340101073
  68. Annane D, Bellissant E, Bollaert PE, et al. Corticosteroids for treating sepsis in children and adults. Cochrane Database Syst Rev. Dec 06 2019;12(12):CD002243. doi:10.1002/14651858.CD002243.pub4
  69. Weiss SL, Peters MJ, Alhazzani W, et al. Surviving Sepsis Campaign International Guidelines for the Management of Septic Shock and Sepsis-Associated Organ Dysfunction in Children. Pediatr Crit Care Med. Feb 2020;21(2):e52-e106. doi:10.1097/PCC.0000000000002198
  70. Iijima S. Late-onset glucocorticoid-responsive circulatory collapse in premature infants. Pediatr Neonatol. Dec 2019;60(6):603-610. doi:10.1016/j.pedneo.2019.09.005
  71. Sari FN, Dizdar EA, Oguz SS, et al. Baseline and stimulated cortisol levels in preterm infants: is there any clinical relevance? Horm Res Paediatr. 2012;77(1):12-8. doi:10.1159/000332157
  72. Aucott SW, Watterberg KL, Shaffer ML, Donohue PK, group Ps. Early cortisol values and long-term outcomes in extremely low birth weight infants. J Perinatol. Jul 2010;30(7):484-8. doi:10.1038/jp.2009.191
  73. Aucott SW, Watterberg KL, Shaffer ML, Donohue PK, Group PS. Do cortisol concentrations predict short-term outcomes in extremely low birth weight infants? Pediatrics. Oct 2008;122(4):775-81. doi:10.1542/peds.2007-2252
  74. Shaffer ML, Baud O, Lacaze-Masmonteil T, Peltoniemi OM, Bonsante F, Watterberg KL. Effect of Prophylaxis for Early Adrenal Insufficiency Using Low-Dose Hydrocortisone in Very Preterm Infants: An Individual Patient Data Meta-Analysis. J Pediatr. Apr 2019;207:136-142.e5. doi:10.1016/j.jpeds.2018.10.004
  75. Sousa AG, Cabral JV, El-Feghaly WB, de Sousa LS, Nunes AB. Hyporeninemic hypoaldosteronism and diabetes mellitus: Pathophysiology assumptions, clinical aspects and implications for management. World J Diabetes. Mar 10 2016;7(5):101-11. doi:10.4239/wjd.v7.i5.101
  76. Pai B, Shaw N, Hogler W. Salt-losing crisis in infants-not always of adrenal origin. Eur J Pediatr. Feb 2012;171(2):317-21. doi:10.1007/s00431-011-1541-3
  77. Capalbo D, Moracas C, Cappa M, et al. Primary Adrenal Insufficiency in Childhood: Data From a Large Nationwide Cohort. J Clin Endocrinol Metab. Mar 08 2021;106(3):762-773. doi:10.1210/clinem/dgaa881
  78. Perry R, Kecha O, Paquette J, Huot C, Van Vliet G, Deal C. Primary adrenal insufficiency in children: twenty years experience at the Sainte-Justine Hospital, Montreal. J Clin Endocrinol Metab. Jun 2005;90(6):3243-50. doi:10.1210/jc.2004-0016
  79. Huffnagel IC, Laheji FK, Aziz-Bose R, et al. The Natural History of Adrenal Insufficiency in X-Linked Adrenoleukodystrophy: An International Collaboration. J Clin Endocrinol Metab. Jan 01 2019;104(1):118-126. doi:10.1210/jc.2018-01307
  80. Buonocore F, Maharaj A, Qamar Y, et al. Genetic Analysis of Pediatric Primary Adrenal Insufficiency of Unknown Etiology: 25 Years' Experience in the UK. J Endocr Soc. Aug 01 2021;5(8):bvab086. doi:10.1210/jendso/bvab086
  81. Guran T, Buonocore F, Saka N, et al. Rare Causes of Primary Adrenal Insufficiency: Genetic and Clinical Characterization of a Large Nationwide Cohort. J Clin Endocrinol Metab. Jan 2016;101(1):284-92. doi:10.1210/jc.2015-3250
  82. Arlt W, Allolio B. Adrenal insufficiency. Lancet. May 31 2003;361(9372):1881-93. doi:10.1016/S0140-6736(03)13492-7
  83. Regal M, Paramo C, Sierra SM, Garcia-Mayor RV. Prevalence and incidence of hypopituitarism in an adult Caucasian population in northwestern Spain. Clin Endocrinol (Oxf). Dec 2001;55(6):735-40. doi:10.1046/j.1365-2265.2001.01406.x
  84. Tomlinson JW, Holden N, Hills RK, et al. Association between premature mortality and hypopituitarism. West Midlands Prospective Hypopituitary Study Group. Lancet. Feb 10 2001;357(9254):425-31. doi:10.1016/s0140-6736(00)04006-x
  85. Hietamaki J, Karkinen J, Iivonen AP, et al. Presentation and diagnosis of childhood-onset combined pituitary hormone deficiency: A single center experience from over 30 years. EClinicalMedicine. Sep 2022;51:101556. doi:10.1016/j.eclinm.2022.101556
  86. Miao Y, Fan K, Peng X, et al. Postoperative hypothalamic-pituitary dysfunction and long-term hormone replacement in patients with childhood-onset craniopharyngioma. Front Endocrinol (Lausanne). 2023;14:1241145. doi:10.3389/fendo.2023.1241145
  87. Harsha GS, Dabadghao P, Sudhanshu S. Long-Term Outcomes of Paediatric-Onset Craniopharyngioma: A Retrospective Analysis from a Tertiary Care Centre in North India. Neurol India. Mar-Apr 2022;70(2):600-605. doi:10.4103/0028-3886.344661
  88. Monge Chacón AG, Wang C, Waqar D, Syeda SA, Kumar R, Meghana DR. Long-Term Usage of Oral Glucocorticoids Leading to Adrenal Insufficiency: A Comprehensive Review of the Literature. Cureus. May 2023;15(5):e38948. doi:10.7759/cureus.38948
  89. Overman RA, Yeh JY, Deal CL. Prevalence of oral glucocorticoid usage in the United States: a general population perspective. Arthritis Care Res (Hoboken). Feb 2013;65(2):294-8. doi:10.1002/acr.21796
  90. Zöllner EW, Lombard CJ, Galal U, Hough FS, Irusen EM, Weinberg E. Hypothalamic-pituitary-adrenal axis suppression in asthmatic school children. Pediatrics. Dec 2012;130(6):e1512-9. doi:10.1542/peds.2012-1147
  91. Smith RW, Downey K, Gordon M, et al. Prevalence of hypothalamic-pituitary-adrenal axis suppression in children treated for asthma with inhaled corticosteroid. Paediatr Child Health. May 2012;17(5):e34-9. doi:10.1093/pch/17.5.e34
  92. Todd GR, Acerini CL, Ross-Russell R, Zahra S, Warner JT, McCance D. Survey of adrenal crisis associated with inhaled corticosteroids in the United Kingdom. Arch Dis Child. Dec 2002;87(6):457-61. doi:10.1136/adc.87.6.457
  93. Goldbloom EB, Mokashi A, Cummings EA, et al. Symptomatic adrenal suppression among children in Canada. Arch Dis Child. Apr 2017;102(4):338-339. doi:10.1136/archdischild-2016-311223
  94. Fleseriu M, Hashim IA, Karavitaki N, et al. Hormonal Replacement in Hypopituitarism in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. Nov 2016;101(11):3888-3921. doi:10.1210/jc.2016-2118
  95. Charoensri S, Auchus RJ. A Contemporary Approach to the Diagnosis and Management of Adrenal Insufficiency. Endocrinol Metab (Seoul). Feb 2024;39(1):73-82. doi:10.3803/EnM.2024.1894
  96. Sbardella E, Isidori AM, Woods CP, et al. Baseline morning cortisol level as a predictor of pituitary-adrenal reserve: a comparison across three assays. Clin Endocrinol (Oxf). Feb 2017;86(2):177-184. doi:10.1111/cen.13232
  97. Debono M, Elder CJ, Lewis J, et al. Home Waking Salivary Cortisone to Screen for Adrenal Insufficiency. NEJM Evid. Feb 2023;2(2):EVIDoa2200182. doi:10.1056/EVIDoa2200182
  98. Choi MH. Clinical and Technical Aspects in Free Cortisol Measurement. Endocrinol Metab (Seoul). Aug 2022;37(4):599-607. doi:10.3803/EnM.2022.1549
  99. Stoye DQ, Boardman JP, Osmond C, et al. Preterm birth and infant diurnal cortisol regulation. Arch Dis Child Fetal Neonatal Ed. Sep 2022;107(5):565-567. doi:10.1136/archdischild-2021-323296
  100. Brar PC. Clinical utility of stimulation tests in infants with suspected adrenal insufficiency (AI). J Pediatr Endocrinol Metab. May 27 2019;32(5):529-531. doi:10.1515/jpem-2019-0025
  101. Javorsky BR, Raff H, Carroll TB, et al. New Cutoffs for the Biochemical Diagnosis of Adrenal Insufficiency after ACTH Stimulation using Specific Cortisol Assays. J Endocr Soc. Apr 01 2021;5(4):bvab022. doi:10.1210/jendso/bvab022
  102. Ospina NS, Al Nofal A, Bancos I, et al. ACTH Stimulation Tests for the Diagnosis of Adrenal Insufficiency: Systematic Review and Meta-Analysis. J Clin Endocrinol Metab. Feb 2016;101(2):427-34. doi:10.1210/jc.2015-1700
  103. Burgos N, Ghayee HK, Singh-Ospina N. Pitfalls in the interpretation of the cosyntropin stimulation test for the diagnosis of adrenal insufficiency. Curr Opin Endocrinol Diabetes Obes. Jun 2019;26(3):139-145. doi:10.1097/MED.0000000000000473
  104. Ueland G, Methlie P, Øksnes M, et al. The Short Cosyntropin Test Revisited: New Normal Reference Range Using LC-MS/MS. J Clin Endocrinol Metab. Apr 01 2018;103(4):1696-1703. doi:10.1210/jc.2017-02602
  105. Birtolo MF, Antonini S, Saladino A, et al. ACTH Stimulation Test for the Diagnosis of Secondary Adrenal Insufficiency: Light and Shadow. Biomedicines. Mar 15 2023;11(3)doi:10.3390/biomedicines11030904
  106. Cortez S, Arbeláez AM, Wallendorf M, McNerney K. Peak Serum Cortisol Cutoffs to Diagnose Adrenal Insufficiency Across Different Cortisol Assays in Children. J Clin Res Pediatr Endocrinol. Nov 22 2023;15(4):375-379. doi:10.4274/jcrpe.galenos.2023.2023-2-3
  107. Coshway LK, Indyk JA, Bowden SA. Repeating ACTH Stimulation Test Is Necessary to Diagnose ACTH Deficiency in Neonatal Hypopituitarism With Initial False Negative Result. Glob Pediatr Health. 2014;1:2333794X14563385. doi:10.1177/2333794X14563385
  108. El-Farhan N, Tennant S, Rees SE, Evans C, Rees DA. Salivary Cortisol Response to ACTH Stimulation Is a Reliable Alternative to Serum Cortisol in Evaluating Hypoadrenalism. J Clin Endocrinol Metab. Jan 18 2024;109(2):e579-e588. doi:10.1210/clinem/dgad576
  109. Arai K, Papadopoulou-Marketou N, Chrousos GP. Aldosterone Deficiency and Resistance. In: Feingold KR, Anawalt B, Blackman MR, et al, eds. Endotext. 2000.
  110. Travers S, Martinerie L, Boileau P, Lombès M, Pussard E. Alterations of adrenal steroidomic profiles in preterm infants at birth. Arch Dis Child Fetal Neonatal Ed. Mar 2018;103(2):F143-F151. doi:10.1136/archdischild-2016-312457
  111. Martinerie L, Pussard E, Foix-L'Hélias L, et al. Physiological partial aldosterone resistance in human newborns. Pediatr Res. Sep 2009;66(3):323-8. doi:10.1203/PDR.0b013e3181b1bbec
  112. Viengchareun S, Pussard E, Castanet M, et al. The invention of aldosterone, how the past resurfaces in pediatric endocrinology. Mol Cell Endocrinol. Sep 15 2021;535:111375. doi:10.1016/j.mce.2021.111375
  113. Linder BL, Esteban NV, Yergey AL, Winterer JC, Loriaux DL, Cassorla F. Cortisol production rate in childhood and adolescence. J Pediatr. Dec 1990;117(6):892-6. doi:10.1016/s0022-3476(05)80128-3
  114. Johannsson G, Nilsson AG, Bergthorsdottir R, et al. Improved cortisol exposure-time profile and outcome in patients with adrenal insufficiency: a prospective randomized trial of a novel hydrocortisone dual-release formulation. J Clin Endocrinol Metab. Feb 2012;97(2):473-81. doi:10.1210/jc.2011-1926
  115. Neumann U, Braune K, Whitaker MJ, et al. A Prospective Study of Children Aged 0-8 Years with CAH and Adrenal Insufficiency Treated with Hydrocortisone Granules. J Clin Endocrinol Metab. Mar 08 2021;106(3):e1433-e1440. doi:10.1210/clinem/dgaa626
  116. Mushtaq T, Ali SR, Boulos N, et al. Emergency and perioperative management of adrenal insufficiency in children and young people: British Society for Paediatric Endocrinology and Diabetes consensus guidance. Arch Dis Child. Nov 2023;108(11):871-878. doi:10.1136/archdischild-2022-325156
  117. Zhang CD, Ioachimescu AG. Challenges in the postsurgical recovery of cushing syndrome: glucocorticoid withdrawal syndrome. Front Endocrinol (Lausanne). 2024;15:1353543. doi:10.3389/fendo.2024.1353543

 

Infections Of The Hypothalamic-Pituitary Region

ABSTRACT

 

Infections of the hypothalamic-pituitary region are rare lesions, accounting for less than 1% of all pituitary lesions. The clinical diagnosis of these infections can be difficult due to the nonspecific nature of the disease (in many patients without symptoms of infection) and may be misdiagnosed as other pituitary lesions. The risk factors for infections of the hypothalamic-pituitary region are meningitis, paranasal sinusitis, head surgery, and immunocompromised host (diabetes mellitus, Cushing’s syndrome, HIV infections, solid organ transplantation, malignancy). Infections can develop in a normal pituitary gland or in pre-existing pituitary lesions (adenoma, Rathke´s cleft cyst, craniopharyngioma). There are several modes of dissemination of the infection to the hypothalamic-pituitary region: hematogenous, iatrogenic (after neurosurgical procedures), and spread from paranasal or nasal cavity (through venous channels of the sphenoid bone). Hypothalamic-pituitary infections most commonly present with visual disturbances and headache, in some cases with fever and leukocytosis. A significant proportion of patients develop hypothalamic-pituitary dysfunction during the acute phase of the disease or months and years after successful antimicrobial therapy. Diagnosis can be challenging and the hypothalamic-pituitary infection with formation of abscess or granuloma may be misdiagnosed as a pituitary tumor. Transsphenoidal drainage followed by antibiotics, antimycotics, or anti-tuberculous drugs are usually efficient in successful treatment of these patients.

 

INTRODUCTION

 

Infections of the hypothalamic-pituitary region are rare and commonly described in case reports or small case series. These infections include bacterial infections (pituitary abscess), tuberculosis, fungal, viral, and parasitic infections (Table 1). An infection in the hypothalamic-pituitary region may present as a sella/suprasellar mass and may be misinterpreted as a pituitary tumor. Also, these infections may cause hypopituitarism and be misdiagnosed as post-encephalitic syndrome (1-4).

 

Table 1. Infectious Agents which Cause Hypothalamic-Pituitary infections

BACTERIA

·       Gram-positive cocci (Staphylococcus, Streptococcus)

·       Gram-negative cocci (Neisseria, Esherichia coli , Pseudomonas, Brucella)

·       Spirochete (Treponema pallidum, Leptospira Interrogans)

·       Mycobacteria (M. Tuberculosis)

VIRUS

·       Herpes simplex virus

·       Varicella zoster virus

·       Cytomegalovirus

·       Tick-borne

·       Hantaan (Hantan) virus

·       Enterovirus

·       Neuroborreliosis

·       SARS-CoV-2 virus

FUNGI

·       Candida

·       Aspergillus

PARASITES

·       Toxoplasma gondii, Echinococcus, Taenia solium

 

Infections of the hypothalamic-pituitary region may be primary (without an identifiable source) or secondary in origin (3, 5). The more common is a primary pituitary infection, which occurs in previously healthy normal pituitary glands. Secondary pituitary infections occur in patients with a pre-existing lesion in the pituitary region (pituitary adenoma, Rathke´s cleft cyst, craniopharyngioma, or prior pituitary surgery).

 

There are several sources of infections in the hypothalamic-pituitary region (Table 2). Dissemination from the sphenoid sinus to the pituitary is possible by direct contact and through shared venous drainage.

 

Table 2. Sources of Infections Spreading to the Hypothalamic-Pituitary Region

Spread

Comments

·       Hematogenous spread

In immunocompromised host

·       Direct extension from adjacent anatomical sites

Meningeal infection, sphenoid sinus, cavernous sinus, skull

·       Previous infectious diseases of the CNS

 

·       Iatrogenic

Surgical intervention in sellar and suprasellar region, tooth extraction

 

Infections in the hypothalamic-pituitary region are rare and several predisposing factors have been identified (2) (Table 3).

 

Table 3. Predisposing Factors for Hypothalamic-Pituitary Infections

·       Diabetes mellitus

·       Tuberculosis

·       Solid organ transplantation (renal, liver, etc.)

·       Human immunodeficiency virus (HIV) infection

·       Non-Hodgkin lymphoma

·       Chemotherapy

·       Cushing´s syndrome

·       Previous pituitary surgery

·       Immunosuppressive therapy

 

Infections of the hypothalamic-pituitary region may present with neurological signs and symptoms and signs of neuroendocrine dysfunction (Table 4).

 

Table 4. Clinical Features of Hypothalamic-Pituitary Infections

NEUROLOGICAL SYMPTOMS

ENDOCRINE DYSFUNCTION

Headache

Hyponatremia

Visual disturbances

Hypopituitarism

Cranial neuropathy (III, IV, VI)

Hypogonadotropic hypogonadism

 

Isolated ACTH deficiency

 

Hyperprolactinemia

 

Central diabetes insipidus

 

HYPOTHALAMIC-PITUITARY BACTERIAL INFECTIONS

 

Pituitary Abscess

 

Pituitary abscesses are rare pituitary lesions accounting for less than 1% of all pituitary lesions (6, 7). The first case of a pituitary abscess was described in 1848 and since then it has been mostly described in case reports or small series. In two-thirds of patients, pituitary abscesses occur in previously healthy normal glands (primary pituitary abscess) (8). In other patients, there is a preexisting lesion in the pituitary region, such as a pituitary adenoma, Rathke´s cleft cyst, granulomatous hypophysitis, or craniopharyngioma or prior pituitary surgery (secondary pituitary abscess) (5, 9-12). The infection can be caused by hematogenous dissemination or by direct extension from surrounding structures (meningitis, sphenoid sinusitis, cavernous sinus thrombophlebitis) (Table 2). Pituitary surgery and immunocompromised condition are also risk factors for pituitary abscesses (Table 3).

 

According to the clinical presentation and duration of the disease, pituitary abscesses can be acute, subacute (the disease course less than 1 month), or chronic (disease course longer than 1 month) (13). Infective manifestations (fever, leukocytosis, meningism) have been reported in patients with acute and subacute pituitary abscesses, while chronic pituitary abscesses have a more indolent course.

 

Many patients with pituitary abscess were misdiagnosed as having a pituitary adenoma, pituitary adenoma with apoplexy, or Rathke´s cleft cyst prior to surgery (14). The diagnosis of this potentially life-threatening disease is based on intraoperative detection of pus and postoperative histopathological analysis. Clinically, pituitary abscesses usually present with neurological signs and symptoms (headache, visual disturbances), signs of neuroendocrine dysfunction (anterior hypopituitarism, AVP deficiency) and signs and symptoms related to infections (fever, leukocytosis) (5, 8, 14). The largest series of primary pituitary abscesses (84 patients) during a 20-year period reported asthenia as the most common clinical presentation (75%), followed with visual impairment (71%), and headache (50%) (6).

 

Compared to patients with a pituitary adenoma who rarely have neuroendocrine dysfunction, most patients with pituitary abscesses have hypopituitarism (8, 9, 13). The large case series of 66 pituitary abscesses reported anterior pituitary hypopituitarism in 81.8% of patients, while AVP deficiency was diagnosed in 47.9% of patients (8). Nine percent of patients (9.3%) had isolated hypogonadism, 3.7% had isolated ACTH deficiency, 1.8% had isolated hypothyroidism, and 1.8% had combined hypogonadism and ACTH deficiency (8). The possible source of the pituitary infection was found in 14 out of 66 patients (sepsis, sinusitis, pulmonary tuberculosis) (8). Recently published the largest series of 84 patients with primary pituitary abscesses confirmed the high incidence (73%) of preoperative neuroendocrine dysfunction in patients with pituitary abscess: panhypopituitarism in 46%, isolated corticotropic insufficiency in 13%, isolated thyrotropic insufficiency in 10%, isolated gonadotropic insufficiency in 8%, and combined two pituitary axes insufficiencies in 22% (6).

 

On MRI, pituitary abscesses present as a sella masses, hypointense or isointense on T1-weighted imaging, hyperintense or isointense on T2-weighed imaging, with typical rim enhancement after gadolinium injection, mimicking apoplexy of the pituitary adenoma or other cystic sella lesions (7-9, 13, 15-17; Fig. 1). Diffusion-weighted imaging (DWI) sequences of pituitary abscesses often demonstrate high signal intensity with a reduction in the apparent diffusion coefficient, different from necrotic brain tumors (18).

Figure. 1. Pituitary abscess: gadolinium-enhanced T1-weighted MRI scan (sagittal view) shows sellar and suprasellar mass with peripheral contrast enhancement.

 

Neuroimaging with nuclear medicine investigations (18-FDG PET scan, labelled leukocyte scintigraphy) could increase the preoperative diagnostic rate in challenging patients with pituitary abscess (6).

 

The majority of patients are treated with transsphenoidal surgery, rarely with a transcranial approach (8). Intraoperatively, pus is found in the sella (19, 20). In patients with secondary pituitary abscesses, the sphenoid sinus is the most common site of extrasellar invasion (5).

On histopathological analysis, there is evidence of acute or chronic inflammation, while Gram staining and bacterial cultures in some cases may identify the infecting pathogen. In most cases, the etiological agents cannot be isolated (6, 12). In the two largest studies on pituitary abscesses, positive results on gram staining or bacterial cultures were found in only 19.7% and 25% of patients, respectively (6, 8). The most prevalent organisms are Gram-positive cocci (Staphylococcus Aureus and Streptococcus species), but also Gram-negative bacteria (Neisseria, Esherichia coli, Pseudomonas, Brucella) (5, 6, 9, 21).

 

Patients with bacterial pituitary abscesses are treated with intravenous and oral antibiotics for three to six weeks to prevent the recurrence of the pituitary abscesses, but in some cases, reoperation was required. In rare cases, AVP deficiency and hypopituitarism are reversible, occasionally followed by secondary empty sella (22). In most cases, neuroendocrine dysfunction and AVP deficiency are irreversible findings (6, 13). The preoperative diagnosis of pituitary abscess represented a protective factor for pituitary function recovery (in 23% of patients) (6).

 

Although pituitary abscesses are more indolent than other intracranial abscesses, secondary pituitary abscesses in patients with pituitary adenomas (not in Rathke´s cleft cyst) is associated with high mortality rate (26%) due to the dissemination of the infection or meningitis (5). In patients with infected Rathke´s cleft cyst, clinical manifestations are commonly subacute, without septic symptoms (9, 11).

 

Recently published the largest systematic review of 488 cases of pituitary abscess examined presentation, radiological findings, endocrinological abnormalities, treatment, and mortality of these patients (23). The most common symptoms were headache (76%) and visual field loss. The median time from onset of symptoms to presentation was 120 days. Symptoms and biochemical markers of infection were absent in 57% of cases. The appearance of hypointensity on T1 weighted images and hyperintensity on T2 weighted images, as well as peripheral contrast enhancement of the pituitary on MRI were the most common radiological findings. Fifty-five percent of patients had negative culture results. Endocrinological abnormalities were present in 84.5% of cases (panhypopituitarism in 41%, AVP deficiency in 25%) and persisted in over half of cases. The mortality rate was 5.1%, with delayed presentation increasing risk of mortality (23).

 

Hypopituitarism Caused by Treponema Pallidum Infection (Syphilis)

 

Syphilis is a sexually transmitted chronic bacterial infection caused by Treponema Pallidum which progresses over years through a series of clinical stages. Syphilis is a well-recognized cause of hypopituitarism, with granulomatous hypophysitis (noncaseating giant cell granuloma), syphilitic gumma in sella region, or congenital syphilis causing hypothalamic-pituitary dysfunction. The first cases of hypopituitarism caused by syphilis were described almost 70 years ago, mostly in postmortem cases (24). The use of penicillin caused a decline in syphilis presentations and late complications, including congenital syphilis. Nowadays, the incidence of syphilis has been rising again and this sexually transmitted disease should be considered again in the differential diagnosis of neurological, psychiatric, and endocrine cases in high-income countries in risk groups (HIV positive patients, men-men relationships, crack cocaine users, and among intravenous drug users) (25, 26). Spirochete Treponema Pallidum is also described as a cause of hypophysitis and pituitary gland enlargement with hypopituitarism, mostly in immunocompromised patients (HIV-infected patients) with syphilitic meningitis (27). Syphilis may cause a sella mass with suprasellar extension mimicking a pituitary tumor and causing severe headache and hypopituitarism (28). The diagnosis is confirmed by positive treponemal antibody or by detection of Treponema Pallidum by immunohistochemistry or PCR on the resected pituitary. This disorder is treated with antibiotics.

 

Hypopituitarism Caused by Leptospira Interrogans Infection (Sy Weil)

 

Leptospirosis is a common tropical febrile, zoonotic infectious disease caused by spirochete Leptospira Interrogans. The bacteria are spread through the urine of infected animals (cattle, pigs, horses, dogs, rodents, wild animals). Humans can become infected through contact with urine or other body fluids from infected animals or through contact with water, soil, or food contaminated with the urine of infected animals. This disease presents with hepatorenal syndrome and systemic hemorrhagic manifestations. The first case of pituitary apoplexy and panhypopituitarism caused by leptospirosis in a 56-year-old male with type 2 diabetes mellitus was recently published (29). The patient developed fever, nausea, vomiting and acute kidney injury. Leptospirosis was diagnosed by positive leptospira antibody test, and he started treatment with antibiotics. After five days of admission, he developed signs and symptoms of pituitary apoplexy. A brain MRI scan was consistent with apoplexy in a pituitary adenoma (the mass showed T2W hyper intensity and TIW isointensity with hypo intense areas which suggested hemorrhage). The patient developed hypopituitarism and was replaced with glucocorticoids and thyroid hormones. The follow-up MRI scan showed resolution of the hemorrhagic focus and regression of the pituitary adenoma. The proposed mechanism of pituitary apoplexy is platelet dysfunction (caused by uremia and directly by leptospira) and non-inflammatory vasculopathy (increased vascular permeability due to disruption of endothelial cell-cell junctions, cell retraction and opening of intercellular gaps) (29). Leptospirosis clinically presents very similar to another zoonosis, hantavirus infection, is more common worldwide and serology and PCR are necessary to distinguish between these two diseases (30).

 

Hypothalamic-Pituitary Dysfunction Following Bacterial CNS Infections

 

Hypothalamic-pituitary dysfunction is a well-recognized complication of acute infectious diseases of the central nervous system (meningitis and encephalitis) and may occur in the acute phase or in the late stage of these diseases (1, 4, 31, 32). The clinical spectrum of neuroendocrine dysfunction may range from an isolated pituitary hormone deficiency to panhypopituitarism. Endocrine dysfunction in the acute phase of meningitis may return to normal after the acute period or be irreversible (32). The most common deficit is isolated GH deficiency diagnosed 6-48 months after the infection, reported at a rate of 28.6% (31).

 

Hypopituitarism following acute viral or bacterial meningitis in children is not as common as in adulthood (33, 34). The GH neurosecretory dysfunction (low IGF1 with normal GH response in clonidine test) was found in 3 out of 37 children tested at least 6 months following the diagnosis of bacterial meningitis (34). There are rare case reports on hypopituitarism during acute meningitis caused by Streptococcus Group B or sepsis caused by Salmonella enteritidis in a neonatal period (35, 36).

 

The pathophysiological mechanism responsible for hypothalamic-pituitary dysfunction following acute meningitis is not fully understood. In some patients, anti-pituitary and anti-hypothalamus antibodies are detected (37). It is proposed that acute infection provokes an autoimmune process and may cause axonal injury with consequent neuroendocrine dysfunction (38).

 

Idiopathic Granulomatous Inflammation of the Cavernous Sinus - the Tolosa-Hunt Syndrome      

 

Tolosa-Hunt syndrome is defined as an idiopathic granulomatous inflammation of the cavernous sinus, therefore not infectious in origin, with variable extension into the superior orbital fissure/orbital apex, usually unilateral. The diagnosis is made by exclusion of other more common causes of cavernous sinus lesions (thrombosis, tumors, fungal infections, systemic granulomatous diseases-sarcoidosis, tuberculosis, Wegener´s granulomatosis) (39). In less than 5% of cases it can be bilateral, mimicking a pituitary adenoma in imaging studies (40). The etiology of Tolosa-Hunt syndrome is not fully understood. The disease is characterized by nonspecific granulomatous inflammation with infiltration of lymphocytes and plasmocytes. The patient presents with severe unilateral orbital pain and ipsilateral ocular motor neuropathy. The paralysis of one or more cranial nerves passing through the cavernous sinus (III, IV, VI) develops with orbital pain after less than 2 weeks. The signs of infection (fever, leukocytosis) are usually present. Granulomatous inflammation develops within the cavernous sinus causing acute throbbing orbital pain and disordered eye movement. Brain MRI scan demonstrate the inflammation in the cavernous sinus, orbital apex, and rarely in the sella. MR venography of the brain is important to exclude cavernous sinus thrombosis. Treatment consists of high dose corticosteroids and antibiotics. Refractory and steroid-intolerant cases may be treated with immunosuppressants (Methotrexate or Azathioprine) and gamma knife radiotherapy (41). Periorbital pain intensity is rapidly decreasing and resolving within 72h, while the resolution of ophthalmoplegia improves gradually and takes a longer time to resolve (several weeks) (42). If the patient is not responding to standard therapy, biopsy of the lesion is necessary to exclude other diseases, such as lymphoma.

 

Hypothalamic-Pituitary Tuberculosis

 

The incidence of tuberculosis is rising not only in developing countries, but also in developed countries, especially given the increasing population migration. Mortality has begun to increase after years of decline and since 2018 more than 7 million people have died of tuberculosis (43). Extrapulmonary tuberculosis may affect the brain causing tuberculous meningitis and tuberculoma of the central nervous cases. Tuberculous meningitis has a tendency to affect basal parts of the brain from where it can spread to the sella region. In rare cases, CNS tuberculosis may present as tuberculous hypophysitis or sella/suprasellar tuberculoma mimicking a pituitary adenoma or pituitary apoplexy (44-48). It may occur in the absence of systemic tuberculosis, but the majority of patients have a past history of pulmonary tuberculosis or tuberculosis of other organs (spine). Tuberculosis may affect the hypothalamus, pituitary, paranasal sinuses (sphenoid sinus), or tuberculoma may be located only in the pituitary stalk. Hypothalamic-pituitary dysfunction and AVP deficiency during the acute phase or years after recovery from acute tuberculous meningitis suggests a more destructive and more extensive hypothalamic and pituitary damage compared to other causes of acute viral and bacterial meningitis.

 

A significant proportion of patients with sella tuberculoma or tuberculous meningitis develop hypothalamic-pituitary dysfunction. In 18 cases of histologically proved sella tuberculoma (5 of them with past history of tuberculosis), 5 patients had hypopituitarism and 3 had hyperprolactinemia due to pituitary stalk compression (44).

 

In patients with tuberculous meningitis half of them developed neuroendocrine dysfunction: hyperprolactinemia (23-50% of patients), hypocortisolism (13-43%), hypothyroidism (17-31%), hypogonadism (34%), SIADH (10%) (4, 49, 50) during the acute phase. Multiple hormonal axes were affected in 23.5% of patients (49, 50). In young adult patients who survived tuberculous meningitis in childhood and were tested several years after recovery, hypopituitarism was diagnosed in 20% of patients (51). This is a consequence of fibrosis, gliosis, and calcification in the hypothalamus and pituitary after recovery of active tuberculous brain infection. In rare cases, pituitary function recovered after successful treatment with anti-tuberculous drugs (52).

 

The diagnosis of sella tuberculoma is a challenge, especially in cases without systemic tuberculosis. The signs and symptoms are nonspecific: fever, neurological abnormalities (headache, visual disturbances), and neuroendocrine dysfunction (44, 53). Sellar tuberculoma on MRI scans presents as thickening of the pituitary stalk or abnormal enhancement pattern of the sella lesion (54) (Fig. 2).

 

Figure 2. Tubercular hypophysitis: sellar MRI scan (coronal view) shows stalk thickening.

 

If the correct diagnosis is established, anti-tuberculosis drugs are effective and surgery is not indicated for tuberculous hypophysitis. With surgery the histological examination shows granulomas with central caseous necrosis surrounded by giant Langhans cells. In cases in whom Ziehl-Nielsen staining for acid fast bacilli and the culture on Lowenstein-Jensen media are negative, PCR for detection of mycobacterial DNA in tissue or CSF may help.

 

HYPOTHALAMIC-PITUITARY FUNGAL INFECTIONS

 

Hypothalamic-pituitary fungal infections are extremely rare and occur usually in immunocompromised patients (diabetes mellitus, granulocytopenia, solid organ transplantation). There are only few case reports of Candida and Aspergillus sella abscesses and reviews of fungal sellar abscesses (55-61). Aspergillus is a ubiquitous saprophitic fungus found in the nasal mucosa of healthy people and patients with chronic sinusitis. This fungus may cause CNS infection, such as meningitis, encephalitis, brain abscess, subdural abscess, pituitary abscess, and mycotic arteritis with thrombosis and aneurysm (61-63). Fungal sella abscesses have a nonspecific presentation, with neurological signs and symptoms (headache, visual disturbances) and hypothalamic-pituitary dysfunction. Sella MRI images are nonspecific, with a T1W hypointense or isointense mass with rim enhancement and may be misdiagnosed as a pituitary adenoma (Fig. 3). It is proposed that a low signal on T2W images due to iron deposition may be a more specific sign of fungal abscess (58, 64; Fig. 4). Diagnosis of fungal pituitary abscess is made by histopathological finding (Grocott–Gömöri's methenamine silver stain demonstrates septate fungal hyphae), cultivation, or PCR identification of fungus DNA. A combination of transsphenoidal drainage and antifungal therapy (liposomal Amphotericin B, itraconazole, voriconazole, caspofungin, micafungin) can result in a good prognosis (55, 59, 60, 65). Endocrinopathies caused by fungal abscess have a low rate of recovery (59).

 

Figure 3. Fungal pituitary abscess spreading from fungal sinusitis: sellar MRI scan (coronal and sagittal views) shows a large sellar mass pushing the pituitary upwards.

Figure 4. Fungal infections in the sella: a) CT scan of the sinuses (axial view) shows a large sellar mass, erosion in sellar floor, propagation of the pathological process and opacification of the sinuses, and B) sellar MRI scan (coronal view) shows a giant hypointense lesion in the sellar region.

 

An unusual form of allergic fungal sinusitis which expands from the sphenoid sinus through a bone erosion to the sella in immunocompetent patient has been described (64) (Fig. 4). This patient had hyperprolactinemia (in the setting with no pituitary stalk compression), which resolved after successful transsphenoidal operation followed by anti-mycotics and corticosteroids (64). It is speculated that fungal glucans may directly stimulate glucan-specific receptors on somatomammotroph cells to stimulate prolactin secretion (66).

 

HYPOTHALAMIC-PITUITARY VIRAL INFECTIONS

 

Hypothalamic-pituitary dysfunction (hypopituitarism and cranial diabetes insipidus) may develop in the acute phase of viral infections of the CNS (meningitis and encephalitis) or in the late stage of these diseases (1, 31, 32). Infectious agents which cause CNS viral infections are listed in Table 5.

 

Table 5 – Infectious Agents Which Cause CNS Viral Infections

MENINGITIS

ENCEPHALITIS

Herpes virus

Tick-borne

Varicella

Herpes simplex

Enterovirus

Cytomegalovirus

 

Neuroborreliosis

SARS-CoV-2 virus

SARS-CoV-2 virus

 

The investigation of hypothalamic-pituitary function at least 6 months after recovery from mild-to-moderate meningitis/encephalitis showed that 21% patients developed isolated corticotroph deficiency, while other neuroendocrine abnormalities or AVP deficiency were not found (1).

 

Hantavirus

 

Hemorrhagic fever with renal syndrome (HFRS), caused by Hantaviruses in the Bunyaviridae family, is an endemic zoonotic disease transmitted by rodents. There are several serotypes of these RNA viruses causing systemic infection, milder form called nephropathia endemica (Puumala) or severe form (Dobrava, Belgrade). The disease is endemic in Europe (Balkans, Finland, Germany) and Asia (Korea), where several outbreaks have been recorded. Farmers and solders are exposed to the virus by inhalation of infected rodent urine, feces, or saliva. Hantavirus infiltrates the vascular system causing increased capillary permeability, renal failure, thrombocytopenia, hemorrhage, fever, hypotension, and shock. The mortality rate is 6.6%. Autopsy findings reported a slightly enlarged pituitary with ischemia/infarction, hemorrhage, and necrosis (67-69). Direct viral invasion was confirmed in the pituitary causing viral hypophysitis (69). Hypothalamic-pituitary dysfunction has been reported during the acute phase of the disease or after long-term follow-up (70-79). A milder form of HFRS infection caused by Puumala virus (nephropathia epidemica) is associated with a lower incidence of hypopituitarism (80). It is speculated that in some patients with no signs of hemorrhage in the sella, hantavirus may cause autoimmune hypophysitis and hypopituitarism (81). Sellar MRI imaging in hypopituitary patients reveals an edematous pituitary gland or increased signal intensity in the pituitary due to hemorrhage during the acute phase, while pituitary atrophy and secondary empty sella develops months and years after acute infection (76, 79-81) (Fig 5). A retrospective study of 60 adults who had recovered from HFRS reported that 18% of patients developed hypopituitarism (82). Ten percent of patients had a single pituitary deficit (three GH, two gonadal, and one adrenal), and 8.3% had multiple pituitary hormone deficiencies (82). In rare cases, HFRS may cause injury of the pituitary stalk or acute/subacute hemorrhage in the pituitary gland and AVP deficiency with panhypopituitarism (30, 77).

 

Figure. 5. Hemorrhagic fever with renal syndrome: sellar MRI scan (sagittal view) shows pituitary atrophy and secondary empty sella.

 

SARS-CoV-2

 

The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was identified as a cause of coronavirus disease 2019 (COVID-19). This virus is responsible for a variety of clinical manifestations, ranging from an asymptomatic stage to severe pulmonary disease (respiratory distress syndrome) and various extrapulmonary manifestations, including endocrine axes (83-85).

 

SARS-CoV-2 is a neuroinvasive virus which enters the brain through the nasopharyngeal epithelium via the olfactory nerve, passes through the blood-brain barrier, or enters the brain through the median eminence where this barrier is absent (86). Neurons, glial cells, and cerebrovascular endothelia cells, as well as hypothalamus and pituitary cells express an angiotensin-converting enzyme 2 (ACE2) receptor responsible for the entry of the virus into these cells which induce neuroinflammation (86-88). SARS-CoV-2 virus is also detected in cerebrospinal fluid of patients with encephalitis caused by COVID-19 and in pituitary tissues from patients who died from SARS (89, 90). SARS-CoV-2 is responsible for a variety of neurological complications, including headache, anosmia, confusion, ataxia, neuropathic pain, seizures, and delirium (89).

 

There are also indirect systemic effects of COVID-19 virus: an altered immune response (cytokine storm), infection-induced thrombocytopenia, platelet dysfunction, coagulopathy (hypercoagulable state), endothelitis, and endothelial dysfunction (91). All these direct and indirect systemic effects of COVID-19 virus may be responsible for ischemic and hemorrhagic vascular syndromes, affecting also the hypothalamo-pituitary region. Another possible indirect effect of COVID-19 may be mediated by cytokines that can trigger hypothalamo-pituitary inflammation with consequent neuroendocrine dysfunction (92). Also, it has been shown that SARS-CoV-2 express an amino-acid sequence that mimics human ACTH and induces the production of autoantibodies against ACTH causing cortisol insufficiency and inadequate adrenal response to the stress (83).

 

SARS-CoV-2 infection may involve the hypothalamo-pituitary axis causing pituitary apoplexy, hypophysitis, and hyponatremia (83, 85, 91, 93-97).

 

There are nine case reports of pituitary adenoma (6 males/3 females, between 27 and 56 years of age, eight with pituitary macroadenoma) and two series of three patients with pituitary adenomas, complicated by apoplexy during COVID-19 infection (93-102). In one patient this occurred during the third trimester of pregnancy. Four patients had transsphenoidal surgery and recovered, one patient had transcranial resection, three patients were conservatively managed, and one patient died 12 hours after admission. There is also a report of a 65-year-old woman with no underlying pituitary disease who developed acute pituitary apoplexy one month after the initial diagnosis of COVID-19 (103). She developed anterior hypopituitarism with no evidence of AVP deficiency. An MRI pituitary scan showed the resolution of intrapituitary hemorrhage, with normal size of the pituitary gland after six months of follow-up. A small case series reported three patients with previously unknown pituitary adenoma complicated by apoplexy during COVID-19 infection: a 54-year-old female with null cell adenoma, a 52-year-old and a 56-year-old men, both obese with hypertension, both with a lactotroph pituitary adenoma (94). In another series of three patients with pituitary macroadenoma complicated with apoplexy following a symptomatic COVID-19 infection, two of these patients’ developed symptoms of pituitary apoplexy days following the viral infection, whereas the third patient developed pituitary apoplexy after a 2-month period (96).

 

In some patients COVID-19 caused an acute lymphocytic hypophysitis with temporal evolution of symptoms, suggesting an immune-mediated parainfectious pattern of disease (104, 105). An 18-year-old previously healthy girl with a history of symptomatic COVID-19 three weeks prior to the acute onset of headache and dizziness, presented with diffuse thickening and enlargement of the infundibulum with homogenous contrast enhancement of the pituitary (104). She was treated with glucocorticoids with a significant clinical improvement on day 3 and complete resolution of MRI finding on day 5. A similar case is a  16-year-old girl who presented with headaches, polyuria/polydipsic syndrome, and impaired visual acuity three weeks after COVID-19 infection (105). She had pituitary enlargement on MRI, was treated with methylprednisolone and improved on day 5. Such a rapid headache resolution after steroid treatment suggests a transitory acute hypophysitis, an immune-mediated process, triggered by viral infection. It has been hypothesized that SARS-CoV-2 may induce hypothalamo-pituitary autoimmunity, with positive anti-hypothalamus (AHA) and anti-pituitary (APA) antibodies (106). The current results in the literature about antigens which are targets of AHA and APA are still contradictory (83). It is possible that patients at high-risk for such a complication are carriers of specific HLA alleles. The human leukocyte antigen (HLA) complex has a central role in the recognition and presentation of viral antigens to immune system. HLA class I molecules mediates innate defense strategies against viral infection. HLA-c 04:01, DRB1 08, DQB1 06 carriers have an increased risk of a severe clinical course of COVID-19 and generate a robust response and susceptibility to autoimmune diseases (107-110).

 

Even these two acute pituitary disorders (pituitary apoplexy with transition to acute hypophysitis) can occur in patients with COVID-19 and preexisting unknown pituitary adenoma (97).

 

Several cases of AVP deficiency as a late complication of COVID-19 infection were published (111-114). All patients (2 males/2 females, 28-60 years of age) suddenly developed polyuria, nocturia, and polydipsia four to eight weeks after the diagnosis of COVID-19 infection. In one of the published cases, a sellar MRI scan showed an enlarged pituitary with an absent posterior pituitary bright spot on T1W images associated with thickening of the pituitary stalk of 3.5 mm suggesting infundibulo-neurohypophysitis (113). Other pituitary hormone evaluations were normal. They were treated with oral desmopressin.

 

A case of a young woman with hypothalamic amenorrhea following COVID-19, with normal appearance of the sella turcica and regular dimensions of the pituitary was reported (95). This case suggests clinicians need to follow patients for possible delayed neuroendocrine dysfunctions after COVID-19 infection.

 

Hyponatremia was reported in 30-60% of patients with SARS-CoV-1 infection and was associated with worse outcomes and increased mortality (115). The syndrome of inappropriate anti-diuretic hormone secretion (SIADH) was the most common reason for hyponatremia, followed by adrenal insufficiency (116).

 

There are also data on the association between COVID-19 vaccination and the subsequent development of pituitary diseases (85, 117-120). A recently published systematic review analyzed 23 case reports of post COVID-19 vaccination pituitary diseases: hypophysitis in 9 patients, pituitary apoplexy in 6 patients, SIADH in 5 patients, and isolated ACTH deficiency in two patients (119). Symptoms of pituitary disease typically occurred shortly (several days) after vaccine administration and the pathogenetic mechanisms potentially include molecular mimicry, vaccine adjuvants, and vaccine-induced thrombotic thrombocytopenia (83, 119). The presence of ACE2 receptors in the hypothalamo-pituitary system contributes to these post vaccinal pituitary diseases. Isolated infundibulo-neurohypophysitis and AVP deficiency or isolated ACTH deficiency may also develop several days after immunization with BNT 162b2 mRNA COVID-19 vaccine (117, 121). In some cases of panhypopituitarism and AVP deficiency due to hypophysitis after COVID-19 vaccination, hormonal secretion partially improved during follow-up (120).

 

Other Viruses

 

Cytomegalovirus, herpes simplex, varicella zoster, and enterovirus have also been described in very rare cases of central diabetes insipidus, mainly in immunocompromised patients with encephalitis (such as HIV infection, Cushing´s syndrome, lymphoma or immunosuppressive therapy) (122-126). Direct cytomegalovirus invasion and reduction in the number of AVP and oxytocin cells was confirmed in the hypothalamus (123).

 

HYPOTHALAMIC-PITUITARY PARASITIC INFECTIONS

 

Parasitic infections of the pituitary are rare and infections in the sellar region caused by Toxoplasma gondii, Echinococcus species, and Taenia solium have been reported anecdotally (4).

 

Toxoplasmosis is a worldwide zoonosis, caused by the protozoan parasite Toxoplasma gondii. This is one of the most common parasitic infections of warm-blooded animals and humans. Approximately one-third of humans have been exposed to T. gondii, mostly with no serious diseases, except in immunocompromised patients (HIV) and congenital toxoplasmosis. Two cases of prolactinomas with T. gondii cysts among tumor cells were reported (127). Toxoplasmosis is the most common CNS infection in immunocompromised patients (patients with HIV infection) and may cause hypopituitarism, accompanied by focal neurological deficits, headache, and fever (128, 129). The brain MRI shows lesions with significant enhancement of T2W images and peri-lesional edema, which may be misdiagnosed as intracranial metastasis. The diagnosis is based on brain biopsy with confirmed presence of T. gondii by PCR. Infection with T. gondii is treated with antimicrobial therapy and with hormone replacement therapy as needed.

 

The most common parasitic infestation of the brain is neurocysticercosis, caused by Taenia solium. A systematic review of 23 patients with intrasellar cysticercosis reported endocrine abnormalities (panhypopituitarism, hyperprolactinemia, AVP deficiency, and isolated hypothyroidism) in 56% of the affected population (130). These infections present with a cystic mass in the sella with hypopituitarism caused by compression, subarachnoid cysts, obstructive hydrocephalus, or neuroinflammation (ventriculitis, focal arachnoiditis) (4, 131). Neurocysticercosis may involve the pituitary stalk too (132). Transsphenoidal or transcranial operation is required for the definitive histopathological diagnosis and cure, because medical therapy with praziquantel is usually ineffective (130).

 

ACKNOWLEDGMENT

 

This study was supported by The Science Fund of the Republic of Serbia, Grant No: 7754282 — Prediction, prevention and patient’s participation in diagnosis of selected fungal infections (FI): an implementation of a novel method for obtaining tissue specimens, “FungalCaseFinder”.

 

REFERENCES

 

  1. Schaefer S, Boegershausen N, Meyer S, Ivan D, Schepelmann K, Kann PH. Hypothalamic-pituitary insufficiency following infectious diseases of the central nervous system. Eur J Endocrinol 2008; 158: 3-9.
  2. Pekic S, Popovic V. Alternative causes of hypopituitarism: traumatic brain injury, cranialirradiation, and infections.Handb Clin Neurol 2014; 124: 271-290
  3. Pekic S, Popovic V. Diagnosis of endocrine disease: Expanding the cause of hypopituitarism. Eur J Endocrinol2017; 176: R269-R282
  4. Das L, Dutta P. Approach to the patient: a case with an unusual cause of hypopituitarism. J Clin Endocrinol Metab 2023; 108: 1488-1504. 
  5. Awad AJ, Rowland NC, Mian M, Hiniker A, Tate M, Aghi MK. Etiology, prognosis, and management of secondary pituitary abscesses forming in underlying pituitary adenomas. J Neurooncol 2014; 117: 469–476
  6. Mallereau CH, Todeschi J, Ganau M, Cebula H, Bozzi MT, Romano A, Le Van T, Ollivier I, Zaed I, Spatola G, Nannavecchia B, Mahoudeau P, Djennaoui I, Debry C, Signorelli F, Ligarotti GKI, Pop R, Baloglu S, Fasciglione E, Goichot B, Bund C, Gaudias J, Proust F, Chibbaro S. Pituitary abscess: a challenging preoperative diagnosis-a multicenter study. Medicina (Kaunas) 2023; 59: 565.
  7. Naser PV, Papadopoulou P, Teuber J, Kopf S, Jesser J, Unterberg AW, Beynon C. Characteristics of inflammatory and infectious diseases of the pituitary gland in patients undergoing transsphenoidal surgery. Pituitary 2023; 26: 451-460. 
  8. GaoL, Guo X, Tian R, Wang Q, Feng M, Bao X, Deng K, Yao Y, Lian W, Wang R, Xing B. Pituitary abscess: clinical manifestations, diagnosis and treatment of 66 cases from a large pituitary center over 23 years. Pituitary 2017; 20: 189-194
  9. Aranda F, Garcia R, Guarda FJ, Nilo F, Cruz JP, Callejas C, Balcells ME, Gonzales G, Rojas R, Villanueva P. Rathke´s cleft cyst infections and pituitary absesses: case series and review of the literature. Pituitary 2021; 24: 374-383
  10. Warmbier J, Lüdecke DK, Flitsch J, Buchfelder M, Fahlbusch R, Knappe UJ, Kreutzer J, Buslei R, Bergmann M, Heppner F, Glatzel M, Saeger W. Typing of inflammatory lesions of the pituitary. Pituitary 2022; 25: 131-142.
  11. Rabiei MM, Ebrahimzadeh K, Davoudi Z, Bidari Zerehpoosh F, Javandoust Gharehbagh F, Sedaghati R, Lotfollahi L, Kalhor F, Alavi Darazam I. A case of pituitary gland abscess associated with granulomatous hypophysitis. BMC Neurol 2023; 23:11.
  12. Inoue E, Kesumayadi I, Fujio S, Makino R, Hanada T, Masuda K, Higa N, Kawade S, Niihara Y, Takagi H, Kitazono I, Takahashi Y, Hanaya R. Secondary hypophysitis associated with Rathke's cleft cyst resembling a pituitary abscess. Surg Neurol Int 2024; 15: 69.
  13. LiuF, Li G, Yao Y, Yang Y, Ma W, Li Y, Chen G, Wang R. Diagnosis and management of pituitary abscess: experiences from 33 cases. Clin Endocrinol 2011; 74: 79-88
  14. Xue Q, Shi X, Fu X, Yin Y, Zhou H, Liu S, Sun Q, Meng J, Bian L, Tan H, He H. Pituitary abscess: a descriptive analysis of a series of 19 patients-a multi-center experience. Eur J Med Res 2024; 29: 262.
  15. Vates GE, Berger MS, Wilson CB. Diagnosis and management of pituitary abscess: a review of twenty-four cases. J Neurosurg 2001; 95: 233-241
  16. Ciappetta P, Calace A, D'Urso PI, De Candia N. Endoscopic treatment of pituitary abscess: two case reports and literature review. Neurosurg Rev 2008; 31: 237-246
  17. Dalan R, Leow MK. Pituitary abscess: our experience with a case and a review of the literature. Pituitary 2008; 11: 299-306
  18. Takao H, Doi I, Watanabe T. Diffusion-weighted magnetic resonance imaging in pituitary abscess. J Comp Assist Tomogr 2006; 30: 514–516
  19. Zhang X, Sun J, Shen M, Shou X, Qiu H, Qiao N, Zhang N, Li S, Wang Y, Zhao Y. Diagnosis and minimally invasive surgery for the pituitary abscess: A review of twenty nine cases. Clin Neurol Neurosurg 2012; 114: 957-961
  20. Al Salman JM, Al Agha RAMB, Helmy M. Pituitary abscess. BMJ Case Rep 2017; 2017
  21. De la Peña-Sosa G, Cabello-Hernández AI, Gómez-Ruíz RP, Gómez-Sámano MA, Gómez-Pérez FJ. Pituitary abscess causing panhypopituitarism in a patient with neurobrucellosis: case report. AACE Clin Case Rep 2023; 10: 10-13.  
  22. Pepene CE, Ilie I, Mihu D, Stan H, Albu S, Duncea I. Primary pituitary abscess followed by empty sella syndrome in an adolescent girl. Pituitary 2010; 13: 385-389
  23. Stringer F, Foong YC, Tan A, Hayman S, Zajac JD, Grossmann M, Zane JNY, Zhu J, Ayyappan S. Pituitary abscess: a case report and systematic review of 488 cases. Orphanet J Rare Dis 2023; 18: 165.
  24. Oelbaum M. Hypopituitarism in male subjects due to syphilis with a discussion of androgen treatment. Q J Med 1952; 83: 249–264
  25. Noblett JRoberts E. The importance of not jumping to conclusions: syphilisas an organic cause of neurological, psychiatric and endocrine presentations. BMJ Case Rep 2015; 2015.
  26. Hook EW 3rd. Syphilis. Lancet 2017; 389: 1550-1557.
  27. Spinner CD, Noe S, Schwerdtfeger C, Todorova A, Gaa J, Schmid RM, Busch DH, Neuenhahn M. Acute hypophysitis and hypopituitarism in early syphilitic meningitis in a HIV-infected patient: a case report. BMC Infect Dis 2013; 13: 481-484
  28. Bricaire L, Van Haecke C, Laurent-Roussel S, Jrad G, Bertherat J, Bernier M, Gaillard S, Groussin L, Dupin N. The great imitator in endocrinology: a painful hypophysitis mimicking a pituitarytumor. J Clin Endocrinol Metab 2015; 100: 2837-2840
  29. Gohil J, Gowda A, George T, Easwer HV, George A, Nair P. Pituitary apoplexy and panhypopituitarism following acute leptospirosis. Pituitary 2021 May 21; 1-5.
  30. Schwab S, Lissmann S, Schafer N, Isaak A, Klingmuller D, Attenberger U, Eis-Hubinger AM, Hofmann J, Strassburg C, Lutz P. When polyuria does not stop: a case report on an unusual complication of hantairus infection. BMC Infectious Diseases 2020; 20: 713
  31. Tanriverdi F, Alp E, Demiraslan H, Dokmetas HS, Unluhizarci K, Doganay M, Casanueva FF, Kelestimur F. Investigation of pituitary functions in patients with acute meningitis: a pilot study. J Endocrinol Invest 2008; 31: 489-491
  32. Tsiakalos A, Xynos ID, Sipsas NV, Kaltsas G. Pituitary insufficiency after infectious meningitis: a prospective study.J Clin Endocrinol Metab 2010; 957: 3277-3281
  33. Giavoli C, Tagliabue C, Profka E, Senatore L, Bergamaschi S, Rodari G, Spada A, Beck-Peccoz P, Esposito S. Evaluation of pituitary function after infectious meningitis in childhood. BMC Endocr Disord2014; 14: 80-84
  34. Karadag-Oncel E, Cakir M, Kara A, Gonc N, Cengiz AB, Ozon A, Ciftci E, Alikasifoglu A, Ceyhan M, Kandemir N. Evaluation of hypothalamic-pituitary function in children following acute bacterial meningitis. Pituitary 2015; 18: 1-7
  35. Saranac L, Bjelakovic B, Djordjevic D, Novak M, Stankovic T. Hypopituitarism occurring in neonatal sepsis. J Pediatr Endocrinol Metab 2012; 25: 847-848
  36. Ferreira AS, Fernandes AL, Guaragna-Filho G. Hypopituitarism as consequence of late neonatal infection by Group B streptococcus: a case report. Pan Afr Med J2015; 20: 308
  37. Tanriverdi F, De Bellis A, Teksahin H, Alp E, Bizzarro A, Sinisi AA, Bellastella G, Paglionico VA, Bellastella A, Unluhizarci K, Doganay M, Kelestimur F. Prospective investigation of pituitary functions in patients with acute infectious meningitis: is acute meningitis induced pituitary dysfunction associated with autoimmunity? Pituitary 2012; 5: 579–588
  38. Gerber J, Seitz RC, Bunkowski S, Bruck W, Nau R. Evidence for frequent focal and diffuse acute axonal injury in human bacterial meningitis. Clin Neuropathol 2009; 28:33–39
  39. BhatkarS, Goyal MK, Takkar A, Mukherjee KK, Singh P, Singh R, Lal V. Cavernous sinus syndrome: A prospective study of 73 cases at a tertiary care centre in Northern India. Clin Neurol Neurosurg 2017; 155: 63-69
  40. Swiątkowska-Stodulska R, Stodulski D, Babinska A, Piskunowicz M, Sworczak K. Bilateral Tolosa-Hunt syndrome mimicking pituitary adenoma. Endocrine 2017; 58: 582-586
  41. LeeJMPark JSKoh EJ. Gamma Knife radiosurgery in steroid-intolerant Tolosa-Hunt syndrome: case report. Acta Neurochir (Wien) 2016; 158: 143-145
  42. Arshad ANabi SPanhwar MSRahil A.Tolosa-Huntsyndrome: an arcane pathology of cavernous venous sinus. BMJ Case Rep 2015; 2015.
  43. Reid M, Agbassi YJP, Arinaminpathy N, Bercasio A, Bhargava A, Bhargava M, Bloom A,Cattamanchi A, Chaisson R, Chin D, Churchyard G, Cox H, Denkinger CM, Ditiu L, Dowdy D, Dybul M, Fauci A, Fedaku E, Gidado M, Harrington M, Hauser J, Heitkamp P, Herbert N, Herna Sari A, Hopewell P, Kendall E, Khan A, Kim A, Koek I, Kondratyuk S, Krishnan N, Ku CC, Lessem E, McConnell EV, Nahid P, Oliver M, Pai M, Raviglione M, Ryckman T, Schäferhoff M, Silva S, Small P, Stallworthy G, Temesgen Z, van Weezenbeek K, Vassall A, Velásquez GE, Venkatesan N, Yamey G, Zimmerman A, Jamison D, Swaminathan S, Goosby E. Scientific advances and the end of tuberculosis: a report from the Lancet Commission on Tuberculosis. Lancet 2023; 402: 1473-1498.
  44. Sharma MC, Arora R, Mahapatra AK, Sarat-Chandra P, Gaikwad SB, Sarkar C. Intrasellar tuberculoma--an enigmatic pituitary infection: a series of 18 cases. Clin Neurol Neurosurg 2000; 102: 72-77
  45. Arunkumar MJ, Rajshekhar V. Intrasellar tuberculoma presenting as pituitary apoplexy. Neurol India 2001, 49: 407-410
  46. Husain N, Husain M, Rao P. Pituitary tuberculosis mimicking idiopathic granulomatous hypophysitis. Pituitary 2008; 11: 313-315
  47. Aziz Khan A, Ahuja S, Malik S, Naaz S, Zaheer S. Pituitary tuberculoma with panhypopituitarism masquerading as a pituitary adenoma. Neuropathology 2023; 43: 496-499.
  48. Elfarissi MA, Dahamou M, Dehneh Y, Lhamlili M, Khoulali M, Oulali N, Moufid F. Pediatric sellar-suprasellar tuberculosis: A case report and review of the literature. Surg Neurol Int 2023; 14: 379. 
  49. Dhanwal DK, Vyas A, Sharma A, Saxena A. Hypothalamic pituitary abnormalities in tubercular meningitis at the time of diagnosis. Pituitary 2010; 13: 304-310
  50. More A, Verma R, Garg RK, Malhotra HS, Sharma PK, Uniyal R, Pandey S, Mittal M. A study of neuroendocrine dysfunction in patients of tuberculous meningitis. J Neurol Sci2017; 379: 198-206
  51. Lam KS, Sham MM, Tam SC, Ng MM, Ma HT. Hypopituitarism after tuberculous meningitis in childhood. Ann Intern Med 1993; 118: 701-706
  52. Tanimoto K, Imbe A, Shishikura K, Imbe H, Hiraiwa T, Miyata T, Ikeda N, Kuroiwa T, Terasaki J, Hanafusa T. Reversible hypopituitarism with pituitarytuberculoma. Intern Med 2015; 54: 1247-1251
  53. Jain A, Dhanwal DK, Kumar J. A rare case of pituitarytuberculoma-diagnosed and managed conservatively.Pituitary 2015; 18: 579-580
  54. Bonifacio-Delgadillo D, Aburto-Murrieta Y, Salinas-Lara C, Sotelo J, Montes-Mojarro I, Garcia-Marquez A. Clinical presentation and magnetic resonance findings in sellar tuberculomas. Case Rep Med2014; 2014: 961913. 
  55. Liu W, Chen H, Cai B, Li G, You C, Li H. Successful treatment of sellar aspergillus abscess. J Clin Neurosci 2010; 17: 1587-1589
  56. Vijayvargiya PJaved IMoreno JMynt MAKotapka MZaki ROrtiz J. Pituitaryaspergillosis in a kidney transplant recipient and review of the literature. Transpl Infect Dis 2013; 15(5): E196-200
  57. Hong WLiu YChen MLin KLiao ZHuang S. Secondary headache due to aspergillus sellar abscesssimulating a pituitaryneoplasm: case report and review of literature. Springerplus 2015; 4: 550
  58. Lim JC, Rogers TW, King J, Gaillard F. Successful treatment of pituitarysella Aspergillus abscess in a renal transplant recipient. J Clin Neurosci 2017; 45: 138-140
  59. Strickland BA, Pham M, Bakhsheshian J, Carmichael J, Weiss M, Zada G. Endoscopic endonasal transsphenoidal drainage of a spontaneous candidaglabrata pituitary abscess. World Neurosurg 2018; 109: 467-470
  60. Saffarian A, Derakhshan N, Taghipour M, Eghbal K, Roshanfarzad M, Dehghanian A. Sphenoid Aspergilloma with headache and acute vision loss. World Neurosurg 2018; 115: 159-161
  61. Funakoshi S, Nishiyama M, Komori M, Hyodo M, Kawanishi Y, Ueba T, Fujimoto S, Terada Y. Hypopituitarism due to CNS Aspergillus infection. Intern Med 2024 Mar 11. doi: 10.2169/internalmedicine.3390-23.
  62. Iplikcioglu AC, Bek S, Bikmaz K,Ceylan DGökduman CA. Aspergillus pituitary abscess. Acta Neurochir 2004; 146: 521-524
  63. Hao L, Jing C, Bowen C, Min H, Chao Y. Aspergillus sellar abscess: case report and review of the literature. Neurol India 2008; 56: 186-188
  64. Pekic S, Arsenijevic VA, Gazibara MS, Milojevic T, Pendjer I, Stojanovic M, Popovic V. What lurks in the sellar?Lancet 2010; 375: 432
  65. Ouyang TZhang NWang LJiao JZhao YLi ZChen J. Primary Aspergillus sellar abscesssimulating pituitary tumor in immunocompetent patient. J Craniofac Surg 2015; 26: e86-88
  66. Breuel KF, Kougias P, Rice PJ, Wei D, De Ponti K, Wang J, Laffan JJ, Li C, Kalbfleisch J, Williams DL. Anterior pituitary cells express pattern recognition receptors for fungal glucans: implications for neuroendocrine immune involvement in response to fungal infections. Neuroimmunomodulation 2004; 11: 1–9
  67. Klebanov IuA. Analysis of fatal outcome in hemorrhagic fever with renal syndrome. Klin Med 1990; 68: 64-67
  68. Valtonen M, Kauppila M, Kotilainen P, Lähdevirta J, Svartbäck CM, Kosunen O, Nurminen J, Sarkkinen H, Brummer-Korvenkontio M. Four fatal cases of nephropathia epidemica. Scand J Infect Dis 1995; 27: 515-517
  69. Hautala T, Sironen T, Vapalahti O, Pääkkö ESärkioja TSalmela PIVaheri APlyusnin AKauma H. Hypophyseal hemorrhage and panhypopituitarism during Puumala Virus Infection: Magnetic Resonance Imaging and detection of viral antigen in the hypophysis. Clin Infect Dis 2002; 35: 96-101
  70. Lim TH, Chang KH, Han MC, Chang YB, Lim SM, Yu YS, Chun YH, Lee JS. Pituitary atrophy in Korean (epidemic) hemorrhagic fever: CT correlation with pituitary function and visual field. Am J Neuroradiol 1986; 7: 633-637
  71. Forslund T, Saltevo J, Anttinen J, Auvinen S, Brummer-Korvenkontio M, Korhonen A, Poutiainen M. Complications of nephropathia epidemica: three cases. J Intern Med 1992; 232: 87-90
  72. Settergren B, Boman J, Linderholm M, Wiström J, Hägg E, Arvidsson PA. A case of nephropathia epidemica associated with panhypopituitarism and nephrotic syndrome. Nephron 1992; 61: 234-235
  73. Suh DC, Park JS, Park SK, Lee HK, Chang KH. Pituitary hemorrhage as a complication of hantaviral disease. Am J Neuroradiol 1995; 16: 175-178
  74. Park JE, Pyo HJ. Delayed onset of diuresis in a patient with acute renal failure due to hemorrhagic fever with renal syndrome who also developed anterior hypopituitarism. Clin Nephrol 1996; 46: 141-145
  75. Kim NH, Cho JG, Ahn YK, Lee SU, Kim KH, Cho JH, Kim HG, Kim W, Jeong MH, Park JC, Kang JC. A case of torsade de pointes associated with hypopituitarism due to hemorrhagic fever with renal syndrome. J Korean Med Sci 2001; 16: 355-359
  76. Pekic S, Cvijovic G, Stojanovic M, Kendereski A, Micic D, Popovic V. Hypopituitarism as a late complication of hemorrhagic fever. Endocrine 2005; 26: 79-82
  77. Ahn HJChung JHKim DMYoon NRKim CM. Hemorrhagic fever with renal syndrome accompanied by panhypopituitarism and central diabetes insipidus: a case report. J Neurovirol  2018; 24: 382-387
  78. Bhoelan S, Langerak T, Noack D, van Schinkel L, van Nood E, van Gorp ECM, Rockx B, Goeijenbier M. Hypopituitarism after Orthohantavirus infection: what is currently known? Viruses 2019; 11: 340
  79. Shi S, Zhang A, Zhang J, Xu S. Partial hypopituitarism with ACTH deficiency as the main manifestation as a complication of hemorrhagic fever with renal syndrome. BMC Endocr Disord 2024; 24: 61.
  80. Partanen T, Koivikko M, Leisti P, Salmela P, Pääkkö E, Karttunen A, Sintonen H, Risteli L, Hautala N, Vapalahti O, Vaheri A, Kauma H, Hautala T. Long-term hormonal follow-up after human Puumala hantavirus infection. Clin Endocrinol2016; 84: 85-91
  81. Tarvainen MMäkelä SMustonen JJaatinen P. Autoimmune polyendocrinopathy and hypophysitis after Puumalahantavirus infection. Endocrinol Diabetes Metab Case Rep 2016; 2016
  82. Stojanovic M, Pekic S, Cvijovic G, Miljic D, Doknic M, Nikolic-Djurovic M, Micic D, Hrvacevic R, Nesic V, Popovic V.High risk of hypopituitarism in patients who recovered from hemorrhagic fever with renal syndrome. J Clin Endocrinol Metab 2008; 93: 2722-2728
  83. Bellastella G, Cirillo P, Carbone C, Scappaticcio L, Maio A, Botta G, Tomasuolo M, Longo M, Pontillo A, Bellastella A, Esposito K, De Bellis A. Neuroimmunoendocrinology of SARS-CoV 2 Infection. Biomedicines 2022; 10: 2855. 
  84. Clarke SA, Abbara A, Dhillo WS. Impact of COVID-19 on the endocrine system: a mini-review. Endocrinology 2022; 163: bqab203. 
  85. Capatina C, Poiana C, Fleseriu M. Pituitary and SARS CoV-2: An unremitting conundrum. Best Pract Res Clin Endocrinol Metab 2023; 37: 101752
  86. Chigr F, Merzouki M, Najimi M Autonomic brain centers and pathophysiology of COVID-19. ACS Chem Neurosci 2020; 11: 1520–1522
  87. Conde Cardona G, Quintana Pajaro LD, Quintero Marzola ID, Ramos Villegas Y, Moscote Salazar LR. Neurotropism of SARS-CoV 2: mechanisms and manifestations. J Neurol Sci 2020; 412: 116824
  88. Han T, Kang J, Li G, Ge J, Gu J. Analysis of 2019-nCoV receptor ACE2 expression in different tissues and its significance study. Ann Transl Med 2020; 8: 1077
  89. Abdel-Mannan O, Eyre M, Lobel U, Bamford A, Eltze C, Hameed B, Hemingway C, Hacohen Y. Neurologic and radiographic findings associated with COVID-19 infection in children. JAMA Neurol 2020; 77: 1440-1445
  90. Ding Y, He L, Zhang Q, Huang Z, Che X, Hou J, Wang H, Shen H, Qiu L, Li Z, Geng J, Cai J, Han H, Li X, Kang W, Weng D, Liang P, Jiang S. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: implications for pathogenesis and virus transmission pathways. J Pathol 2004; 203: 622–630
  91. Frara S, Allora A, Castellino L, di Filippo L, Loli P, Giustina A. COVID-19 and the pituitary. Pituitary, 2021; 24: 465-481
  92. Puig-Domingo M, Marazuela M, Yildiz BO, Giustina A. COVID-19 and endocrine and metabolic diseases. An updated statement from the European Society of Endocrinology. Endocrine 2021; 72: 301-316.
  93. Ghosh R, Roy D, Roy D, Mandal A, Dutta A, Naga D, Benito-Leon J. A rare case of SARS-CoV-2 infection associated with pituitary apoplexy without comorbidities. J Endocr Soc 2021; 5: bvaa203.
  94. Martinez-Perez R, Kortz MW, Carroll BW, Duran D, Neill JS, Luzardo GD, Zachariah MA. Coronavirus disease 2019 and pituitary apoplexy: a single-center case series and review of the literature. World Neurosurg 2021; 152: e678-e687.
  95. Facondo P, Maltese V, Delbarba A, Pirola I, Rotondi M, Ferlin A, Cappelli C. Case report: hypothalamic amenorrhea following COVID-19 infection and review of literatures. Front Endocrinol (Lausanne) 2022; 13: 840749.
  96. Hazzi C, Villemure-Poliquin N, Nadeau S, Champagne PO. SARS-CoV-2 infection, a risk factor for pituitary apoplexy? A case series and literature review. Ear Nose Throat J 2024; 103: 153S-161S.  
  97. Racca G, D'Agnano S, Fasano N, Gianotti L. Pituitary apoplexy with transition to acute hypophysitis in a patient with Sars-CoV-2 pneumonia. JCEM Case Rep 2022; 1: luac010.
  98. Bray DP, Solares CA, Oyesiku NM. Rare case of a disappearing pituitary adenoma during the COVID-19 pandemic. World Neurosurg 2020; 146: 148-149
  99. Chan JL, Gregory KD, Smithson SS, Naqvi M, Mamelak AN. Pituitary apoplexy associated with acute COVID-19 infection and pregnancy. Pituitary 2020; 23: 716-720
  100. Santos CDSE, Filho LMDCL, Santos CAT, Neill JS, Vale HF, Kurnutala LN. Pituitary tumor resection in a patient with SARSCoV-2 (COVID-19) infection. A case report and suggested airway management guidelines. Braz J Anesthesiol 2020; 70: 165-170
  101. Solorio-Pineda S, Almendarez-Sanchez CA, Tafur-Grandett AA, Ramos-Martinez GA, Huato-Reyes R, Ruiz-Flores MI, Sosa-Najera A. Pituitary macroadenoma apoplexy in a severe acute respiratory syndrome-coronavirus-2-positive testing: causal or casual? Surg Neurol Int 2020; 11: 304
  102. LaRoy M & McGuire M. Pituitary apoplexy in the setting of COVID-19 infection: a case report. Am J Emerg Med 2021; S0735-6757(21)00153-4
  103. Bordes SJ, Phang-Lyn S, Najera E, Borghei-Razavi H, Adada B. Pituitary apoplexy attributed to COVID-19 infection in the absence of an underlying macroadenoma or other identifiable cause. Cureus 2021; 13: e13315
  104. Joshi M, Gunawardena S, Goenka A, Ey E, Kumar G. Post COVID-19 Lymphocytic hypophysitis: a rare presentation. Child Neurol Open 2022; 9: 2329048X221103051
  105. Zerrouki D, Assarrar I, Rami I, Rouf S, Latrech H. Coronavirus as a trigger of lymphocytic hypophysitis in an adolescent girl: An exceptional case report. Int J Surg Case Rep 2024; 115: 109218.
  106. Gonen MS, De Bellis A, Durcan E, Bellastella G, Cirillo P, Scappaticcio L, Longo M, Bircan BE, Sahin S, Sulu C, Ozkaya HM, Konukoglu D, Kartufan FF, Kelestimur F. Assessment of neuroendocrine changes and hypothalamo-pituitary autoimmunity in patients with COVID-19. Horm Metab Res 2022; 54: 153-161. 
  107. Littera R, Campagna M, Deidda S, Angioni G, Cipri S, Melis M, Firinu D, Santus S, Lai A, Porcella R, Lai S, Rassu S, Scioscia R, Meloni F, Schirru D, Cordeddu W, Kowalik MA, Serra M, Ragatzu P, Carta MG, Del Giacco S, Restivo A, Deidda S, Orrù S, Palimodde A, Perra R, Orrù G, Conti M, Balestrieri C, Serra G, Onali S, Marongiu F, Perra A, Chessa L. Human leukocyte antigen complex and other immunogenetic and clinical factors influence susceptibility or protection to SARS-CoV-2 infection and severity of the disease course. The Sardinian experience. Front Immunol 2020; 11: 605688. 
  108. Weiner J, Suwalski P, Holtgrewe M, Rakitko A, Thibeault C, Müller M, Patriki D, Quedenau C, Krüger U, Ilinsky V, Popov I, Balnis J, Jaitovich A, Helbig ET, Lippert LJ, Stubbemann P, Real LM, Macías J, Pineda JA, Fernandez-Fuertes M, Wang X, Karadeniz Z, Saccomanno J, Doehn JM, Hübner RH, Hinzmann B, Salvo M, Blueher A, Siemann S, Jurisic S, Beer JH, Rutishauser J, Wiggli B, Schmid H, Danninger K, Binder R, Corman VM, Mühlemann B, Arjun Arkal R, Fragiadakis GK, Mick E, Comet C, Calfee CS, Erle DJ, Hendrickson CM, Kangelaris KN, Krummel MF, Woodruff PG, Langelier CR, Venkataramani U, García F, Zyla J, Drosten C, Alice B, Jones TC, Suttorp N, Witzenrath M, Hippenstiel S, Zemojtel T, Skurk C, Poller W, Borodina T, Pa-Covid SG, Ripke S, Sander LE, Beule D, Landmesser U, Guettouche T, Kurth F, Heidecker B. Increased risk of severe clinical course of COVID-19 in carriers of HLA-C*04:01. EClinicalMedicine 2021; 40: 101099.
  109. Hovhannisyan A, Madelian V, Avagyan S, Nazaretyan M, Hyussyan A, Sirunyan A, Arakelyan R, Manukyan Z, Yepiskoposyan L, Mayilyan KR, Jordan F. HLA-C*04:01 affects HLA class I heterozygosity and predicted affinity to SARS-CoV-2 peptides, and in combination with age and sex of armenian patients contributes to COVID-19 severity. Front Immunol 2022; 13: 769900.
  110. Yasuda S, Suzuki S, Yanagisawa S, Morita H, Haisa A, Satomura A, Nakajima R, Oikawa Y, Inoue I, Shimada A. HLA typing of patients who developed subacute thyroiditis and Graves' disease after SARS-CoV-2 vaccination: a case report. BMC Endocr Disord 2023; 23: 54.
  111. Rajevac H, Bachan M, Khan Z. Diabetes insipidus as a symptom of covid-19 infection: case report. Chest 2020, 158: A2576
  112. Sheikh AB, Javed N, Sheikh AA, Upadhyay S, Shekhar R. Diabetes insipidus and concomitant myocarditis: a late sequelae of COVID-19 infection. J Investig Med High Impact Case Rep 2021, 9: 1–4
  113. Misgar RA, Rasool A, Wani AI, Bashir MI. Central diabetes insipidus (infudibuloneuro hypophysitis): a late complication of COVID-19 infection. J Endocrinol Invest 2021 Jul 3; 1-2
  114. Yavari A, Sharifan Z, Larijani B, Mosadegh Khah A. Central diabetes insipidus secondary to COVID-19 infection: a case report. BMC Endocr Disord 2022; 22: 134.
  115. Berni A, Malandrino D, Corona G, Maggi M, Parenti G, Fibbi B, Poggesi L, Bartoloni A, Lavorini F, Fanelli A, Scocchera G, Nozzoli C, Peris A, Pieralli F, Pini R, Ungar A, Peri A. Serum sodium alterations in SARS CoV-2 (COVID-19) infection: impact on patient outcome. Eur J Endocrinol 2021; 185: 137-144. 
  116. Khidir RJY, Ibrahim BAY, Adam MHM, Hassan RME, Fedail ASS, Abdulhamid RO, Mohamed SOO. Prevalence and outcomes of hyponatremia among COVID-19 patients: A systematic review and meta-analysis. Int J Health Sci (Qassim) 2022; 16: 69-84.
  117. Morita S, Tsuji T, Kishimoto S, Uraki S, Takeshima K, Iwakura H, Furuta H, Nishi M, Inaba H, Matsuoka TA. Isolated ACTH deficiency following immunization with the BNT162b2 SARS-CoV-2 vaccine: a case report. BMC Endocr Disord 2022; 22:185.
  118. Panos LD, Bargiotas P, Hadjigeorgiou G, Panos GD. Neurovascular adverse effects of Sars-Cov-2 vaccination. Drug Des Devel Ther 2024; 18: 1891-1905.
  119. Verrienti M, Marino Picciola V, Ambrosio MR, Zatelli MC. Pituitary and COVID-19 vaccination: a systematic review. Pituitary 2024 May 18. doi: 10.1007/s11102-024-01402-2.
  120. Watanabe S, Tamura Y, Oba K, Kitayama S, Sato M, Kodera R, Toyoshima K, Chiba Y, Araki A. Hypopituitarism with secondary adrenocortical insufficiency and arginine vasopressin deficiency due to hypophysitis after COVID-19 vaccination: a case report. BMC Endocr Disord 2024; 24(1): 71.
  121. Bouça B, Roldão M, Bogalho P, Cerqueira L, Silva-Nunes J. Central diabetes insipidus following immunization with BNT162b2 mRNA COVID-19 vaccine: a case report. Front Endocrinol (Lausanne) 2022; 13: 889074.
  122. Torres AM, Kazee AM, Simon H, Knudson PE, Weinstock RS. Central diabetes insipidus due to herpes simplex in a patient immunosuppressed by Cushing's syndrome. Endocr Pract 2000; 6: 26-28
  123. Moses AM, Thomas DG, Canfield MC, Collins GH. Central diabetes insipidus due to cytomegalovirus infection of the hypothalamus in a patient with acquired immunodeficiency syndrome: a clinical, pathological, and immunohistochemical case study. J Clin Endocrinol Metab 2003; 88: 51-54.
  124. Scheinpflug K, Schalk E, Reschke K, Franke A, Mogren M. Diabetes insipidus due to herpes encephalitis in a patient with diffuse large cell lymphoma. A case report. Exp Clin Endocrinol Diabetes 2006; 114: 31-34
  125. Jones GMuriello MPatel ALogan L. Enteroviral meningoencephalitis complicated by central diabetes insipidusin a neonate: a case report and review of the literature. J Pediatric Infect Dis Soc 2015; 4: 155-158.
  126. Tsuji HYoshifuji HFujii T, Matsuo TNakashima RImura YYukawa NOhmura KSumiyoshi SMimori T. Visceral disseminated varicella zoster virus infection after rituximab treatment for granulomatosis with polyangiitis. Mod Rheumatol  2017; 27: 155-161
  127. Zhang X, Li Q, Hu P, Cheng H, Huang G. Two case reports of pituitary adenoma associated with Toxoplasma gondii infection. J Clin Pathol 2002; 55: 965-966
  128. Milligan SA, Katz MS, Craven PC, Strandberg DARussell IJBecker RA. Toxoplasmosis presenting as panhypopituitarism in a patient with the acquired immune deficiency syndrome. Am J Med 1984; 77: 760-764
  129. Hamdeh S, Abbas A, Fraker J, Lambrecht JE. Intracranial toxoplasmosis presenting as panhypopituitarism in an immunocompromised patient. Am J Emerg Med2015; 33: 1848.e1-2
  130. Del Brutto OH, Del Brutto VJ. Intrasellar cysticercosis: a systematic review. Acta Neurol Belg 2013; 113: 225-227.
  131. Dutta D, Kumar M, Ghosh S, Mukhopadhyay S, Chowdhury S. Pituitary hormone deficiency due to racemose neurocysticercosis. Lancet Diabetes Endocrinol 2013; 1: e13.
  132. Cheong JH, Kim JM, Kim CH. Neurocysticercosis involving the pituitary stalk: case report and literature review. J Korean Neurosurg Soc 2010; 48: 91-93

Development And Microscopic Anatomy Of The Pituitary Gland

ABSTRACT

 

The pituitary gland is an organ of dual origin. The anterior part (adenohypophysis) arises from embryonic buccal mucosa, whereas the posterior part (neurohypophysis) derives from neural ectoderm. Precise spatial and temporal co-ordination of transcription factor expression in both structures is critical for pituitary gland formation and the differentiation of hormone-producing cells. Disruption of this regulation, for instance by transcription factor mutation, can lead to numerous developmental disorders and disturbances in endocrine function and regulation. We provide an overview of the molecular drivers of pituitary organogenesis and illustrate the anatomy and histology of the mature pituitary gland, comprising adenohypophysis (anterior lobe), neurohypophysis (posterior lobe), pars intermedia (intermediate lobe), and infundibulum (pituitary stalk).

 

PITUITARY ORGANOGENESIS

 

The pituitary is an organ of dual origin. The anterior part (adenohypophysis) is derived from oral ectoderm and is epithelial in origin, whereas the posterior part (neurohypophysis) derives from neural ectoderm. The mixed embryological derivation of the pituitary gland requires that developmental signals from both the neural and oral ectoderm must interact temporally and spatially to control maturation of the pituitary gland and the cellular differentiation of the various hormone-producing cell types within the adenohypophysis. The expression of transcription factors that control cell lineage commitment in the developing adenohypophysis must be regulated precisely 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 tumors (1) (2).

 

Pituitary organogenesis begins during week 4 of fetal development. A thickening of cells in the oral ectoderm forms the hypophyseal placode which gives rise to Rathke’s pouch; an upward evagination of the buccal ectoderm 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 bilobed structure of the adult pituitary gland.  Rathke’s pouch constricts at its base and eventually separates altogether from the oral epithelium during gestational week 6-8. The cells of the anterior wall of Rathke’s pouch undergo extensive proliferation to form the anterior lobe. In humans, the posterior wall proliferates more slowly to form the vestigial intermediate lobe (3). Cell patterning and terminal differentiation occurs within the anterior lobe to form the five principal specialized endocrine cell types of the pituitary gland; the somatotrophs, adrenocorticotrophs, gonadotrophs, thyrotrophs, and lactotrophs.

 

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 (Blue in Table 1).
  2. Evagination of Rathke’s pouch and cell proliferation (Green in Table 1).
  3. Lineage determination and cellular differentiation (Yellow in Table 1).

 

Table 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 homeo-domain 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

(52) (53)

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

(54)

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

 

(55) (56)

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

 (57)

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 (6)

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

(58) (59)

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

(60) (61)(59)

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

(62) (6)

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

(63) (7)

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

(64)

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 (5)

BMP2

Induces Isl1 expression

 

Downregulation required for cell differentiation

Prolonged expression results in hyperplastic pituitary and lack of terminal differentiation

 

(65)

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

 

(66) (67)(68)

Shh

 

Expressed in oral ectoderm and ventral diencephalon

 

Induces Lhx3 expression

Antagonism in mouse oral ectoderm results in hypoplastic Rathke’s pouch

 

(69)

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 (6) and (70) (71)(72) (73)(74)

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

(73) (75)(76) (77)

GATA2

 

Specifies gonadotroph and thyrotroph lineages

 

Induces expression of Nr5a1

In mice, overexpression associated with gonadotroph and thyrotroph hypoplasia.

Expression persists in adult gland

(75)

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

 

(18) (78)

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

(79) (80)

 

Much of our understanding of the process of pituitary organogenesis comes from mouse studies (4) (5), but the phenotypes associated with human disorders often share aspects with mouse models of defective pituitary development. Various transcription factors that are involved are presented in Figure 1 and summarized in Table 1 (6), (7), (8) & (9).

 

Figure 1. Signaling molecules and transcription factors control pituitary development. Arrows represent regulation of expression in the direction indicated (See Table 1). The clinically used transcription factors are in blue.

 

Transcriptional Control of Pituitary Organogenesis: Lineage Determination and Cellular Differentiation

 

Significant advances have been made in recent years in elucidating the molecular mechanisms underlying the commitment and differentiation of Rathke’s pouch progenitors and the role of endocrine stem cells in the adenohypophysis (8) (10).  Many factors are involved in pituitary differentiation, but a few later acting transcription factors have pituitary-specific effects and have become clinically relevant (11).  In clinical terms, cells of the adenohypophysis can be divided into those of the Pit-1 lineage (somatotrophs, lactotrophs, somatomammotrophs, and thyrotrophs), those of t-PIT lineage (corticotrophs and melanotrophs), and those of SF-1 lineage (Gonadotrophs).

 

PIT-1 (POU1F1)

 

PIT-1 was first identified as a trans-activating factor of the growth hormone and prolactin genes. It is a member of the POU homeodomain family of transcription factors (5). Inactivating mutations produce recessive hypopituitarism characterized by a congenital lack of growth hormone, prolactin and TSH (12).  Two naturally occurring recessive mouse mutants have Pit-1 defects; the Snell Dwarf (dw) mouse and the Jackson dwarf (dwJ).  Homozygous recessive variants of these mice both exhibit post-natal, but not embryonic, anterior pituitary hypoplasia with GH, PRL and TSH deficiencies. 

 

T-PIT

 

t-PIT (TBX19) is a member of the T-Box family of transcription factors.  The T-Box represents the DNA binding domain of these factors.  T-Pit is expressed in the developing pituitary and expression generally persists into the adult gland.  T-Pit depletion in mice induces severe ACTH and glucocorticoid deficiencies in addition to adrenal hypoplasia and pigmentation defects.  The phenotype of t-Pit -/- mice suggests that T-PIT promotes the development of corticotrophs but can also actively repress gonadotroph formation (13). 

 

STEROIDOGENIC FACTOR (SF-1)

 

SF-1 is a nuclear receptor encoded by the gene Nr5a1 or steroidogenic factor 1 (SF-1).    It is an orphan nuclear receptor involved as a transactivating factor in steroid biosynthesis.  SF-1 is expressed throughout the adrenal and reproductive axes during development and postnatal life (14) (15).  Mutations of SF-1 in humans are associated with 46XY sex reversal with adrenal failure, 46XY gonadal dysgenesis and 46XX ovarian insufficiency and premature ovarian failure (16) (17).

 

In some cases, transcription factor deficiency does not result in the complete absence of a cell type.  SF-1 deficient mice do not spontaneously produce gonadotrophins but hyperstimulation with GnRH can induce hormone production suggesting that SF-1 is not essential for gonadotroph differentiation (18).  Similarly, t-Pit expression is not essential for corticotroph development, but POMC synthesis is delayed if they are deficient (19). The failure to promote differentiation along a particular lineage can be permissive for other pathways and the idea that single transcription factors direct cell differentiation is overly simplistic.  Nevertheless, these differentiation markers (colored blue in Figure 1) have proven to be extremely useful in diagnostic pituitary pathology, particularly in demonstrating the cell lineage of silent or non-expressing pituitary adenomas, and their expression can be detected in histological samples of pituitary tissue removed at trans-sphenoidal adenohypophysectomy by immunohistochemistry (Figure 2).

 

Figure 2. Pituitary transcription factor expression in the normal pituitary gland.

 

ANATOMY AND HISTOLOGY OF THE MATURE PITUITARY GLAND

 

Hypophysis Cerebri

 

The mature pituitary gland, or hypophysis cerebri, is a small, oval body weighing approximately 0.5g. The adenohypophysis of the pituitary is generally smaller in men than women and, within the female population, the gland in nulliparous women is generally smaller than that seen in multiparous women (20) (21).  Indeed, during pregnancy, the gland may increase by approximately 30% due to a combination of lactotroph hyperplasia and vascular engorgement. 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 (a thin film of dura), laterally by the walls of the cavernous sinus, and antero-inferiorly by the posterior wall of the sphenoid sinus; the latter used as the standard route for pituitary surgery or transsphenoidal adenectomy (Figure 3).  Antero-superiorly, the pituitary lies in close proximity to the optic chiasm and bilateral, inferomedial compression of the chiasm by the expanding gland explains why space-occupying lesions of the pituitary commonly present with bitemporal hemianopia. Inferiorly, the adenohypophysis is separated from the floor of the sella turcica by a large, loculated venous sinus that communicates with the circular sinus. 

 

Figure 3. High resolution 3D imaging reconstruction of the sella turcica in a volunteer (courtesy of Prof Mark Gurnell, Cambridge, UK). These detailed scans are proving invaluable in planning the surgical approach for transsphenoidal adenectomy, especially where vascular anomalies are expected.
A) Section across the roof of the sella with the optic nerves and chiasm (green) in situ revealing the intimate relationship between the chiasm and the pituitary gland (turquoise).
B) A similar level with the optic nerve and chiasm removed.
C) Detail of the sella region showing the relationship between the pituitary gland and stalk (turquoise), base of hypothalamus (orange), and internal carotid arteries (red).

 

The Hypothalamus

 

The structure of the hypothalamus and its complex connections are well known and have been described in detail elsewhere (22) (23) (24). The hypothalamic nuclei that give rise to the neurohypophysis arise from the preoptic, supraoptic, lateral, tuberal and mamillary regions (3) and hypothalamic regulatory neurons have their cell bodies in the supraoptic, paraventricular, infundibular and ventromedial nuclei of the hypothalamus.  These hypothalamic nuclei have projections that terminate throughout the median eminence, infundibulum, and the posterior lobe (Figure 4).  Neurons derived from hypothalamic nuclei produce peptidergic “releasing hormones” that are transported along axons and released into the portal system from where they are carried to the dense capillary plexus of the adenohypophysis to influence hormone synthesis and release.  The parvocellular neurosecretory pathway of the tuberoinfundibular tract is proposed as the main route for the synthesis of releasing hormones and of transmission to portal blood.   The major trophic factors originating in the hypothalamic nuclei include growth hormone releasing factor (GRF), corticotrophin releasing factor (CRF), thyrotropin releasing factor (TRF), gonadotrophin releasing hormone (GnRH) and other peptides such as somatostatin (SS).  Other trophic and inhibitory factors are also released in this region including dopaminergic compounds. 

 

Figure 4. Sagittal T1-weighted image of the pituitary fossa demonstrates normal pituitary anatomy. The T1-weighted study is particularly helpful in identifying the posterior pituitary bright spot (white arrow)- a focus of T1 signal hyperintensity posteriorly in the sella, which corresponds with the neurohypophysis. The intrinsic T1 signal shortening through the posterior pituitary is thought to be due to the presence of vasopressin. The zoomed image of the pituitary gland, consisting of the anterior (A) and posterior (P) lobes, sits within the sella turcica, a “saddle”-shaped depression in the sphenoid bone, with the tuberculum sellae anteriorly, and the dorsum sellae posteriorly. Superior to the pituitary gland is CSF within the hypophyseal and suprasellar cisterns. The pituitary stalk (S), also known as the infundibulum or infundibular stalk, extends through the suprasellar cistern, extending between the hypothalamus and the superior surface of the pituitary gland. The optic chiasm (C) can be seen in the suprasellar space more superiorly. Anteroinferior to the pituitary gland is the sphenoid sinus (SS). (B)- brainstem, (M)- mammillary bodies, (3V)- third ventricle.

 

In addition to nerve fibers, the long and short portal blood vessels (distributed between sinusoids in the median eminence, infundibulum, and the adenohypophysis) are also of major importance to this region. The adenohypophysis receives no major nerve supply and almost all of its blood comes from hypothalamic-hypophysial portal vessels. The predominant direction of blood flow is from the tufts of sinusoids in the median eminence and infundibulum via the portal vessels to sinusoids between glandular cells in the anterior lobe.  This neurovascular arrangement is of vital importance to understand the mechanism by which the hypothalamus regulates the neuroendocrine function of the gland. 

 

Neurosecretion is the physiological process involved in the production and release of posterior lobe oxytocin and vasopressin and their related proteins: including the precursor neurophysins.  In terms of these magnocellular neurosecretory pathways, both the supraoptic and paraventricular nuclei are involved in the production both of vasopressin and oxytocin, however, vasopressin originates predominantly from the supraoptic nucleus and oxytocin originates predominantly from the paraventricular nucleus. 

 

Infundibulum (Pituitary Stalk)

 

The infundibular 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 (see below) and contains a few scattered gonadotroph or corticotroph cells. Anteriorly and superficially, it surrounds the pars infundibularis (infundibular stem), which contains the unmyelinated axons of the magnocellular supraoptic and paraventricular neurons.  The neurons contain large intra-axonal accumulations of oxytocin and vasopressin that may be seen as eosinophilic ovoid granular swellings along the trajectory of these axons in the infundibular stem. These ovoid swellings are known as “Herring bodies” named after Percy Theodore Herring who first described them (figure 4).  It also contains the neural connections of the hypophysis that are continuous with the median eminence of the tuber cinereum. 

 

The Adenohypophysis

 

The adenohypophysis comprises the vast majority of the pituitary gland by volume and is composed of three parts: the pars distalis, the pars intermedia and the pars tuberalis.  The pars distalis forms the majority of the adenohypophysis, the pars intermedia is rudimentary in the human but represents the vestigial posterior limb of Rathke’s pouch and the pars tuberalis is an upward extension of the adenohypophysis that surrounds the lower regions of the hypophysial stalk.  The anatomy of this region is shown in figure 5.

 

Figure 5. Coronal T2-weighted image demonstrates normal sellar and parasellar anatomy and delineates the local anatomical boundaries of the pituitary fossa. The pituitary gland (P) is sited within the sella turcica, with the sphenoid sinus (SS) demonstrated inferiorly. The pituitary stalk (S) is seen extending vertically through the suprasellar space in the midline. Superior to this is the optic chiasm (C). The lateral borders of the pituitary fossa are formed by the cavernous sinuses. On this coronal image, the T2 flow voids through the cavernous segments of the ICAs occupy the cavernous sinuses.

 

Pars Distalis

 

The pars distalis of the adenohypophysis is a highly vascular structure, consisting of epithelial cells of varying size and shape arranged in cords, irregular masses or follicles, separated by thin-walled vascular sinusoids and supported by a complex network of reticular tissue best appreciated in reticulin preparations (Figure 6).  This region of the adenohypophysis contains several cell types, generally divided into acidophils and basophils (depending on their staining properties with mixed acidic and basic dyes such as Orange-G/aldehyde fuchsin, (figure 7).  These cells release trophic hormones including growth hormone (GH), prolactin (PRL), adrenocorticotrophic hormone (ACTH), thyroid stimulating hormone (TSH), follicle stimulating hormone (FSH) and luteinizing hormone (LH).  The regulation and production of these trophic hormones is complex and is thought to be the result of a combination of the release of hypothalamic releasing factors into the median eminence, the regulation of portal blood flow by vascular systems, feedback by systemic hormones, autocrine interactions and complex paracrine interactions within the gland (25) (26)(27). 

 

Figure 6. Section of adenohypophysis stained with reticulin showing nested acini of adenohypophysial cells.

Figure 7. Section of adenohypophysis stained with PAS/Orange G showing the distribution of acidophils (Orange) and basophils (Purple).

 

The cellular heterogeneity within acini may be demonstrated tinctorially in PAS-OG preparations (Figures 7, 8) which were widely used before the adoption of antibody-based stains (Figure 9). Corticotroph cells are generally strongly basophilic (PAS-positive), somatotroph and lactotroph cells mostly acidophilic (orangeophilic), whilst gonadotrophs and thyrotrophs may be basophilic or chromophobic (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. In a normal adult adenohypophysis, approximately 10% of endocrine cells are basophils, 40% acidophils and 50% chromophobes.

 

Figure 8. The axial section of the adenohypophysis of the human pituitary gland 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).

 

The PAS-OG stain is still a useful supplementary method in the differential diagnosis of some pituitary lesions (hyperplasia, corticotroph microadenoma, Crooke’s cell changes or adenoma) but these tinctorial stains have all largely been replaced by immunohistochemical methods (Figure 9).   Although most acini contain a mixture of different hormone-producing cells, there is evidence of zonation (Figure 8). 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) harbors clusters of thyrotrophs. Gonadotroph (LH/FSH) cells are diffusely scattered throughout the gland.

  

 

Figure 9. Examples of immunohistochemistry. It can be seen that although growth hormone (GH) is generally enriched in the wings, the distribution is more diffuse. Corticotrophs (ACTH) are more obviously restricted to the central “mucoid wedge”. Compare this with tinctorial stains in Figures 7 & 8.

 

Cell Types of the Adenohypophysis

 

SOMATOTROPHS

 

Somatotrophs represent about 50% of cells in the adenohypophysis and are located predominantly, but not exclusively, in the lateral wings.  They are readily identified by the expression of growth hormone.  A subset of somatotrophs also express the common glycoprotein hormone alpha-subunit.  They are the second cell type to form during fetal development and are detected by 8 weeks of gestation. 

 

LACTOTROPHS

 

Lactotrophs express prolactin.  The number of prolactin-secreting cells varies widely between the sexes and with parity.  The Golgi apparatus is well developed in lactotrophs and contains immature, pleomorphic secretory granules.  Granule extrusions are common in lactotrophs, not only at the basal cell surface, but also on the lateral cell borders; the latter is a distinctive feature of lactotrophs seen on electron microscopy and is known as “misplaced exocytosis”. Lactotrophs appear to arise from mammosomatotroph cells and pure lactotrophs appear late in gestation (24 weeks).  The expansion and secretion of lactotrophs is controlled by exogenous hormones including estrogen, paracrine effects from adjacent adenohypophysial cells and inhibitory substances from the hypothalamus. 

 

MAMMOSOMATOTROPHS

 

These bihormonal cells contain both growth hormone and prolactin.  Mammosomatotrophs are the precursors of lactotrophs and, as in lactotrophs, misplaced exocytosis can be seen on electron microscopy (figure 10). These cells are thought to be the source of lactotroph increases during pregnancy (28)

 

Figure 10. Electron micrograph of a mammosomatotroph adenoma displaying misplaced exocytosis. This feature is seen as the extrusion of secretory granules into the extracellular spaces or from intercellular extensions of the basement membrane.

 

CORTICOTROPHS

 

Corticotrophs synthesis the molecule pro-opiomelanocrortin (POMC) which is cleaved post-transcriptionally to produce bioactive peptides including adrenocorticotrophic hormone (ACTH), melanotrophin (MSH) lipotrophic hormone (LPH) and endorphins (29).  These cells comprise about 15-20% of the cell component of the adenohypophysis and the cells are concentrated in the central mucoid wedge.  They are best identified histologically by using antibodies raised against ACTH.  Corticotrophs are the first cell type to differentiate in the fetal pituitary gland at 6 weeks of gestation and ACTH can be detected at 7 weeks. Exposure to glucocorticoid access, either by exogenous corticosteroid administration or any cause of endogenous glucocorticoid hypersecretion, including ectopic secretion of ACTH, causes corticotrophs to undergo a distinctive, but reversible, morphological alteration known as Crooke hyaline change (30).

 

THYROTROPHS

 

These are the most infrequent cell type within the adenohypophysis and are detected by the expression of the glycoprotein hormone common alpha-subunit and the beta-subunit of TSH.  This can be demonstrated by immunohistochemical double labelling for both subunits.  Thyrotrophs are detectable at about 12 weeks of gestation.

 

GONADOTROPHS

 

Gonadotrophs produce both luteinizing hormone (LH) and follicle stimulating hormone (FSH) and both hormone specific beta-subunits and the common alpha-subunit can be detected in these cells.  Gonadotrophs are scattered throughout the pars distalis and are the major constituent of the pars tuberalis.  Gonadotrophs are intimately associated with lactotrophs and electron microscopy reveals gap junctions between them.  The synthesis and release of the gonadotrophins is differentially regulated by the hormonal milieu, paracrine interactions and the pulsatile frequency of GnRH (31) (26,32). 

 

There is growing evidence that the precise control of pituitary gland secretion also involves a contribution from cells with the capacity to demonstrate plasticity with self-renewal and it is likely that the pituitary contains a pool of stem cells (33)(34) (35) (36).  It is possible that these stem cells, possibly expressing Nestin, Lhx3 and Sox2, may exhibit self-renewal properties and have the ability to differentiate into different hormone-producing cell types in response to differing physiological demands (36) (37) (38).

 

FOLLICULOSTELLATE CELLS  

 

Folliculostellate (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 (39), but whether this is true in humans is not certain. FS cells are small, chromophobe, with slender processes that extend between the endocrine cells. They form small follicles at the center of acini, comprised of apical tips of multiple FS cells. They may be visualized with antibodies raised against the S100 protein and GFAP, 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 (Figure 11).  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 (40) or act as antigen-presenting cells.  FS cells are also postulated to play a role in paracrine regulation (26,41).

 

Figure 11. 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 Tuberalis

 

The pars tuberalis is characterized by a large number of traversing vessels.  Between these are cords or balls of undifferentiated cells admixed with acidophilic and basophilic cells.  The secretion of adenohypophyseal hormones into the circulation appears to occur by exocytosis of the vesicular contents into the perivascular spaces of the sinusoids, the latter being lined with a fenestrated endothelium which facilitates diffusion into the bloodstream. The signal for secretion is the liberation of chemical releasing factors from neurons in the median eminence, and other hypothalamic centers, into the portal system of veins by which they are carried and distributed within the adenohypophysis. 

 

Pars Intermedia

 

In contrast to rodents, the pars intermedia is rudimentary in adult humans (Figure 12). 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 age, 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 hypercortisolemia.  Secretory cells of the pars intermedia have granules containing either alpha- or beta-endorphin; these cells have been shown to contain various peptide hormones including ACTH and alpha-MSH (melanocyte stimulating hormone). 

 

Figure 12. The 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 (center) 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 13).

Figure 13. ‘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

 

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 (Figure 14). Some of these axons are short and terminate in the median eminence and infundibular stem among the superior capillary beds of the venous portal circulation.  Longer axons pass into the main neurohypophysis thereby forming the neurosecretory hypothalamo-hypophyseal tract with their terminals ending near the sinusoids of the posterior lobe. These 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 characterized by the expression of the TTF-1 transcription factor (absent in classic GFAP-positive astrocytes) (42) (43) (44). These cells show elongated processes often running in parallel with axons and demonstrate only patchy GFAP and S100 expression.

 

Figure 14. The cytoarchitecture of the neurophypophysis (right) is strikingly different from the adenohypophysis, which contains the nested (inset, left) collections of neuroendocrine cells (left). The neurohypophysis does not contain neurosecretory cell bodies; instead, it is composed of specialized 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 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.

 

The Vessels of the Hypophysis & Hypothalamic-Hypophysial Portal System

 

The vessels of the hypophysis cerebri have been extensively described and reviewed (45) (46) (47) (48).  The arteries of the hypophysis arise from the internal carotids via a single inferior and multiple superior hypophysial arteries. The arteries of the median eminence and infundibulum end in sprays of capillaries. In the median eminence, these form the external (mantle) plexus and the internal (deep) plexus that are both drained by the long portal vessels some of which terminate in the adenohypophysis.  Short portal vessels run from the lower infundibulum to the pars anterior. Both types of portal vessel open into the vascular sinusoids lying between the secretory cords in the adenohypophysis, providing most of the blood; there is no direct arterial supply to this region (49) (50). The portal system is considered to carry hormone-secreting factors from the hypothalamus (46) (45) (49).  Another notable feature of the rostral portion of the stalk are tortuous capillary loops surrounding a central capillary, termed gomitoli (51). These structures are composed of a central muscular artery surrounded by a spiral of capillaries.  Although their function is unknown, the complexity of these gomitoli suggests that they may regulate the rate of blood flow to the adenohypophysis thereby regulating the flow of trophic factors.

 

The venous drainage of the neurohypophysis is by three possible routes: to the adenohypophysis via the long and short portal vessels, via the large inferior hypophyseal veins into the dural venous sinuses or to the hypothalamus via capillaries passing to the median eminence.  The venous drainage carries hypophysial hormones from the gland to their target tissues and also facilitates feedback of secretions.  In contrast, the venous drainage of the adenohypophysis appears restricted.  It is possible that flow in the short postal vessels could be reversed explaining the postulated “short feedback loop”.  These models of hypophysial blood flow are of importance to the mechanism of hormone secretion.  It is not certain whether the median eminence represents the final common pathway for neural control of the adenohypophysis or the entire neurohypophysis may be involved; its capillary bed may selectively determine the destination of both hypothalamic and pituitary secretions, conveying some to the glandular pituitary, others to distant target organs and yet others to the brain (48). 

 

REFERENCES

 

  1. Davis SW, Potok MA, Brinkmeier ML, Carninci P, Lyons RH, MacDonald JW, Fleming MT, Mortensen AH, Egashira N, Ghosh D, Steel KP, Osamura RY, Hayashizaki Y, Camper SA. Genetics, gene expression and bioinformatics of the pituitary gland. Horm Res 2009;71 Suppl 2:101–15.
  2. Srirangam Nadhamuni V, Korbonits M. Novel Insights into Pituitary Tumorigenesis: Genetic and Epigenetic Mechanisms. Endocr Rev 2020 12 01;41(6):bnaa006.
  3. Asa SL Kovacs. Functional morphology of the human fetal pituitary. Pathol. Ann. 1984;19(Pt.1):275–315.
  4. Rizzoti K. Genetic regulation of murine pituitary development. J Mol Endocrinol 2015;54(2):R55-73.
  5. 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(7):790–829.
  6. 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(9):2442–55.
  7. 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. Dev Biol 2008;324(2):322–33.
  8. Davis SW, Castinetti F, Carvalho LR, Ellsworth BS, Potok MA, Lyons RH, Brinkmeier ML, Raetzman LT, Carninci P, Mortensen AH, Hayashizaki Y, Arnhold IJ, Mendonça BB, Brue T, Camper SA. Molecular mechanisms of pituitary organogenesis: In search of novel regulatory genes. Mol Cell Endocrinol 2010;323(1):4–19.
  9. Zhu X, Gleiberman AS, Rosenfeld MG. Molecular physiology of pituitary development: signaling and transcriptional networks. Physiol Rev 2007;87(3):933–63.
  10. Drouin J, Bilodeau S, Roussel-Gervais A. Stem cells, differentiation and cell cycle control in pituitary. Front Horm Res 2010;38:15–24.
  11. Davis SW, Ellsworth BS, Peréz Millan MI, Gergics P, Schade V, Foyouzi N, Brinkmeier ML, Mortensen AH, Camper SA. Pituitary gland development and disease: from stem cell to hormone production. Curr Top Dev Biol 2013;106:1–47.
  12. Li S, Crenshaw EB, Rawson EJ, Simmons DM, Swanson LW, Rosenfeld MG. Dwarf locus mutants lacking three pituitary cell types result from mutations in the POU-domain gene pit-1. Nature 1990;347(6293):528–33.
  13. Pulichino AM, Vallette-Kasic S, Tsai JP, Couture C, Gauthier Y, Drouin J. Tpit determines alternate fates during pituitary cell differentiation. Genes Dev 2003;17(6):738–47.
  14. Barnhart KM, Mellon PL. The orphan nuclear receptor, steroidogenic factor-1, regulates the glycoprotein hormone alpha-subunit gene in pituitary gonadotropes. Mol Endocrinol 1994;8(7):878–85.
  15. Ngan ES, Cheng PK, Leung PC, Chow BK. Steroidogenic factor-1 interacts with a gonadotrope-specific element within the first exon of the human gonadotropin-releasing hormone receptor gene to mediate gonadotrope-specific expression. Endocrinology 1999;140(6):2452–62.
  16. Lin L, Achermann JC. Steroidogenic factor-1 (SF-1, Ad4BP, NR5A1) and disorders of testis development. Sex Dev 2008;2(4–5):200–9.
  17. Lourenço D, Brauner R, Lin L, De Perdigo A, Weryha G, Muresan M, Boudjenah R, Guerra-Junior G, Maciel-Guerra AT, Achermann JC, McElreavey K, Bashamboo A. Mutations in NR5A1 associated with ovarian insufficiency. N Engl J Med 2009;360(12):1200–10.
  18. 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 Dev 1994;8(19):2302–12.
  19. Lamolet B, Poulin G, Chu K, Guillemot F, Tsai MJ, Drouin J. Tpit-independent function of NeuroD1(BETA2) in pituitary corticotroph differentiation. Mol Endocrinol 2004;18(4):995–1003.
  20. Elster AD Sanders. Size and shape of the pituitary gland during pregnancy and post-partum: measurement with MR imaging.. Radiology 1991;181:531–535.
  21. Bergland RM Ray. Anatomical variations in the pituitary gland and adjacent structures in 225 human autopsy cases. J. Neurosurg. 1968;28:93–99.
  22. LeGros Clark W. The topography and homologies of the hypothalamic nuclei in man. J. Anat. 1936;70:203–214.
  23. Harris GW. Neural control of the pituitary gland. London: Arnold; 1955.
  24. Watts AG. 60 YEARS OF NEUROENDOCRINOLOGY: The structure of the neuroendocrine hypothalamus: the neuroanatomical legacy of Geoffrey Harris. J Endocrinol 2015;226(2):T25-39.
  25. Stojilkovic SS, Tabak J, Bertram R. Ion channels and signaling in the pituitary gland. Endocr Rev 2010;31(6):845–915.
  26. Denef C. Paracrinicity: the story of 30 years of cellular pituitary crosstalk. J Neuroendocrinol 2008;20(1):1–70.
  27. Santiago-Andres Y, Golan M, Fiordelisio T. Functional Pituitary Networks in Vertebrates. Front Endocrinol (Lausanne) 2020;11:619352.
  28. Stefaneanu L, Kovacs K, Lloyd RV, Scheithauer BW, Young WF, Sano T, Jin L. Pituitary lactotrophs and somatotrophs in pregnancy: a correlative in situ hybridization and immunocytochemical study. Virchows Arch B Cell Pathol Incl Mol Pathol 1992;62(5):291–6.
  29. Smith AI Funder. Proopiomelanocortin processing in the pituitary, central nervous system, and peripheral tissues. Endocr. Reviews 1988;9:159–179.
  30. Crooke AC. A change in the basophil cells of the pituitary gland common to conditions which exhibit the syndrome attributed to basophil adenoma. J. Pathol. Bacteriol. 1935;41:339–349.
  31. Charlton HM, Halpin DM, Iddon C, Rosie R, Levy G, McDowell IF, Megson A, Morris JF, Bramwell A, Speight A, Ward BJ, Broadhead J, Davey-Smith G, Fink G. The effects of daily administration of single and multiple injections of gonadotropin-releasing hormone on pituitary and gonadal function in the hypogonadal (hpg) mouse. Endocrinology 1983;113(2):535–44.
  32. Scott IS, Bennett MK, Porter-Goff AE, Harrison CJ, Cox BS, Grocock CA, O’Shaughnessy PJ, Clayton RN, Craven R, Furr BJ. Effects of the gonadotrophin-releasing hormone agonist “Zoladex” upon pituitary and gonadal function in hypogonadal (hpg) male mice: a comparison with normal male and testicular feminized (tfm) mice. J Mol Endocrinol 1992;8(3):249–58.
  33. Childs GV, MacNicol AM, MacNicol MC. Molecular Mechanisms of Pituitary Cell Plasticity. Front Endocrinol (Lausanne) 2020;11:656.
  34. Laporte E, Vennekens A, Vankelecom H. Pituitary Remodeling Throughout Life: Are Resident Stem Cells Involved? Front Endocrinol (Lausanne) 2020;11:604519.
  35. Camilletti MA, Martinez Mayer J, Vishnopolska SA, Perez-Millan MI. From Pituitary Stem Cell Differentiation to Regenerative Medicine. Front Endocrinol (Lausanne) 2020;11:614999.
  36. Perez-Castro C, Renner U, Haedo MR, Stalla GK, Arzt E. Cellular and molecular specificity of pituitary gland physiology. Physiol Rev 2012;92(1):1–38.
  37. Florio T. Adult pituitary stem cells: from pituitary plasticity to adenoma development. Neuroendocrinology 2011;94(4):265–77.
  38. Gleiberman AS, Michurina T, Encinas JM, Roig JL, Krasnov P, Balordi F, Fishell G, Rosenfeld MG, Enikolopov G. Genetic approaches identify adult pituitary stem cells. Proc Natl Acad Sci U S A 2008;105(17):6332–7.
  39. Horvath E, Kovacs K. Folliculo-stellate cells of the human pituitary: a type of adult stem cell. Ultrastruct Pathol 2002;26(4):219–28.
  40. 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(12):949–59.
  41. Baes M Allaerts. Evidence for functional communication between folliculostellate cells and hormone-secreting cells in perfused anterior pituitary aggregates. Endocrinology 1987;120:685–691.
  42. Roncaroli F, Chatterjee D, Giannini C, Pereira M, La Rosa S, Brouland JP, Gnanalingham K, Galli C, Fernandes B, Lania A, Radotra B. Primary papillary epithelial tumour of the sella: expanding the spectrum of TTF-1-positive sellar lesions. Neuropathol Appl Neurobiol 2020;46(5):493–505.
  43. Lee EB, Tihan T, Scheithauer BW, Zhang PJ, Gonatas NK. Thyroid transcription factor 1 expression in sellar tumors: a histogenetic marker? J Neuropathol Exp Neurol 2009;68(5):482–8.
  44. Hagel C, Buslei R, Buchfelder M, Fahlbusch R, Bergmann M, Giese A, Flitsch J, Lüdecke DK, Glatzel M, Saeger W. Immunoprofiling of glial tumours of the neurohypophysis suggests a common pituicytic origin of neoplastic cells. Pituitary 2017;20(2):211–217.
  45. Popa GT; F U. A portal circulation from the pituitary to the hypothalamic region. J. Anat. 1930;65:88–91.
  46. Harris GW. Blood vessels of rabbit’s pituitary gland, and significance of pars and zona tuberalis. J. Anat.1947;81:343–351.
  47. Green JD; H GW. Observations of hypophysioportal vessels of living rat. J. Physiol.1949;108:359–361.
  48. Page RB; B RM. Pituitary Vasculature. In: Allen MB; M VB, ed. The Pituitary: a current review. New York: Academic Press; 1977:9–17.
  49. Bergland RM Page RB. Pituitary-brain vascular relations: a new paradigm. Science 1979;204:18–24.
  50. Bergland RM Page. Can the pituitary secrete directly to the brain? Endocrinology 1978;102:1325–1338.
  51. Coates PJ. The distribution of immunoreactive corticotrophin-releasing factor in the human pituitary stalk. Acta Endocrinol (Copenh) 1985;109(4):433–9.
  52. 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(28):11482–7.
  53. 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. Invest Ophthalmol Vis Sci 2004;45(11):3871–6.
  54. 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. J Pediatr Endocrinol Metab 2011;24(9–10):779–82.
  55. Mortensen AH, Schade V, Lamonerie T, Camper SA. Deletion of OTX2 in neural ectoderm delays anterior pituitary development. Hum Mol Genet 2015;24(4):939–53.
  56. Tajima T, Ishizu K, Nakamura A. Molecular and Clinical Findings in Patients with LHX4 and OTX2 Mutations. Clin Pediatr Endocrinol 2013;22(2):15–23.
  57. 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(2):329–37.
  58. Pfaeffle RW, Savage JJ, Hunter CS, Palme C, Ahlmann M, Kumar P, Bellone J, Schoenau E, Korsch E, Brämswig 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(5):1909–19.
  59. Mullen RD, Colvin SC, Hunter CS, Savage JJ, Walvoord EC, Bhangoo AP, Ten S, Weigel J, Pfäffle RW, Rhodes SJ. Roles of the LHX3 and LHX4 LIM-homeodomain factors in pituitary development. Mol Cell Endocrinol 2007;265–266:190–5.
  60. Machinis K, Pantel J, Netchine I, Léger 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. Am J Hum Genet 2001;69(5):961–8.
  61. 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(3):1062–71.
  62. 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(5):1865–73.
  63. 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(5):673–85.
  64. 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(6):1458–66.
  65. Treier M, Gleiberman AS, O’Connell SM, Szeto DP, McMahon JA, McMahon AP, Rosenfeld MG. Multistep signaling requirements for pituitary organogenesis in vivo. Genes Dev 1998;12(11):1691–704.
  66. 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(8):2822–31.
  67. 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(10):E1709-18.
  68. Dodé C, Levilliers J, Dupont JM, De Paepe A, Le Dû N, Soussi-Yanicostas N, Coimbra RS, Delmaghani S, Compain-Nouaille S, Baverel F, Pêcheux 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. Nat Genet 2003;33(4):463–5.
  69. 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(3):377–86.
  70. Kelberman D, Dattani MT. Role of transcription factors in midline central nervous system and pituitary defects. Endocr Dev 2009;14:67–82.
  71. Cogan JD, Wu W, Phillips JA, 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(9):3346–9.
  72. Deladoëy J, Flück C, Büyükgebiz A, Kuhlmann BV, Eblé 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(5):1645–50.
  73. 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(1):10–8.
  74. 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. Arq Bras Endocrinol Metabol 2007;51(7):1097–103.
  75. 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(5):587–98.
  76. 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(6):785–94.
  77. 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(1):60–4.
  78. Shinoda K, Lei H, Yoshii H, Nomura M, Nagano M, Shiba H, Sasaki H, Osawa Y, Ninomiya Y, Niwa O. Developmental defects of the ventromedial hypothalamic nucleus and pituitary gonadotroph in the Ftz-F1 disrupted mice. Dev Dyn 1995;204(1):22–9.
  79. 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 Dev 2003;17(6):711–6.
  80. Vallette-Kasic S, Brue T, Pulichino AM, Gueydan M, Barlier A, David M, Nicolino M, Malpuech G, Déchelotte 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(3):1323–31.

Graves’ Disease and the Manifestations of Thyrotoxicosis

ABSTRACT

 

Graves' disease (GD) is an autoimmune disorder characterized by hyperthyroidism and various systemic manifestations, including thyroid eye disease (TED) and, less commonly, dermopathy. This chapter provides an in-depth review of GD, covering its history, epidemiology, risk factors, and the molecular mechanisms underlying autoimmune hyperthyroidism. Emphasis is placed on emerging insights into the genetic, environmental, and immunological factors contributing to GD's multifactorial pathogenesis. The chapter also explores the pathogenic role of TSH receptor antibodies and their significance in diagnosis and treatment, alongside key clinical features of thyrotoxicosis.

 

INTRODUCTION

 

Graves' disease (GD) is an autoimmune form of hyperthyroidism characterized by multisystemic manifestations of thyrotoxicosis and, in some cases, extrathyroidal manifestations of thyroid autoimmunity including, frequently, thyroid eye disease (TED) and rarely pretibial dermopathy. There are therefore diverse phenotypic presentations of thyroid autoimmunity, and patients do not necessarily present with thyrotoxicosis as the main manifestation of their autoimmune phenomena. Autoimmune thyroid disease (AITD) typically includes two major categories of manifestations. Those specific to GD, and caused by the autoimmunity per se, including goiter, TED, and dermatopathy (TED and dermopathy are discussed in the chapter “Graves’ disease: Complications”). The second set of problems is caused by the excess thyroid hormone and its widespread disturbance in metabolism. These thyrotoxic manifestations do not differ from those induced by any other cause of excess of thyroid hormone. Alternative etiologies of thyrotoxicosis are described in other chapters.

 

HISTORY

 

The triad of goiter, tachycardia, and ophthalmopathy was independently described by Caleb Parry (1755-1822) and Karl A. Von Basedow (1799 - 1854), but it is the description by the Irish physician Robert James Graves (1796 - 1853) (Figure 1) (1) that is the source of the acronym in the English literature, Graves' disease (GD). However, in many countries, the term Morbus Basedow is widely used to describe autoimmune hyperthyroidism. The autoimmunity reflected in GD is related to antibodies directed against the TSH receptor (TSH-R) expressed at the basolateral membrane of thyroid follicular cells. Initially, serum factors that were thought to be a modified TSH molecule and produced exophthalmos in experimental animal models were given the eponym exophthalmos producing substance (2, 3). Later, Adams et al. identified a factor in human serum that could stimulate the release of thyroid hormone in guinea pigs and a human subject (Figure 2) (4, 5). Because of the time course of its action being longer than TSH, this material was named Long-Acting Thyroid Stimulator, or LATS. Subsequent studies better characterized this substance as an immune gamma globulin that acts on a thyroid antigen, and mimics the action of the natural thyroid stimulator, TSH. Nearly three decades later, the antigen to which this antibody was directed was identified as the TSH-R (6, 7). Over the past four decades, there has been improved understanding of the accurate identification, quantitation, and pathophysiologic importance of the thyroid receptor antibodies (8-10), as will be further detailed throughout this chapter.

 

Figure 1. Robert James Graves, Statue. Marble statue of Robert James Graves, former president (1843–1849) of the Royal College of Physicians of Ireland. The statue was commissioned by John Henry Foley who died before completing it. Graves Hall, the Royal College of Physicians of Ireland, No. 6, Kildare Street, Dublin, Ireland. Photographer: Osama Shukir Muhammed Amin FRCP(Glasgow). Copyrighted work available under Creative Commons Attribution only license CC BY 4.0.

Figure 2. Stimulation of thyroid hormone secretion by LATS-P. The subject's thyroid iodine was labeled by administration of I131, and serial observations were made on the appearance of I131-labeled hormone in blood (Y axis) over one month (X axis). An infusion of 280 ml control plasma had no effect, but 280 ml plasma from a patient with Graves' disease caused a marked stimulation of secretion of hormone from the thyroid. (Adapted from: D.D. Adams et al., J. Clin. Endocrinol. Metab., 39:826, 1974).

 

EPIDEMIIOLOGY

 

GD has an estimated incidence of 30 to 80 cases per 100.000 persons per year and occurs more frequently in the white population while it appears to be less frequent in the Asian and Sub-Saharan African populations (3.8 to 5, and 0.7 to 51 cases per 1000.000 persons per year, respectively) (11-13). Female (lifetime risk 3%, eight-folds greater than in men) and middle-aged individuals (30 to 50 years of age) are at higher risk. The manifestations of disease tend to be more pronounced in patients younger than 65 years, while older patients tend to have an abbreviated presentation, with mainly cardiac manifestations (14, 15). Around 50% of patients with GD have a family history of autoimmune thyroid disease (AITD). Aside from the infrequent occurrence of postnatal thyrotoxicosis due to maternal antibodies, the incidence of spontaneous GD in children before the age of ten is unusual, but the incidence climbs with each decade until about age 60 (16-20).

 

RISK FACTORS

 

GD is a consequence of autoimmunity against the TSH-R. The next question revolves around the trigger for this specific autoimmunity. Contemporary understanding is that a multi-factorial process allows thyroid self-reactivity to occur. While our immune system is designed to prevent self-reactivity, to some extent, exceptionally low levels of self-reactivity are normally present (21). Sex, environmental, immunologic, and genetic factors interact to augment this immune response to a degree that predisposes to clinically manifest autoimmunity (Figure 3). Several such factors have been identified with some certainty, and others have been suggested.

 

Figure 3. Risk factors for Graves’ disease. MHC: Major histocompatibility complex; HLA: Human leukocyte antigen; CTLA: Cytotoxic T-lymphocyte-associated protein; RNA: Ribonucleic acid. (Created with Biorender.com)

 

Sex

 

The clearest association with any autoimmune condition is female sex, which carries a 10-20-fold risk compared to men (22). Although, the mechanism behind the increased incidence of autoimmune conditions in female is not fully understood, several factors leading to greater autoreactivity towards the thyroid have been proposed. Polymorphisms in the estrogen receptor genes might be at least partially responsive for the increased susceptibility of female to developing GD (23). In addition, marked fluctuations in leptin and other hormonal secretion patterns throughout the different endocrine states during the female lifespan (Including puberty, pregnancy, and menopause) have been linked with changes in the regulation of the innate and adaptive immune systems, and disbalance between pro and anti-inflammatory states, which altogether increase the risk of developing autoimmune diseases (22, 24).

 

The post-partum period is associated with a higher risk of developing GD. This relates to the transition from a skewed T helper 2 (Th2) cell response that suppresses the immune response and antibody production (Including TSH-R antibodies) during pregnancy, to a post-partum state in which the proinflammatory Th1 cell response returns to normal (25). Furthermore, it has been shown that fetal cells from male infants can persist in the maternal circulation for up to 20 years (fetal micro chimerism). These cells have been found in frozen thyroid tissue specimens from patients with GD and thyroid nodules, and it is hypothesized that they could modulate AITD by induction of immune response or development of a graft-versus-host immune response to the mother (26).

 

Environmental Factors

 

Considerable information has accumulated about factors in the environment that can induce GD. Cigarette smoking increases the risk of GD and complicates its extra thyroidal manifestations through direct cell damage, downregulation of the innate immune system, and induction of epigenetic changes (27-31). Conversely, moderate alcohol use and obesity seem to decrease the risk of developing the disease (32), while physical activity seems neutral in that respect (33). Other factors include:

 

INFECTIONS AND MOLECULAR MIMICRY

 

A persistent theory on the etiology of autoimmune diseases is that exposure to a particular peptide epitope in an environmental antigen might lead to immune reactivity to an amino acid sequence identical to that present in a human antigen such as the TSH-R, thyroperoxidase (TPO), or thyroglobulin (Tg). Through this molecular mimicry, exposure to a virus or bacteria could produce heightened immune reactivity towards these autoantigens. This sequence is believed to play a role in rheumatic fever and glomerulonephritis. There is some evidence that proteins present in a common intestinal parasite, Yersinia enterocolitica, may induce antibody reactivity to the TSH-R (34-36). While a clear association has not been established, a higher proportion of patients with GD have been infected with this bacteria than people without the disease (34) and exposure to the bacteria can induce TSH-R antibodies (TRAb) (35).

 

There is also evidence that autoimmunity to the thyroid can be induced by infection with the human T-lymphotropic virus type 1 (HTLV-1), which causes lymphocytic leukemia (37). Whether this is due to molecular mimicry of the virus, viral damage to the thyroid, or stimulation through another mechanism such as cytokine secretion, remains uncertain. In addition, it has been suggested that Helicobacter pylori (H. pylori) antigens may be involved in the development of AITD, and one study reported that up to 85% of patients with autoimmune atrophic thyroiditis have H. pylori infections (38). While molecular mimicry remains a tantalizing explanation, the factual evidence for its role in the pathogenesis of GD is minimal.

 

THYROID INJURY AND ANTIGEN RELEASE

 

It is accepted that certain types of injury to the thyroid are followed by the development of thyroid autoimmunity, including GD. In fact, this is one of the few proven causes of GD. The release of thyroid antigens following destruction of thyroid tissue may add a significant stimulation to a latent low level of thyroid autoimmunity, causing the development of GD. Radiation to the thyroid has been associated with a subsequent higher incidence of positive thyroid antibody tests (39), and an increased risk of GD, Hashimoto's thyroiditis, and TED (40, 41). Around 1% of patients with autonomous thyroid nodules who undergo radioactive iodide treatment or ethanol injection can subsequently develop GD (42, 43), with a tenfold higher risk if anti-TPO antibodies are present (44). Whether viral injury, as in the case of HTLV-1 and SARS-CoV (45, 46), plays a similar role in human AITD is uncertain. However, Covid-19 disease caused by the SARS-CoV has been associated with higher risk of developing subacute thyroiditis, GD, and TED.

 

IMMUNE THERAPIES

 

Administration of cytokines such as INF-α, IL-2, and GM-CSF can augment AITD, or in some cases appear to induce it de novo (47-49). Alemtuzumab therapy, used for depletion of circulating lymphocytes in the therapy of multiple sclerosis is followed by development of GD in a third of such treated patient within 6 months of recovery from T cell depletion (50). This and other immunosuppressive treatments may deviate the immune system from a Th1 to a Th2 type of response, reducing the number of regulatory T cells or altering the balance between T helper 17 (Th17) cells and regulatory T cells, and leading to overexpression of pro-inflammatory cytokines IL-17, IL-22, IL-23R, and IL-32 that are commonly seen in patients with AIDT (51-55). In addition, treatment with INF-α for chronic hepatitis or INF-β for multiple sclerosis has also been suggested to cause AITD and destructive thyroiditis, especially in females (RR: 4.4) and people with pre-existing anti-TPO antibodies (RR: 3.9) (56); however, a definitive association has not been established (57). In addition, while immune check point inhibitors can commonly induce hypothyroidism and thyrotoxicosis due to destructive thyroiditis (58), in rare instances they can also induce GD (59, 60).

 

EXCESS IODINE

 

Excess iodide can induce hyperthyroidism in patients with multinodular goiter (61, 62), a phenomenon known as “Jod Basedow”. Presumably, autonomous nodules in the goiter are in a subclinical state and unable to produce an excess of thyroid hormones as their synthesis is limited by iodide supply; yet, when the iodine supply is high, the autonomous nodules produce an excess of hormone. Well studied epidemics of iodide-induced thyrotoxicosis occurred for example in Tasmania, Denmark, and India after the introduction of salt iodization and were clearly associated with multinodular goiters rather than typical GD (63-66). In addition, increased iodine intake can actually augment thyroid autoimmunity through other mechanisms such as heavy iodination of Tg which appears to increase immunogenicity in animal studies (67), or hypothetical induction of follicular thyroid cell injury with secondary liberation of thyroid related antigens (68, 69). On the other hand, the addition of 2 - 6 mg per day of iodide to the intake of most patients with GD causes a dramatic, but probably temporary, reduction in hormone release, a phenomenon referred to as Plummer effect (70-73). Overall, whether an excess of iodine can induce true GD and autoimmunity remains unknown.

STRESS

 

The incidence of GD increased in Denmark during World War II (74), and in Serbia during the civil war in the former Yugoslavia (75). In general, studies have shown mixed results; however, meta-analysis suggests physical trauma and psychologic stress are possible environmental triggers of GD (76). Some authors indicated that patients with GD had suffered on average more stressful episodes than control subjects, but other similar studies failed to show this relationship (77-80). Stress induces a variety of physiologic responses including anxiety, tachycardia, and restlessness, among others, which are not unlike symptoms of GD. Its role remains enigmatic in causation of GD to this date. Theoretically, stress might cause activation of the adrenal cortex or the sympathetic nervous system which might cause stimulation of thyroid secretion, as has been shown in experimental animals (81). Other specific stressors have been reported. Aggressive weight loss programs that involve using exogenous thyroid hormone have been reported to induce GD (82).

 

Immunologic Factors

 

THYMIC SELECTION OF LYMPHOCYTES

 

Lymphocytes develop from precursors present in the bone marrow that undergo progressive maturation and selection in the thymus. Lymphocytes which fail to recognize endogenous human leukocyte antigens (HLA) undergo negative selection, as do those which strongly react with endogenous epitopes presented by HLA molecules (83, 84). In this process, more than 95% of all lymphocytes undergo apoptosis. As with other human molecules, thyroid molecules like the sodium iodine symporter (NIS), TSH-R, TPO, and Tg-RNAs are presented as immunoreactive peptides in the human thymus as part of the lymphocyte selection process (85, 86). Pre-T lymphocytes are thus educated in the thymus to recognize thyroid-related epitopes, and to generate self-tolerance against them. Expression of these thyroid antigens in the fetal thymus is under control of the AIRE gene, and absence of this gene leads to polyglandular autoimmune syndrome type 1 whose hallmarks are adrenal insufficiency, hypoparathyroidism and muco-candidiasis. In Down’s syndrome, despite having 3 copies of the AIRE gene, expression of thyroid antigens in the thymus is reduced, and this is thought to cause the elevated incidence of autoimmunity in these patients (87). Clearly the thymic selection process is imperfect, in a developmental process designed to provide the maximum repertoire of lymphocytes, some lymphocytes which weakly recognize autologous thyroid antigens do persist in the circulation, and can be found in normal patients and those with AITD (Table 1) (85).

 

REGULATORY T CELL ABNORMALITIES

 

Every T cell-mediated response is tempered by the interaction of effector and regulatory cells (88). Thus, a disproportionately low number or diminished function of regulatory T cells is, in part, responsible for the development of autoimmunity (89, 90). Several studies have demonstrated abnormalities in T cell regulation in GD with conflicting results. Some groups have proposed that patients with GD have a lower proportion of regulatory T cells (91-95), while others report a normal or high number of T regulatory cells but with impaired immune suppressive function (91, 96-100). In addition, Marazuela et al. reported a high proportion of defective intrathyroidal regulatory T cells in patients with autoimmune thyroiditis (101). Gangi et al. found that administration of GM-CSF induced development of regulatory T cells capable of suppressing immunity to Tg in mice (102). Molteni et al. reported that CD8 T cells can induce anergy in TSH-R specific CD4 T cell clones to prevent thyroid related auto reactivity (103) and, ultimately, Vaidya et al. reported higher numbers of naïve activated T cells and lower memory T cells in patients with TED (104). Overall, while there remains some uncertainty, decreased T regulation due to low cell counts or impaired function is involved in the development of GD.

 

EXPRESSION OF MAJOR HISTOCOMPATIBILITY COMPLEX (MHC) CLASS II

 

Exposure of thyroid epithelial cells to interferon, presumably secreted from infiltrating lymphocytes or other immune cells, can lead to the expression of MHC class II molecules on the thyroid cell surface (105, 106). Expression of these molecules allows the thyroid epithelial cell to function as a weak antigen presenting cell (APC) (107, 108). Culture of human thyroid cells from patients with GD in vitro shows that the expression of MHC class II molecules expression disappears (109), as it does when the cells are transplanted into nude mice (110). This is a compelling argument indicating that MHC class II expression on GD thyroid cells is secondary rather than primary event.

 

Table 1. Possible Factors in the Immunological Etiology of Graves' Disease

-       Persistence of some autoreactive T cells and B cells (failure of negative selection)

-       Genetic polymorphisms

-       Re-exposure to antigens secondary to thyroid cell damage

-       Reduced or dysfunctional regulatory T cells

-       Cross-reacting epitopes on environmental and thyroid antigens

-       Inappropriate HLA-DR expression

-       Mutated T or B cell clones

-       Activation of T cells by polyclonal stimuli

-       Stimulation of the thyroid by cytokines

HLA: Human leukocyte antigen; CTLA: Cytotoxic T-lymphocyte-associated protein

 

Genetic Factors

 

The increased incidence of GD in certain families and in identical twins has for decades indicated a powerful genetic influence on disease development (111-113). Studies of large samples of pairs of twins suggest that the genetic factors account for 73-79% of the liability to have positive TSH-R antibodies and to develop GD, whereas environmental factors account presumably for the remainder (113, 114). Adjusting for covariates (age, TSH and others), the estimate for genetic influence on serum anti-TPO and anti TG antibodies was also found to be between 60 – 80%. This inheritance turns out to be polygenic. Rather than inheriting one gene which, in a dominant fashion, would induce GD, individuals inherit many different genetic polymorphisms which are conducive to the development of AITD (115-117).

 

HUMAN LEUKOCYTE ANTIGEN (HLA) MOLECULES

 

HLA molecules are expressed in different isotypes. There are between 50 and 100 different HLA-DR molecules, and a much smaller number of HLA-DQ and HLA-DP molecules, all coded on chromosome 6, in the human genome (118). Of these, the HLA-DR are most abundantly expressed and most important. These molecules exist as dimers on the surface of antigen presenting cells. In the initiation of an immune response, the antigen presenting cell displays a specific epitope complexed in an HLA-DR. Recognition of this bi-molecular complex by the T cell receptor leads to stimulation of T cells. The amino acid sequences of the HLA-DR molecule determine the shape and affinity to the antigen presenting cleft (119). Thus, certain HLA-DR molecules are more efficient to present certain epitopes during the lymphocyte selection and maturation process in the thymus (84). Therefore, the inheritance of certain HLA genes, and the matching of the HLA-DR molecules with the structure of the TSH-R epitopes, or other thyroid related epitopes, plays a significant role in determining the development of AITD.

 

The first genetic factor to be associated with GD was HLA-B8 (120), a class I major histocompatibility component (MHC). Subsequently, this relation was found to be more specifically significant with an MHC class II molecule, HLA-DR3 (117). Inheritance of this gene, expressed on the surface of antigen presenting cells, was found to confer up to 5.7-fold increased risk of developing GD (121-123). Additional data demonstrated that the HLA molecules DQA1*0501, DRB1*0301, DQB1*0202, DQB1*0603, DQB1*0609, DQB1*0302, DQB1*0303 were also closely associated with higher risk of developing GD (124-128). In contrast, inheritance of HLA-DR beta 1*07, DQB1*0201, DQB1*0502, and DQB1*0602 appear to be protective (128, 129).

 

CYTOTOXIC T LYMPHOCYTE ASSOCIATED PROTEIN (CTLA)

 

When an immune reaction begins, the “first signal” is the recognition by the T cell receptor of an HLA molecule. If only one signal occurs, the T cell tends to be turned off or “anergized.” In order for a progressive immune response to occur, there must be a "second signal" provided by one of several adhesion molecules which exist on the antigen presenting cells (APCs) and T cells, and which tend to augment the affinity of the interaction (130). Of these, one of the most important is "B7", which exists in two forms, B7.1 and B7.2, present on the surface of APCs. These molecules interact with their cognate receptors on the T cell, CD28 for B7.1 and CTLA-4 for B7.2. In many situations interaction between B7.1 and CD28 give a positive stimulus to growth of the T cell, whereas interaction of B7.1 with CTLA-4 provides a negative signal, suppressing autoimmunity (131). CTLA-4 exists as a gene with several isoforms, and the inheritance of some of them is associated with a higher incidence of GD (132-137), and increased production of anti-TPO and anti-Tg antibodies (138).

 

Interestingly, the HLA association suggests a relationship to GD specifically, since it has to do with the presentation of specific antigen epitopes, whereas the CTLA-4 polymorphism appears to be a general phenomenon, allowing one population group to have augmented lymphocyte proliferation, but is not specifically related to thyroid disease. These observations also fit with the concept that development of GD is mediated by a set of genes rather than one specific gene.

 

NON-CODING RNAs

 

The RNA transcripts that regulate genetic transcription and protein translation, but do not encode a specific protein themselves, are denominated non-coding RNAs (139). These regulators play a key role in immune activation, cellular proliferation, and cytokine production. Thus, their abnormal expression can halt the normal immune response, leading to autoimmunity. In the case of AITD, the understanding of non-coding RNAs associated with development of disease is still limited. Several transcripts have been proposed to play a role in the development of GD but their exact mechanism of action, and their utility for clinical care are unclear (140-142). Ongoing and future efforts aim to clarify the role of non-coding RNAs as well as their potential use as diagnostic biomarkers or therapeutic targets (143).

 

EXOSOMES

 

Exosomes are cell-secreted extracellular vesicles that function as biological carriers, and contain a series of molecules (i.e., cytokines, transduction factors, nucleic acids, proteins, and lipids) involved in processes of cellular communication such as antigen presentation and immune response activation. As in other autoimmune conditions, exosomes are presumed to play a role in AITD but research on their pathophysiologic role is still in the early stages (144, 145). Some authors have previously reported the presence of thyroid follicular cell derived exosomes containing TSH-R which are involved in the development of GD (146, 147). In addition, exosomes from patients with GD have been found to stimulate the production of proinflammatory cytokines such as IL-1, IL-6, and TNF-α (148). Furthermore, exosomes containing metalloproteinases and non-coding RNAs have been found in high proportions in patients with AITD and TED (149, 150).

 

OTHER FACTORS

 

Numerous other gene polymorphisms have been reported to be associated with GD. It is highly likely that these individual variants contribute a real but small increment in the risk of developing the disease. The initial genetic studies on the TSH-R showed mixed results on a potential association between specific polymorphisms and susceptibility to GD (151, 152). However, more recent studies have endorsed this association (153, 154). Certain genetic variants in TPO and Tg have also been associated with TED and AITD but their role in the development of disease is unclear (155-159). In addition, a vitamin D receptor polymorphisms have been associated with GD and other autoimmune conditions (160). Inheritance of specific genes coding for immunoglobulins may carry the same kind of risk. Additionally, several possible genes linked to GD or AITD have been found by linkage studies, including one recently described at a locus on chromosome 18q21 that is also associated with type 1 diabetes (161).

 

Other gene polymorphisms can lead to increased susceptibility to GD including CD40, CD25, and ZFAT (required for the correct functioning of B cells and other immune cells) (117, 162-164), CYP27B1 (involved in the activation of vitamin D) (165), IL-6 (166), IL-13 (167), IL-1 (168), IL-23R (169), TNF-α (170), protein tyrosine phosphatase-22 (PTPN22) (involved in inhibition of T cell activation) (171-174), Fc receptor-like protein 3 (FCRL3)(175-177), transforming growth factor beta1 (TGF-β1) (178), interferon inducible helicase 1 (IFIH-1)(179). Other proposed genes associated with AITD include FOXP3, TBX21, HLX, BTNL2, NOTCH4, and CXCR4 (158, 180).

 

In conclusion, there are several genes harboring polymorphisms that appear to augment the possibility of developing immunity to thyroid gland protein components. These genetic alterations are likely to interplay with epigenetic aspects of DNA methylation and histone modifications (a phenomenon referred to as epistasis), along with environmental interventions, leading to the overall individual susceptibility to GD.

 

PATHOGENESIS

 

GD and Hashimoto's thyroiditis are strongly associated and, in fact, overlapping syndromes. They share immunological abnormalities, histological thyroid changes, and genetic predisposition. Patients can switch from one category to the other, depending upon the stage of their illness. For example, an individual might first present with hypothyroidism, thyroid enlargement and positive anti-thyroperoxidase (TPO) antibodies, and thus qualify as having Hashimoto's thyroiditis. At a later stage, this individual might become hyperthyroid with positive TSH-R autoantibodies and fit in the category of GD. Conversely, a patient with hyperthyroidism might have progressive destruction of the thyroid, or develop blocking antibodies to the TSH-R, and become hypothyroid. The aspects explaining the autoimmunity and pathologic changes at the thyroid level are explained in this section. Additional thyroidal pathophysiologic changes are explained later in the chapter.

 

The common feature of AITD is the immune reactivity, both humoral and cell-mediated, to specific thyroid antigens such as the TSH-R, TPO, and Tg (21, 181). Antibodies also exist, among others, to megalin (the thyroid cell Tg receptor) (182), to the thyroidal iodide symporter (183), and to components of eye muscle and fibroblasts in patients with thyroid eye disease (184) (Table 2). Up to 90% of patients with GD have anti-TPO antibodies, and up to 50% have anti-Tg antibodies (185-189). Rarely patients have antibodies directed against T4 or T3 (187). Peripheral blood mononuclear cells (190), thyroid lymphocytes (191), and lymph node lymphocytes demonstrated cell-mediated immunity to TPO, Tg and TSH-R (192, 193), and to specific epitopes of these molecules (194-199). Anti-TPO antibodies are not known to play a role in GD, but they could cause a certain degree of cytotoxicity, as noted in couple in vitro studies using serum from AITD patients (200, 201). The functional consequence of having anti-Tg antibodies is uncertain, but they do not appear to cause thyroid cell destruction. Tg and anti-Tg antibody immune complexes are rarely deposited in the kidney basement membrane of the glomeruli and can, in extremely rare circumstances, produce nephritis (200, 202, 203).

 

Table 2. Antibodies in Graves' Disease

Antibody

Graves’ disease

Hashimoto’s thyroiditis

Healthy controls

TSH-receptor antibodies (TRab) (204-209)

Thyroid stimulating antibodies (TSAb)

TSH- blocking antibody (TBAb)

Neutral antibodies

99%

 

10%

2%

Anti-thyroperoxidase antibodies (Anti-TPOAb) (204, 205)

80%

90-95%

10-15%

Anti-thyroglobulin antibodies (Anti-TgAb) (205, 210, 211)

30-60%

60-80%

8-11.5%

 

In addition, patients with AITD often develop other organ-specific antibodies, and have higher risk of developing associated conditions such as Addison's disease, premature ovarian failure, chronic hepatitis, celiac disease, primary biliary cirrhosis, pernicious anemia, type 1 diabetes, multiple sclerosis, myasthenia gravis, vitiligo, rheumatoid arthritis, systemic lupus erythematous, systemic sclerosis, idiopathic thrombocytopenic purpura, urticaria, alopecia, and angioedema (212-220). The most frequent extrathyroidal antibodies include antibodies directed to gastric parietal cells, found in 50% of patients with Hashimoto’s thyroiditis (221), to adrenal steroidogenic enzymes, ovarian steroidogenic enzymes, components of the pituitary gland (222), DNA (223), liver mitochondria GD(224), and to cardiolipin (225). Further evidence of ongoing inflammation in GD is the elevation of ICAM-1, IL-6 and IL-8 cytokines seen in hyperthyroid patients (217, 226).

 

TSH Receptor Antibodies

 

These antibodies can be classified into three major categories:

 

  1. Thyroid stimulating antibodies (TSab) interact with the TSH-R and stimulate adenyl cyclase and the protein kinase A pathway, as well as the phospholipase C pathway, thereby triggering thyroid hormone synthesis and cell proliferation (227-229). Functionally, this is identical to the effects induced by TSH itself. When TSab reaches a certain level of function, they cause an increase in thyroid hormone synthesis and secretion, as well as growth of the gland, and cause hyperthyroidism.
  2. Thyroid blocking antibodies (TBAb) interact with the TSH-R by binding to different epitopes on the receptor, and they can block the binding of TSH to the receptor without stimulating function themselves (230-233).
  3. Thyroid neutral antibodies (TNab) bind mostly to the hinge region of the TSH-R. While these antibodies neither stimulate nor inhibit its function, they appear to be involved in signaling activity leading to apoptosis of the thyroid follicular cells (234-236).

 

All these antibodies are commonly recognized by assays which detect their ability to interfere with the binding of TSH to the receptor and are identified as thyrotropin receptor antibody (TRAb), or Thyrotropin binding inhibiting immunoglobulins (TBII) (231). Probably all patients with GD have a mixture of all these three subtypes of antibodies.

 

TRAb/TBII can be measured with enzyme linked immunosorbent assays (ELISA) that measure the ability of the patient’s serum antibodies to inhibit binding of labeled TSH-R ligands (human monoclonal thyroid antibodies) to the TSH-R (209). In addition, TSab can be identified by a bioassay which quantifies the ability of the antibodies to stimulate the adenyl cyclase function of the TSH-R. Either thyroid cells or thyroid cell membranes can be used, and the cyclic AMP produced by this stimulation is quantitated by a luciferase-based assay (209, 227). A cyclic AMP responsive luciferase construct introduced into Chinese hamster ovary (CHO) cells, allows a sensitive luminescent assay for thyroid stimulating antibodies with the capability of high throughput suitable for use in general laboratories (228).

 

The TSH-R is initially formed as a single polypeptide inserted into the thyroid cell membrane. After processing, it becomes a two-peptide structure, one extracellular and one transmembrane, with the chains held together by disulfide bonds. Subsequently, reduction of the disulfide bonds by a disulfide isomerase may separate the two molecules and lead to shedding of the “alpha” subunit, which can be augmented by TSH stimulation of thyroid cells (237). The amino-terminal ectodomain of the human TSH-R has been expressed on the surface of CHO cells as a glycosylphosphatidylinositol-anchored molecule. This material can be released from the cells and is biologically active by binding immunoglobulins from serum of patients with GD, and displaying saturable binding of TSH (8), indicating that all of the "immunologic information" related to production of antibodies resides in the extracellular portion of TSH-R.

 

The initial bioassay developed by Adams et al. (5), and then by McKenzie (238), could quantitate TSAb (or LATS as it was then known) in up to 60% of patients with active GD. Newer assays measuring cyclic-AMP formation can detect TSab in over 99% of patients with GD and thyrotoxicosis (239), with a sensitivity of 97% to identify patients with GD, and 93% for patients with TED (240). The presence of TSab is characteristic of active GD, and if the thyroid can respond (e.g., has not been ablated with RAI), hyperthyroidism ensues. If untreated, the thyroid may be destroyed by the ongoing immune process, or blocking antibodies can develop, and the patient may become hypothyroid. In addition, the coexistence of TSab and TBab can cause a pull-push effect leading to shifting between hypo- and hyperthyroidism (241, 242).

 

During antithyroid therapy, TSab tends to decline. If their titer becomes undetectable there is a high likelihood of disease remission. However, if present in significant concentration, remission is very unlikely, explaining the failure of safely discontinuing antithyroid drug therapy (ATD) in more than 50% of patients after 12-18 months of treatment (243, 244). Similarly, TSab tends to decline after thyroidectomy (245). After radioactive iodide therapy, TSab titer increases for up to a year, probably because of the release of thyroidal antigens (246-248). Antibody levels gradually decrease during the subsequent years, reaching pretherapy values around the third year. During ATD, it is speculated that some immune modulation might occur, and the predominant TSab are replaced by TNab or Tbab.

 

The specific epitopes to which the TSab bind are in the amino terminal portion of the extracellular domain of the TSR-R and have been better characterized (249, 250). This has led to therapeutic efforts to induce tolerance to these epitopes (251, 252). The blocking antibodies tend to bind to sequences at the carboxy terminal portion of the receptor, closer to the plasma membrane, probably preventing the activation of the receptor through inhibition of signal-transmission required conformational changes (253). Details of these actions are revealed by cryo-electron microscopy analysis of the full-length TSH-R structure complexed with the inhibitory antibody K1-70™. This approach revealed key interactions between the receptor's transmembrane and extracellular domains, suggesting that K1-70™ inhibits TSHR by binding without activating it, while the stimulatory autoantibody M22™ likely induces activation through conformational changes in the hinge region (254).

 

The Role of Cellular Immunity

 

Lymphocytes of patients with GD are reactive to the TSH-R (Figure 4), TPO, and synthetic TPO-derived peptides (190, 199, 200, 255). The interaction between antigen presenting cells (APCs) and T cells triggers a multimodal cellular and humoral response. T helper 1 (Th1) cell derived cytokines such as interferon gamma (INF-γ), tumor necrosis factor alpha (TNF-α), interleukin (IL) 1β, IL-6, IL-9, IL-10, and IL-11 play a dominant role in the early phase of AITD (256). Later, there is a predominant T helper 2 (Th2) cell response with its derived IL-4, IL-10, IL-17, and IL-23 (257-260). TSH-R peptides that have aspartic or glutamic acid in the fourth position of their binding motif are particularly susceptible at inducing immune responses in GD and animal T cells (190, 261). Immunoreactivity towards the TSH-R has been reported in samples of orbital fat and preadipocytes, and differentiation of preadipocytes into adipocytes with high TSH-R reactivity has been induced by TSH stimulation (262-264). In addition, immunity to the TSH-R plays a direct role in the development of TED, through the secretion of proinflammatory cytokines by T cells.

 

Figure 4. The role of cellular immunity. Schematic representation of TSH-R endocytosis by antigen presenting cells (APC), subsequent proteolysis and presentation to T cells, and stimulation of thyroid stimulating antibody production by B cells. (Created with Biorender.com).

 

PATHOPHYSIOLOGY

 

Thyroid Function

 

In GD, the thyroid gland is functioning at an accelerated rate. Serum TSab stimulate the TSH-R, as evidenced by the higher activity of the cell membrane adenyl cyclase activity in the thyroid tissue of patients with GD, compared to normal thyroids (265). The plasma iodine clearance, a reflection of the thyroid iodine uptake, is increased from the normal rate of 10-20 ml/min to 40-2000 ml/minute. For this reason, the percentage of a tracer dose of radioactive iodine (RAI, I123) found in the thyroid gland at 12 hours is elevated and distributed in a homogeneous fashion (266). Thyroid hormones, Tg, and iodotyrosine - normally secreted in minute amounts - are released into the blood rapidly and at increased rates (73, 267, 268). Furthermore, the rate of turnover of plasma thyroid hormones is also increased. Accelerated degradation is probably secondary to hypermetabolism and is not a primary event (269), although it has been reported that accelerated T4 turnover can persist after treatment of thyrotoxicosis (270). Finally, after anti-thyroid drugs (ATD) the thyroid hormones tend to fall and that response, along with the required dose of ATD drug required to achieve that response, can be used as predictors of GD remission (271).

 

An interesting historical observation regarding thyroid function during GD is that the uptake of RAI by the thyroid is not suppressed by the administration of exogenous T4 or T3 (272, 273), even if large amounts of hormone were given (old studies performed before the autoimmune basis for GD was fully understood). Non-suppressibility is caused by stimulation of the thyroid by TSab, and independence of feedback control via TSH.

 

Iodine Effect on the Thyrotoxic Gland

 

Iodine affects the metabolism of the diffusely hyperplastic thyrotoxic gland in a way radically different from its action on the normal gland. Years ago, Plummer demonstrated that GD can be temporarily or permanently controlled by the administration of iodide (70). Administration of large doses of iodine to laboratory animals causes a temporary inhibition of iodine organification, the Wolff-Chaikoff effect (274). The same phenomenon occurs in humans, and thyrotoxic patients are especially sensitive to this effect. The thyroid uptake of I131 is acutely depressed in thyrotoxic patients by administration of 2 mg potassium iodine, whereas more than 5 mg is needed to depress uptake in normal subjects. Concentrations of serum iodine above 5 µg/dl block iodine uptake and binding in the thyrotoxic gland (70, 71, 73, 272, 275). The Wolff-Chaikoff effect is transient. With continuous iodine administration, I131 uptake and binding recommences. The adaptation to excess iodine in animals involves a reduction of iodine transport into the thyroid which lowers intrathyroidal iodine content and escape from the Wolff-Chaikoff block. This adaptation occurs independently of TSH action.

 

The biochemical mechanism of the Wolff-Chaikoff effect has not been fully elucidated, but it is partially mediated by the downregulation of the sodium-iodide symporter (NIS) (276). Iodine does not prevent TSH or TSab from binding to the TSH-R but inhibits both the TSH-stimulated adenyl cyclase production of cAMP and its derived actions. In addition, iodine also causes a marked reduction in the release of previously formed hormones from the thyrotoxic gland but does not completely prevent further hormone synthesis. This phenomenon has been repeatedly observed and helps explain the beneficial therapeutic effect of iodide in GD (73). Ochi et al. demonstrated that chronic administration of iodine in GD blocks the stimulating effect on hormone release from both TSH and TSab (277). The block of hormone release that occurs in the thyroid of GD can be observed, although not uniformly, in the normal gland and in the normal gland made hyperactive by repeated administration of exogenous TSH. While early laboratory studies on the effect of iodine on the thyroid gland suggested that methimazole can inhibit somewhat the Wolff-Chaikoff effect (278-280), more recent clinical data indicates their ultimate impact on systemic thyroid hormone levels to be additive (281, 282). Clinical practice therefore favors this combination for treatment of severe cases of hyperthyroidism (278-280).

 

Extrathyroidal Processes

 

The pathogenesis of extrathyroidal complications of GD such as TED and dermopathy is described in detail in another chapter. Recent evidence demonstrates that stimulation of the TSab on the TSH-R in the orbital fibroblasts can lead to a crosstalk of this receptor with the insulin growth factor 1 receptor (IGF-1R) which stimulates hyaluronan accumulation in the orbital muscles and transformation of orbital fibroblasts into adipocytes (283, 284). This mechanism is believed to play a key role in the pathogenesis of TED and is the basis for emergent therapeutic alternatives in patients with TED and, maybe, dermopathy. Antibodies binding to and directly stimulating the IGF-1R have also been proposed to play a role in this process. However, IGF-1R antibodies exist in sera from about 10% of normal subjects, and in a similar percentage of GD patients, and are stable over time, strongly suggesting they have no unique role in GD (285).

 

PATHOLOGY

 

The ophthalmic and dermatologic changes seen in TED and dermopathy are described in the chapter on the Complications of GD. The thyroid gland changes seen in GD and changes in extrathyroidal organs that can be seen with any case of thyrotoxicosis are described below.

 

Thyroid Gland

 

The essential lesion of GD is parenchymatous hypertrophy and hyperplasia (Figure 5). The central features are increased height of the epithelium from cuboidal to columnar, and varying sizes and shapes of the follicles with reduced colloid content (286, 287). Papillary infoldings, cytologic evidence of increased activity, hypertrophy of the Golgi apparatus, increased number of mitochondria, and increased vacuolization of colloid are also seen. In addition, between the follicles, there is a large array of capillaries, together with a characteristic lymphocyte and plasma cell infiltrate. This infiltrate may be mild and diffuse throughout the gland, but more typically occurs as aggregates of mononuclear cells and even lymphoid germinal centers, referred to as focal thyroiditis. Occasionally the histologic pattern completely overlaps that of Hashimoto's thyroiditis. All pathological changes tend to regress when euthyroidism is achieved.

 

Figure 5. Histology of a thyroid gland of a patient with Graves’ disease. Columnar epithelium, vacuolization of colloid, and significant lymphocytic infiltrate are present in the setting of diffuse parenchymal hypertrophy.

 

Extrathyroidal Changes

 

MUSCLE

 

Patients with Graves’s disease can have diffuse degenerative atrophy, fatty infiltration, loss of striation, vacuolization, and proliferation or degeneration of nuclei in striated muscles (288-290). Skeletal muscles can also experience interstitial myositis characterized by plasma cells, tissue macrophages, and atrophy of fibers (291). This partly explains the sense of persistent weakness that patients describe after the normalization of thyroid hormone levels. While cardiac and smooth muscles tend to be spared in most patients, myocardial degenerative lesions have been reported in thyrotoxicosis, with foci of cell necrosis, mononuclear infiltrates, and mucopolysaccharide deposits similar to those described in extraocular and skeletal muscles (290).

 

The extraocular muscle changes are specific for TED, whereas the remainder of the abnormalities may reflect the action of excess hormone.

 

PITUITARY

 

The anterior pituitary demonstrates a dramatic decrease in identifiable thyrotropin containing cells, based on data from patients who died from thyroid storm (292, 293). This is entirely reversed in patients who achieve euthyroidism after treatment.

 

LIVER

 

Initial studies based on autopsies of patients with GD suggested focal and even diffuse liver cell necrosis, atrophy, and cirrhosis (269). In more contemporary series of liver biopsy specimens obtained from thyrotoxic persons, the deviations from normal were minimal (294, 295). Moderate decrease in glycogen content and increase in fat and round cell infiltrates were noted. The differences among these studies could be explained by a lesser severity and duration of the disease in more recent series with patients diagnosed earlier and treated effectively, as the disease has now an extremely low mortality rate (296). Recent studies have documented that thyroid hormone analogs specific for the thyroid hormone receptor b such as resmetirom may be a novel therapeutic approach for the therapy of metabolic dysfunction associated steatotic liver disease (MASLD) (297).

 

BONE

 

Prolonged hyperthyroidism is known to produce the histologic picture of osteoporosis (298), but osteitis fibrosa also occurs (299). Histomorphometry studies show unmistakable evidence of excess bone formation and resorption.

 

NATURAL COURSE OF GD AND CLINICAL MANIFESTATIONS OF THYROTOXICOSIS

 

In the classic presentation, the most common onset is the simultaneous and gradual development, over a period of weeks or months, of thyrotoxicosis-related signs and symptoms, goiter, and exophthalmos. However, GD displays an array of possible clinical patterns. It is possible for GD to develop in a patient with preceding nontoxic multinodular goiter, toxic multinodular goiter (Marine-Lenhart syndrome), to experience thyrotoxicosis without TED, or to have TED preceding the development of thyrotoxicosis. Due to the coexistence of TSab and TBAb some patients can first develop hypothyroidism and later thyrotoxicosis, or vice versa (232). In addition, during pregnancy GD tends to be partially suppressed, only to have a rapid recurrence in the post-partum period (300). Sometimes, human chorionic gonadotrophin (hCG) induced thyrotoxicosis, seen in the first part of pregnancy, can also coexist with GD and mask its presentation (301).

 

Before the general availability of current therapies, hyperthyroidism tended to evolve through periods of exacerbation and remission. In the mild forms, the active disease was self-limited to one year or more, and the patients returned spontaneously to a euthyroid state. In untreated moderate to severe forms, mortality could be observed in 11% of patients (302, 303). Mortality was most frequently attributed to cardiovascular complications (such as myocardial infarction, arrhythmia, or heart failure), or infections, and occurred within the first 4 years after diagnosis. Fortunately, death due to hyperthyroidism is now rare with an excess mortality of 1.2% compared to controls (296). Nowadays, after a period of anti-thyroid drug therapy (ATD) therapy there is re-establishment of normal thyroid homeostasis and over time a sizable proportion of patients can achieve disease remission, allowing them to discontinue ATD and remain euthyroid off therapy.

 

The most common presenting symptoms are weight loss, weakness, dyspnea, palpitations, increased thirst or appetite, diarrhea, irritability, profuse diaphoresis, heat intolerance and increased tolerance to cold, or tremor. Occasionally, exophthalmos or diplopia is the index symptom, but goiter may antedate all other manifestations. The nutritional state varies greatly. In the past, patients were severely emaciated, but these days, on average, the weight loss is 5 - 20 lbs. (2.3 – 9 kg). Facial expressions of flushing, fright, or extreme anxiousness are common. Notably, in elderly patients the disease can manifest as apathetic thyrotoxicosis, in which there is absence of hyperkinetic neuromuscular symptoms, and predominance of cardiac (e.g. arrhythmia), psychiatric (e.g. apathy, depression) and nutritional (e.g.., weight loss) symptoms (304).

 

In patients with GD, the ocular changes of TED, lymphoid hyperplasia, localized abnormalities of skin and connective tissue (e.g., dermopathy and acropachy) and the goiter itself are direct results of the autoimmune processes of GD. The remainder of the changes are entirely attributable to an excess of thyroid hormone.

 

The natural course and manifestations of thyroid storm, TED, and dermopathy are primarily discussed in the chapter on Complications of GD. The clinical manifestations of thyrotoxicosis are described below.

 

Thyroid

 

The diffuse toxic goiter is usually more or less symmetric. The size is related, but not closely, to the severity of the disease. It varies from the barely palpable normal (10- 15 g) to a three-to-six times enlargement (45-100 g) or, rarely, even more. The gland might not be palpable in 1% of cases, either because the thyroid is smaller than usual at baseline or because it is beneath the manubrium. The thyroid may be smooth, lobulated, or rarely nodular. In thyrotoxicosis associated with nodular goiters, the hyperfunctioning tissue may reside between the nodules (305). Usually, the consistency is firm but elastic, or very firm if iodide has been given. The borders are easily demarcated by palpation. Thrills and bruits, usually denote the hypervascularity associated with increased function. Bruits may be continuous or systolic and are usually audible over the entire thyroid. Local pressure symptoms, including dysphagia and the sensation of a lump in the neck, are produced by the enlarged goiter. Vocal cord palsy is only rarely seen in GD (306). In addition, the supraclavicular lymph nodes could become enlarged and, rarely, tender (307). Most thyroid related manifestations tend to regress or disappear with restoration of euthyroidism.

 

Skin

 

Cutaneous manifestations are nearly always present when vasomotor overactivity is significant. Heat intolerance and profuse diaphoresis occur under circumstances that would provoke no response in normal people. On palpation, hands are usually erythematous, hot, and moist (hyperhidrosis). There may be continuous erythema of the face and neck, with superimposed transient blushing after palpation of the thyroid. Occasionally diffuse pruritus or urticaria occur (308). Patchy vitiligo is an associated manifestation of autoimmunity directed toward melanocytes that can be found in 7% of patients with GD.

 

Fingernails can experience onycholysis characterized by ragged appearance, thinning and posterior erosion of the hyponychium. The free margin of the nail leaves the nail bed, producing a concave or wavy margin at the line of contact. Temporary thinning of the hair is common, but alopecia is rare. Hair loss can occur due to marked changes in metabolism throughout the course of the disease.

 

Myxedema can be seen in 0.5-4.3% of patients with GD, and 13% of patients with TED (309, 310). The clinical characteristics of thyroid dermopathy are described in the chapter on Complications of GD.

 

Nervous System

 

Neural and mental findings are diverse. Nervousness, irritability, anxiety, and restlessness are common (Table 3) (311). The behavioral reactions to all sorts of stimuli are typically exaggerated. When asked to sit up, the patient might jump into an upright position. They may simply wish to cooperate but appear to overdo it. Often emotional instability is combined with this pattern, to the point of a notable change in personality. In some patients, the emotional pattern is that of mania or euphoria. In others, hyperactivity produces a state of exhaustion, and profound fatigue or asthenia. The mind is often highly active, and the patient is troubled with insomnia. Rarely, patients develop visual or auditory hallucinations or a frank psychosis. The latter is probably the result on an exacerbation of a baseline psychiatric condition rather than a de novo process (312). Furthermore, impairment of intellectual function has been found in patients with untreated hyperthyroidism. A recent study has documented that in patients over 65 years, a low TSH level from either endogenous or exogenous thyrotoxicosis is associated with higher risk of incident cognitive disorders (313).

 

A fine, rapid tremor of the outstretched fingers is classically found, and a generalized tremulousness, also involving the tongue, may be evident. The speed of muscle contraction and relaxation is increased, tendon reflexes tend to be brisk, and the reflex relaxation time is shortened (314). The tremor of Parkinsonism is intensified during thyrotoxicosis, and manifestations of cerebellar disease or pyramidal tract lesions can be seen (315, 316). Muscle fibrillations are not a usual part of the syndrome, but they may occur in chronic thyrotoxic myopathy. Polyneuropathy has also been reported (317). Rarely, patients manifest disorientation, aphasia, grimacing, choreoathetoid movements, symptoms suggestive of encephalitis, episodes of hemiparesis or bulbar paralysis (318). These symptoms clear up completely after restoration of an euthyroid state.

 

Other possible, although rare, severe neurologic manifestation consist of the new onset, exacerbation, or refractoriness of seizure disorders (319). Electroencephalography reveals increased fast wave activity, and occasionally bursts of tall spike waves. In addition, several reports describe a severe, steroid-responsive encephalopathy in some patients with GD (320).

 

Table 3. Neurologic Manifestations in Graves' Disease

Common:

·       Nervousness

·       Irritability

·       Restlessness

·       Anxiety

·       Emotional lability including hypomania or euphoria, and fatigue or asthenia).

·       Insomnia

·       Tremors

·       Brisk tendon reflexes with short relaxation time

 

Uncommon:

·       Visual or auditory hallucinations

·       Psychosis

·       Impairment of intellectual function

·       Disorientation

·       Aphasia

·       Choreoathetoid movements

·       Hemiparesis or bulbar paralysis

·       Seizures

·       Encephalopathy

·       Neuropathy

 

Muscular System

 

The muscular symptoms vary from mild myasthenia to profound muscular weakness and atrophy, especially of the proximal muscle groups. Wasting of the temporals and interossei used to be noted in a considerable number of patients while a few had generalized muscle wasting in the decades prior to current diagnostic and therapeutic abilities. Occasionally the myopathy may shade into the picture of polymyositis. Muscle cell necrosis and lymphocyte infiltration may be visible histologically, but usually are not found even when the symptoms of weakness are severe (321, 322). The electromyogram is normal in most instances but may occasionally resemble that of muscular dystrophy (323).

 

Work efficiency, measured in terms of the calories of heat produced while performing a given amount of work, has been reported to be either normal or decreased (324, 325). The muscles have decreased ability to incorporate creatine from the blood (326, 327). Creatinine excretion is initially increased by the general catabolism of hyperthyroidism, but as muscle mass diminishes, creatinine excretion in the urine decreases.

 

Myasthenia gravis may simulate thyrotoxicosis, and vice versa (328). The close relationship between these two diseases is apparent in the observation that thyrotoxicosis occurs in 3% of patients with myasthenia gravis. The pathogenic anti-acetylcholine receptor antibodies that occur in myasthenia gravis are clearly comparable to the TSH-R antibodies found in GD.

 

Periodic paralysis is precipitated and worsened by thyrotoxicosis (329). It has been more commonly reported in Southeast Asian males who have a higher frequency of certain genetic susceptibilities associated with higher risk of thyrotoxic periodic paralysis (330-333). Paralysis is usually associated with hypokalemia. Inactivating mutations in the inwardly rectifying potassium channel 2.6 encoded by the KCNJ18 gene (in about 30%) have, among other candidates, been associated with susceptibility for developing thyrotoxic periodic paralysis (334, 335). Beta adrenergic stimulation mediated by thyrotoxicosis augments sodium-potassium ATPase activity in the skeletal muscle leading to increased potassium uptake into the cells (336). The episodes of paralysis tend to be infrequent and sporadic, but most commonly occur after a meal, following exercise, or during sleep, and can be induced by administration of glucose and insulin. Episodes last from minutes to hours, usually involving peripheral muscles, but can cause paralysis of the diaphragm and affect the heart. Serious episodes can be associated with extensive muscle cell damage and necrosis, and electrocardiographic (EKG) abnormalities associated with hypokalemia, such as ST and T wave changes, premature ventricular complexes (PVCs), first degree heart block, prolonged QT intervals, and even ventricular fibrillation (337).

 

Pulmonary System

 

Except for dyspnea on exertion, symptoms deriving from the lungs are not prominent. Nevertheless, pulmonary function tests can show some reduction in vital capacity, expiratory reserve volume, pulmonary compliance, and airway resistance (338, 339). Minute volume response to exercise is excessive for the amount of oxygen consumed (340).

 

Cardiovascular System

 

The first and most common manifestations deriving from the cardiovascular system are palpitations and tachycardia (341, 342). The pulse on palpation is rapid and bounding. When present, it can be effectively controlled with beta- adrenergic blockers (343, 344). PVCs, paroxysmal atrial tachycardia, atrial fibrillation, and shorter P wave duration occur in 6-12% of patients (345-347). When present, atrial fibrillation should be treated with anticoagulation to prevent risk of thromboembolic complications. The systolic blood pressure is frequently elevated. The diastolic blood pressure is characteristically decreased, making the pulse pressure elevated to between 50-80 mm Hg (348). A systolic murmur can sometimes be heard over the precordium due to development of mitral valve prolapse following papillary muscle dysfunction during thyrotoxicosis (349). However, mitral valve insufficiency is usually not clinically relevant and can revert to normal with treatment of thyrotoxicosis.

 

In thyrotoxicosis, the heart rate, stroke volume, left ventricular mass index, and cardiac output are all increased. The pre-ejection period is shortened, and the left ventricular ejection time remains relatively normal. The interval from initiation of the QRS complex to arrival of the arterial pulse in the brachial artery is reduced (350). Circulation time is decreased. There is dilatation of superficial capillaries and decline in systemic vascular resistance (351). Coronary blood flow and myocardial oxygen consumption in each stroke are increased. Circulating plasma volume is increased (352). Long-term mild excess of thyroid hormone causes impaired cardiac reserve and exercise capacity (353). Cardiac enlargement and heart failure may occur with or without prior heart disease (354). These effects tend to normalize when euthyroidism is restored (355).

 

Patients with coronary artery disease often develop angina during thyrotoxicosis. However, angina could also develop de novo in patients with normal coronary arteries, especially young females. This condition has been ascribed to an imbalance between increased cardiac work and blood supply, even with a patent vessel (356). Severe coronary vasospasm has been observed during angiography in patients with GD (357). Myocardial damage can occur in thyrotoxic patients with congestive heart failure (358), even when coronary vessels are normal at baseline (359).

 

It has been suggested that the changes in the cardiovascular system are secondary to increased demand for metabolites and to increased heat production. Dilatation of superficial capillaries for the dissipation of heat does cause increased blood flow and cardiac output (360). However, the direct action of thyroid hormone on the heart is also increased, since the sinus node has higher intrinsic activity, the isolated thyrotoxic heart beats faster than normal, and isolated papillary muscle from a thyrotoxic heart has a shortened contraction time (361-363).

 

Historically, the cardiovascular effects of triiodothyronine have primarily been attributed to its modulatory influence on nuclear adrenergic receptor genes, among others (364, 365). However, more recent evidence has demonstrated a significant physiological role for the non-genomic effects of thyroid hormone, particularly in energy homeostasis (366-368). Cardiac muscle contractility is enhanced by a reduction in alpha-adrenergic and cholinergic receptors within the heart. This effect is coupled with an increase in beta-adrenergic receptors, heightened adenosine transport, and enhanced phosphorylation within myocardial cells. Additionally, there is an augmentation in cardiac Na+-K+ activated membrane ATPase activity, as well as an elevation in sarcoplasmic reticulum Ca++-activated ATPase activity. Finally, the increased synthesis of alpha-myosin heavy chains with increased ATPase activity also contributes to this heightened contractility (363, 369-371).

 

Fatourechi and Edwards used myocardial biopsy to investigate the presence of an autoimmune process in eleven cases of GD with low output cardiac dysfunction. Two patients had lymphocytic infiltrates suggestive of an autoimmune process, whereas the others did not, indicating that myocardial autoimmunity may occur but would not be the usual cause of cardiac dysfunction in thyrotoxicosis (372).

 

Table 4. Cardiovascular Manifestations in Graves' Disease

Common:

·       Palpitations

·       Tachycardia

·       Paroxysmal atrial tachycardia

·       Atrial fibrillation

·       Increased stroke volume and cardiac output.

·       Increased coronary blood flow and myocardial oxygen consumption

·       Decreased exercise capacity

 

Uncommon:

·       Impaired cardiac reserve

·       Heart failure

·       Myocardial infarction

 

Hematologic and Lymphatic Systems

 

In most patients the hemoglobin and hematocrit are in the normal or low-normal range (373). The glucose-6-phosphate dehydrogenase activity of red cells is increased in thyrotoxicosis (374), while blood volume and the red cell mass are also increased in some patients. In the past, severe thyrotoxicosis used to be associated with normocytic anemia with hemoglobin concentrations as low as 8 g/dl, likely related to iron deficiency and malnutrition (375, 376). However, the presence of anemia in a thyrotoxic patient these days requires a search for an additional explanation, other than thyrotoxicosis.

 

The reticuloendothelial and lymphocytic systems undergo hyperplasia. There may be generalized lymphadenopathy, and the thymus may be enlarged (377). The thymus enlargement should be considered when mediastinal abnormalities are noted on chest imaging; thymus enlargement resolves with resolution of thyrotoxicosis and that should inform the decision about possible biopsy (378). Relative lymphocytosis and neutropenia with a normal or slightly low total white cell count, constitute the characteristic blood findings of GD (379). A relative and an absolute increase in the number of monocytes has also been reported (380). Significant pancytopenia with leukocyte counts under 3x109/l and neutrophiles under 2x109/l rarely occur, and if unrelated to drug therapy, tend to recover with restoration of euthyroidism (381).

 

GD is often associated with mild thrombocytopenia, and occasionally with idiopathic thrombocytopenic purpura (382). This co-occurrence is thought to reflect the autoimmune pathogenesis of both diseases. Fourteen percent of patients with immune thrombocytopenic purpura are reported to have coincident GD. Mild thrombocytopenia may disappear spontaneously or with treatment of hyperthyroidism, or if severe, may respond to glucocorticoid therapy (383). Bone marrow examination may show normal or increased megakaryocytes. In addition, platelet life span can be shortened due to a more rapid clearing by the activated reticulo-endothelial system.

 

Usually, thyrotoxicosis results in a mild hypercoagulable state. Although rare, cerebral venous thrombosis has been reported in association with thyrotoxicosis, suggesting that occasionally the propensity for coagulation can lead to profound consequences (384). Mild prolongation of the prothrombin time and elevation of several coagulation factors (Factors VIII, XIII, IX, XI, Von Willebrand, fibrinogen, and plasminogen activator inhibitor 1, among others) are often seen with thyrotoxicosis, and return to normal with treatment. In addition, recent studies emphasize the role of thyroid hormones in promoting coagulation through non-genomic mechanisms involving platelet activation with subsequent endothelial interactions (385).  

 

Gastrointestinal System

 

The appetite and the gastrointestinal track absorption are characteristically increased to try to offset nutritional requirements from the increase in catabolism seen in patients with thyrotoxicosis. Despite this, weight loss is usually predominant. In severe thyrotoxicosis, nausea, emesis, and abdominal pain can be present while intestinal transit time is decreased, and occasionally diarrhea occurs (386). Steatorrhea can be seen if fat intake is excessive. Achlorhydria can be as prevalent as 40% (387, 388). Gastric enzymes production is decreased, and a mild gastritis can be present (389). Fasting serum gastrin levels, and their responses to arginine, are increased (390).

 

Around 55% of patients with thyrotoxicosis can experience at least one abnormality in their liver blood tests. Alkaline phosphatase elevation is the most common, but other common abnormalities include hypoalbuminemia, mild prothrombin time elevation, elevated aminotransferases, hyperbilirubinemia, and elevated lactate dehydrogenase (LDH) (391). Mild to severe liver disease may be found, and the liver is frequently palpable regardless of the coexistence of heart failure (392). Jaundice is possible but often seen when there is significant cholestasis from severe thyrotoxicosis (393, 394). The cause of hepatic disease has been thought to be multifactorial in the setting of congestive hepatopathy from heart failure, malnutrition, previous or concomitant liver disease (i.e., infectious or autoimmune disease), and drug related liver injury, and possible liver ischemia from a mismatch between oxygen consumption and oxygen delivery to the liver (395-399). All gastrointestinal manifestations tend to improve with restauration of the euthyroid state.

 

Renal System

 

Polyuria and occasionally glucosuria are seen in uncomplicated thyrotoxicosis. Polyuria does not indicate insensitivity to vasopressin, for the kidney responds normally to vasopressin with an increase in concentration of urine (400). Glucosuria may reflect accelerated absorption of sugar from the intestine and glucose intolerance. The glomerular filtration rate and renal blood flow are on average increased, probably secondary to increased cardiac output and a direct effect of thyroid hormone on renal function (401). Hyperuricemia with hypercalcemia can be seen in severe thyrotoxicosis, but it rarely injures the kidneys. In addition, occasionally hyposthenuria and uremia occur (402, 403).

 

Female Reproductive System

 

Menstruation is characteristically decreased in volume, and rarely amenorrhea with a proliferative endometrium can happen. The menstrual cycle may be either shortened or prolonged. The relative importance of a primary action of excess thyroid hormone on the gonadotroph function is unclear. However, an alteration in pituitary LH production and subsequent ovulation is suggested (404). Hyperprolactinemia is more common in patients with hyperthyroidism than in healthy controls (405). Premature ovarian failure can occur in association with GD in patients with polyglandular autoimmune syndrome type 3 (222).

 

Fertility is decreased, but pregnancy can develop. The incidence of miscarriage, premature delivery, pre-eclampsia, and gestational heart failure are increased by maternal thyrotoxicosis (406-408). Reduced fertility and increased miscarriage rates are associated with autoimmune thyroid disease (AITD) and positive antibodies. High maternal thyroid hormone levels and/or high titer of TSab (by crossing the placenta in the 3rd trimester) can lead to fetal thyrotoxicosis with suppressed fetal TSH, lower fetal weight, and fetal death (409). However, pregnancy often ameliorates the biochemistry and the symptoms of thyrotoxicosis due to GD. Unfortunately, relapse is prone to occur in the 3-4 months following delivery.

 

Male Reproductive System

 

Peripheral conversion of testosterone and androstenedione to estrone and estradiol is increased in both sexes during hyperthyroidism (293, 410). Thus, men can have elevated circulating levels of free estradiol (411, 412), and some might experience gynecomastia with ductal elongation and epithelial hyperplasia (413, 414). In addition, the slightly elevated LH in men with gynecomastia suggests hypothalamic insensitivity to feedback control and some peripheral unresponsiveness to LH (411).

 

 

Both Leydig cell and spermatogenic abnormalities may be present. Previous small series have described a 71% rate of loss of libido, 56% rate of erectile dysfunction, and 80% rate of low sperm counts (<40 millions) in thyrotoxic men (415, 416). In these patients, the total testosterone level was elevated, but because the sex hormone binding globulin level was also high, the free testosterone level was reduced and the response to hCG was blunted. The abnormalities normalize when the patients become euthyroid (417). In addition, RAI therapy can cause transient reductions in both sperm count and motility but do not seem to cause permanent effects with ordinary treatment doses (under 14 mCi, equivalent to around 500 MBq) (417).

 

Adrenal Function

 

There are no obvious clinical signs or symptoms of altered adrenal cortical function in thyrotoxicosis, but distinct biochemical changes have been detected. In thyrotoxicosis the adrenal cortex is often hyperplastic and exogenous glucocorticoids are cleared from the plasma and metabolized at an accelerated rate (418). Since plasma glucocorticoid levels are normal and their rate of metabolism is increased, total daily metabolism and excretion of 17-ketosteroids and 17 hydroxy-corticoids are usually increased (419, 420). Furthermore, there is a relatively increased excretion of 11-oxycorticoid metabolites (421), which are biologically inactive compounds. There is increased secretion of adrenocorticotropic hormone (ACTH) by the pituitary, and subsequently increased production of steroids by the adrenal gland in order to maintain a normal concentration of active steroids in the peripheral blood and in the tissues (422, 423). However, a reduced response to exogenous ACTH suggests that adrenal reserve is reduced (424), and it has been hypothesized that in severe thyrotoxicosis and in thyroid storm there may be an element of adrenal insufficiency.

 

An increase in the 5-alpha metabolite of testosterone (androsterone) and a relative decrease in the 5-beta metabolite (etiocholanolone) are seen in the urine of thyrotoxic patients (425, 426). Because administration of substantial amounts of androsterone depresses the level of serum lipids, Hellman et al. have hypothesized that this change in steroid metabolism may be a way in which thyroid hormone affects lipid metabolism.

 

Skeletal System

 

Patients with mild thyrotoxicosis can experience some degree of bone mass loss and increased fracture risk, irrespective of age or sex (427-429). Those with thyrotoxicosis extending over several years may develop severe osteoporosis (430, 431). Skeletal mass is augmented after restauration of euthyroidism (428, 429).

 

The thyroid hormone receptors α1 (TRα1), TRβ1, and TRβ2 are expressed in human osteoblasts and bone marrow stromal cells. Histomorphometry evaluations with tetracycline labelling demonstrates accelerated turnover of bone, calcium, and collagen, both in spontaneous hyperthyroidism and in female treated with excess thyroid hormone (298, 432, 433). Serum osteocalcin, carboxy-terminal-1-telopeptide, and alkaline phosphatase may increase in parallel with hormone levels (434-437). These tend to normalize with restauration of euthyroidism.

 

The serum calcium level is usually normal but may be sufficiently elevated to produce nausea and emesis (402), and rarely, renal injury (403, 438). Fecal and urinary calcium excretion is greatly augmented, but kidney stones are infrequent since there is a concomitant polyuria with increase in excretion of colloids that stabilize the calcium (433). Intestinal absorption of calcium is usually reduced (439). The hypercalcemia appears to be a direct manifestation of thyroid hormone action on bone metabolism and can lead to secondary hypoparathyroidism with reduced 1,25-dihydroxyvitamin D (440, 441), and elevated serum phosphorus secondary to increased renal resorption (438, 442). It has been proposed that the hypercalcemia can usually be corrected partially or totally by the administration of glucocorticoids, but data is inconclusive (442, 443). Treatment of thyrotoxicosis is certainly able to normalize these abnormalities (444).

 

Metabolism

 

METABOLIC RATE

 

The basal oxygen consumption in thyrotoxicosis, as measured by the basal metabolic rate (BMR), is elevated compared to a euthyroid person. In extreme thyrotoxicosis, the BMR may be double the standard (445-447). In addition, the total metabolic rate, which is the BMR plus the increments from work, food, or stress, is elevated.

 

Observations regarding energy expenditure are mixed. An increased cost of muscular work with less efficient coupling of oxidation and energy use in patients with thyrotoxicosis was reported many years ago by Plummer and Boothby (448)and Briard et al. (449) among others. However, recent studies suggest that the increase in energy expenditure caused by work is not altered in thyrotoxicosis (450).

 

CARBOHYDRATE METABOLISM

 

Intestinal carbohydrate absorption is accelerated, as is its removal from the plasma. After a standard oral glucose load is given, the thyrotoxic patient characteristically has an early and rapid rise in blood glucose concentration in 30 - 60 minutes followed by a rapid fall, so that by two hours the concentration is normal (451-453). In non-diabetic thyrotoxic patients there is increased insulin demand and there could be some resistance to the action of insulin. Fasting blood glucose levels are associated with double the normal insulin concentration (454), and insulin resistance has been found in vitro utilizing adipocytes from patients with untreated hyperthyroidism (455).

 

Diabetes may develop (in patients with prediabetes) or worsen with the development of thyrotoxicosis and is ameliorated or may disappear when thyrotoxicosis is treated (456). In these patients, there is increased basal hepatic glucose production and reduced ability of insulin and glucose to suppress hepatic glucose production, another marker of insulin resistance (457, 458).

 

LIPID METABOLISM

 

Hypocholesterolemia is associated with thyrotoxicosis. It may be produced without a distinct decrease in total body or liver cholesterol. Part of the cholesterol-lowering action of thyroid hormone relates to malnutrition and weight loss, and part may be simply a manifestation of hypermetabolism. There is an increase both in synthesis and degradation of cholesterol, but the balance results in lower steady-state concentrations in the serum (459-469). Thyroid hormone directly enhances conversion of cholesterol to bile acids and their excretion in the bile, disposing of 70-90% of the cholesterol formed in the body (462). They may also affect cholesterol metabolism by directly increasing the number of membrane surface low-density lipoprotein (LDL) receptors (463). Furthermore, hepatic lipogenesis is also strikingly increased, both by direct action of thyroid hormones and in response to increased insulin levels. Overall, the levels of LDL, HDL, and apolipoproteins are lowered (461, 465-470). Triglyceride levels tend to be normal or slightly elevated (464), and the clearance rate of infused triglycerides might be elevated (461, 465-470). Plasma leptin levels are normal (468), and non-esterified fatty acids are elevated (471).

 

PROTEIN METABOLISM

 

In thyrotoxicosis, protein formation and breakdown are both accelerated. Despite the increased protein turnover, there is an overall protein deficit (472, 473). Nitrogen excretion is increased, and nitrogen balance may be normal or negative, depending on whether intake meets the demands of increased catabolism. Catabolism of collagen is increased, and urinary hydroxyproline excretion is characteristically increased (474). While the exact mechanism behind the effect of thyroid hormone on protein metabolism is not fully elucidated, it is believed to be mediated by a combination of mitochondria-dependent cytoplasmic mechanisms, and modulation of nuclear genetic transcription of genes involved in protein metabolism (475-477).

 

VITAMIN METABOLISM

 

The absorption of vitamin A is increased and conversion of carotene to vitamin A is accelerated in thyrotoxicosis (478). The requirements of the body are likewise increased, and low blood concentrations of vitamin A may be found. In addition, requirements for thiamine and vitamin B6 are increased (479).

 

ACKNOWLEDGEMENT

 

Sincere gratitude is extended to Dr. Leslie DeGroot, the author of the previous version, for laying the foundation of this chapter and his enduring influence on the subject.

 

REFERENCES

 

  1. Graves RJ. Clinical Lectures: Newly Observed Affection of the Thyroid Gland in Females. London Medical and Surgical Journal. 1835;7.
  2. der KP, Houtstra-Lanz M, Schwarz F. Exophthalmos-producing substance in human serum. J Clin Endocrinol Metab. 1960;20:712-8.
  3. Dobyns BM, Steelman SL. The thyroid-stimulating hormone of the anterior pituitary as distinct from the exophthalmos-producing substance. Endocrinology. 1953;52(6):705-11.
  4. Adams DD. The presence of an abnormal thyroid-stimulating hormone in the serum of some thyrotoxic patients. J Clin Endocrinol Metab. 1958;18(7):699-712.
  5. Adams DD, Fastier FN, Howie JB, Kennedy TH, Kilpatrick JA, Stewart RD. Stimulation of the human thyroid by infusions of plasma containing LATS protector. J Clin Endocrinol Metab. 1974;39(5):826-32.
  6. Smith BR, Hall R. Thyroid-stimulating immunoglobulins in Graves' disease. Lancet. 1974;2(7878):427-31.
  7. Huber GK, Safirstein R, Neufeld D, Davies TF. Thyrotropin receptor autoantibodies induce human thyroid cell growth and c-fos activation. J Clin Endocrinol Metab. 1991;72(5):1142-7.
  8. Cornelis S, Uttenweiler-Joseph S, Panneels V, Vassart G, Costagliola S. Purification and characterization of a soluble bioactive amino-terminal extracellular domain of the human thyrotropin receptor. Biochemistry. 2001;40(33):9860-9.
  9. Rees Smith B, McLachlan SM, Furmaniak J. Autoantibodies to the thyrotropin receptor. Endocr Rev. 1988;9(1):106-21.
  10. Ludgate ME, Vassart G. The thyrotropin receptor as a model to illustrate receptor and receptor antibody diseases. Baillieres Clin Endocrinol Metab. 1995;9(1):95-113.
  11. Shapira Y, Agmon-Levin N, Shoenfeld Y. Defining and analyzing geoepidemiology and human autoimmunity. Journal of Autoimmunity. 2010;34(3):J168-J77.
  12. McGrogan A, Seaman HE, Wright JW, De Vries CS. The incidence of autoimmune thyroid disease: a systematic review of the literature. Clinical Endocrinology. 2008;69(5):687-96.
  13. Taylor PN, Albrecht D, Scholz A, Gutierrez-Buey G, Lazarus JH, Dayan CM, et al. Global epidemiology of hyperthyroidism and hypothyroidism. Nat Rev Endocrinol. 2018;14(5):301-16.
  14. Vos XG, Smit N, Endert E, Brosschot JF, Tijssen JG, Wiersinga WM. Age and stress as determinants of the severity of hyperthyroidism caused by Graves' disease in newly diagnosed patients. Eur J Endocrinol. 2009;160(2):193-9.
  15. Boelaert K, Torlinska B, Holder RL, Franklyn JA. Older Subjects with Hyperthyroidism Present with a Paucity of Symptoms and Signs: A Large Cross-Sectional Study. The Journal of Clinical Endocrinology & Metabolism. 2010;95(6):2715-26.
  16. Smith TJ, Hegedüs L. Graves’ Disease. New England Journal of Medicine. 2016;375(16):1552-65.
  17. Furszyfer J, Kurland LT, McConahey WM, Elveback LR. Graves' disease in Olmsted County, Minnesota, 1935 through 1967. Mayo Clin Proc. 1970;45(9):636-44.
  18. Tunbridge WM, Evered DC, Hall R, Appleton D, Brewis M, Clark F, et al. The spectrum of thyroid disease in a community: the Whickham survey. Clin Endocrinol (Oxf). 1977;7(6):481-93.
  19. Vanderpump MP, Tunbridge WM, French JM, Appleton D, Bates D, Clark F, et al. The incidence of thyroid disorders in the community: a twenty-year follow-up of the Whickham Survey. Clin Endocrinol (Oxf). 1995;43(1):55-68.
  20. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, et al. Serum TSH, T4, and Thyroid Antibodies in the United States Population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). The Journal of Clinical Endocrinology & Metabolism. 2002;87(2):489-99.
  21. Degroot LJ, Quintans J. The Causes of Autoimmune Thyroid Disease*. Endocrine Reviews. 1989;10(4):537-62.
  22. Desai MK, Brinton RD. Autoimmune Disease in Women: Endocrine Transition and Risk Across the Lifespan. Frontiers in Endocrinology. 2019;10.
  23. Kisiel B, Bednarczuk T, Kostrzewa G, Kosińska J, Miśkiewicz P, Płazińska MT, et al. Polymorphism of the oestrogen receptor beta gene (ESR2) is associated with susceptibility to Graves' disease. Clin Endocrinol (Oxf). 2008;68(3):429-34.
  24. Merrill SJ, Mu Y. Thyroid autoimmunity as a window to autoimmunity: An explanation for sex differences in the prevalence of thyroid autoimmunity. J Theor Biol. 2015;375:95-100.
  25. Struja T, Kutz A, Fischli S, Meier C, Mueller B, Recher M, et al. Is Graves' disease a primary immunodeficiency? New immunological perspectives on an endocrine disease. BMC Med. 2017;15(1):174.
  26. Ando T, Imaizumi M, Graves PN, Unger P, Davies TF. Intrathyroidal fetal microchimerism in Graves' disease. J Clin Endocrinol Metab. 2002;87(7):3315-20.
  27. Vestergaard P. Smoking and thyroid disorders--a meta-analysis. Eur J Endocrinol. 2002;146(2):153-61.
  28. Wiersinga WM. Smoking and thyroid. Clin Endocrinol (Oxf). 2013;79(2):145-51.
  29. Prummel MF, Strieder T, Wiersinga WM. The environment and autoimmune thyroid diseases. Eur J Endocrinol. 2004;150(5):605-18.
  30. Rodríguez-Paredes M, Esteller M. Cancer epigenetics reaches mainstream oncology. Nat Med. 2011;17(3):330-9.
  31. Wang H, Liao H, Ochani M, Justiniani M, Lin X, Yang L, et al. Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nat Med. 2004;10(11):1216-21.
  32. Effraimidis G, Wiersinga WM. Mechanisms in endocrinology: autoimmune thyroid disease: old and new players. Eur J Endocrinol. 2014;170(6):R241-52.
  33. Holm IA, Manson JE, Michels KB, Alexander EK, Willett WC, Utiger RD. Smoking and other lifestyle factors and the risk of Graves' hyperthyroidism. Arch Intern Med. 2005;165(14):1606-11.
  34. Wenzel BE, Franke TF, Heufelder AE, Heesemann J. Autoimmune thyroid diseases and enteropathogenic Yersinia enterocolitica. Autoimmunity. 1990;7(4):295-303.
  35. Wolf MW, Misaki T, Bech K, Tvede M, Silva JE, Ingbar SH. Immunoglobulins of patients recovering from Yersinia enterocolitica infections exhibit Graves' disease-like activity in human thyroid membranes. Thyroid. 1991;1(4):315-20.
  36. Wenzel BE, Heesemann J, Wenzel KW, Scriba PC. Antibodies to plasmid-encoded proteins of enteropathogenic Yersinia in patients with autoimmune thyroid disease. Lancet. 1988;1(8575-6):56.
  37. Akamine H, Takasu N, Komiya I, Ishikawa K, Shinjyo T, Nakachi K, et al. Association of HTLV-I with autoimmune thyroiditis in patients with adult T-cell leukaemia (ATL) and in HTLV-I carriers. Clin Endocrinol (Oxf). 1996;45(4):461-6.
  38. de Luis DA, Varela C, de La Calle H, Cantón R, de Argila CM, San Roman AL, et al. Helicobacter pylori infection is markedly increased in patients with autoimmune atrophic thyroiditis. J Clin Gastroenterol. 1998;26(4):259-63.
  39. Rochman H, deGroot LJ, Rieger CH, Varnavides LA, Refetoff S, Joung JI, et al. Carcinoembryonic antigen and humoral antibody response in patients with thyroid carcinoma. Cancer Res. 1975;35(10):2689-92.
  40. Hancock SL, Cox RS, McDougall IR. Thyroid diseases after treatment of Hodgkin's disease. N Engl J Med. 1991;325(9):599-605.
  41. Vermiglio F, Castagna MG, Volnova E, Lo Presti VP, Moleti M, Violi MA, et al. Post-Chernobyl increased prevalence of humoral thyroid autoimmunity in children and adolescents from a moderately iodine-deficient area in Russia. Thyroid. 1999;9(8):781-6.
  42. Monzani F, Del Guerra P, Caraccio N, Casolaro A, Lippolis PV, Goletti O. Appearance of Graves' disease after percutaneous ethanol injection for the treatment of hyperfunctioning thyroid adenoma. J Endocrinol Invest. 1997;20(5):294-8.
  43. Nygaard B, Knudsen JH, Hegedüs L, Scient AV, Hansen JE. Thyrotropin receptor antibodies and Graves' disease, a side-effect of 131I treatment in patients with nontoxic goiter. J Clin Endocrinol Metab. 1997;82(9):2926-30.
  44. Schmidt M, Gorbauch E, Dietlein M, Faust M, Stützer H, Eschner W, et al. Incidence of postradioiodine immunogenic hyperthyroidism/Graves' disease in relation to a temporary increase in thyrotropin receptor antibodies after radioiodine therapy for autonomous thyroid disease. Thyroid. 2006;16(3):281-8.
  45. Peng K, Li X, Yang D, Chan SCW, Zhou J, Wan EYF, et al. Risk of autoimmune diseases following COVID-19 and the potential protective effect from vaccination: a population-based cohort study. EClinicalMedicine. 2023;63:102154.
  46. Muller I, Consonni D, Crivicich E, Di Marco F, Currò N, Salvi M. Increased risk of Thyroid Eye Disease following Covid-19 Vaccination. J Clin Endocrinol Metab. 2023.
  47. Rönnblom LE, Alm GV, Oberg KE. Autoimmunity after alpha-interferon therapy for malignant carcinoid tumors. Ann Intern Med. 1991;115(3):178-83.
  48. Berthaud P, Schlumberger M, Comoy E, Avril MF, Le Chevalier T, Spielmann M, et al. Hypothyroidism and goiter during interleukin-2 therapy. J Endocrinol Invest. 1990;13(8):689-90.
  49. Hoekman K, von Blomberg-van der Flier BM, Wagstaff J, Drexhage HA, Pinedo HM. Reversible thyroid dysfunction during treatment with GM-CSF. Lancet. 1991;338(8766):541-2.
  50. Pariani N, Willis M, Muller I, Healy S, Nasser T, McGowan A, et al. Alemtuzumab-Induced Thyroid Dysfunction Exhibits Distinctive Clinical and Immunological Features. The Journal of Clinical Endocrinology & Metabolism. 2018;103(8):3010-8.
  51. Brix TH, Christensen K, Holm NV, Harvald B, Hegedüs L. A population-based study of Graves' disease in Danish twins. Clin Endocrinol (Oxf). 1998;48(4):397-400.
  52. Coles AJ, Wing M, Smith S, Coraddu F, Greer S, Taylor C, et al. Pulsed monoclonal antibody treatment and autoimmune thyroid disease in multiple sclerosis. Lancet. 1999;354(9191):1691-5.
  53. 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;95(2):953-62.
  54. Chen Q. The expression of interleukin-15 and interleukin-17 in tears and orbital tissues of Graves ophthalmopathy patients. Journal of Cellular Biochemistry. 2019;120(4):6299-303.
  55. Yao Q, Wang B, Jia X, Li Q, Yao W, Zhang J-a. Increased Human Interleukin-32 Expression Is Related to Disease Activity of Graves' Disease. Frontiers in Endocrinology. 2019;10.
  56. Prummel MF, Laurberg P. Interferon-alpha and autoimmune thyroid disease. Thyroid. 2003;13(6):547-51.
  57. Durelli L, Ferrero B, Oggero A, Verdun E, Ghezzi A, Montanari E, et al. Thyroid function and autoimmunity during interferon beta-1b treatment: a multicenter prospective study. J Clin Endocrinol Metab. 2001;86(8):3525-32.
  58. Kristan MM, Toro-Tobon D, Francis N, Desale S, Bikas A, Jonklaas J, et al. Immunotherapy-Associated Hypothyroidism: Comparison of the Pre-Existing With De-Novo Hypothyroidism. Frontiers in Endocrinology. 2022;13.
  59. Borodic G, Hinkle DM, Cia Y. Drug-induced graves disease from CTLA-4 receptor suppression. Ophthalmic Plast Reconstr Surg. 2011;27(4):e87-8.
  60. Ryder M, Callahan M, Postow MA, Wolchok J, Fagin JA. Endocrine-related adverse events following ipilimumab in patients with advanced melanoma: a comprehensive retrospective review from a single institution. Endocr Relat Cancer. 2014;21(2):371-81.
  61. Vagenakis AG, Wang CA, Burger A, Maloof F, Braverman LE, Ingbar SH. Iodide-induced thyrotoxicosis in Boston. N Engl J Med. 1972;287(11):523-7.
  62. Stanbury JB, Ermans AE, Bourdoux P, Todd C, Oken E, Tonglet R, et al. Iodine-induced hyperthyroidism: occurrence and epidemiology. Thyroid. 1998;8(1):83-100.
  63. Vidor GI, Stewart JC, Wall JR, Wangel A, Hetzel BS. Pathogenesis of iodine-induced thyrotoxicosis: studies in northern Tasmania. J Clin Endocrinol Metab. 1973;37(6):901-9.
  64. Stewart JC, Vidor GI, Buttifield IH, Hetzel BS. Epidemic thyrotoxicosis in northern Tasmania: studies of clinical features and iodine nutrition. Aust N Z J Med. 1971;1(3):203-11.
  65. Chandrasekaran M, Ramadevi K. Thyromegaly and iodine nutritional status in a tertiary care hospital in South India. Indian J Endocrinol Metab. 2013;17(2):260-4.
  66. Laurberg P, Jørgensen T, Perrild H, Ovesen L, Knudsen N, Pedersen IB, et al. The Danish investigation on iodine intake and thyroid disease, DanThyr: status and perspectives. Eur J Endocrinol. 2006;155(2):219-28.
  67. Boukis MA, Koutras DA, Souvatzoglou A, Evangelopoulou A, Vrontakis M, Moulopoulos SD. Thyroid hormone and immunological studies in endemic goiter. J Clin Endocrinol Metab. 1983;57(4):859-62.
  68. Rasooly L, Burek CL, Rose NR. Iodine-induced autoimmune thyroiditis in NOD-H-2h4 mice. Clin Immunol Immunopathol. 1996;81(3):287-92.
  69. Allen EM, Appel MC, Braverman LE. The effect of iodide ingestion on the development of spontaneous lymphocytic thyroiditis in the diabetes-prone BB/W rat. Endocrinology. 1986;118(5):1977-81.
  70. Kopp PA. Iodine in the Therapy of Graves' Disease: A Century After Henry S. Plummer. Thyroid. 2023;33(3):273-5.
  71. Feinberg WD, Hoffman DL, Owen CAJ. THE EFFECTS OF VARYING AMOUNTS OF STABLE IODIDE ON THE FUNCTION OF THE HUMAN THYROID*. The Journal of Clinical Endocrinology & Metabolism. 1959;19(5):567-82.
  72. Degroot LJ, Greer MA. The effect of stable iodide on thyroid secretion in man. Metabolism. 1956;5(6 Part 1):682-96.
  73. Buhler UK, DeGroot LJ. Effect of stable iodine on thyroid iodine release. J Clin Endocrinol Metab. 1969;29(12):1546-52.
  74. Carlé A, Pedersen IB, Knudsen N, Perrild H, Ovesen L, Rasmussen LB, et al. Epidemiology of subtypes of hyperthyroidism in Denmark: a population-based study. Eur J Endocrinol. 2011;164(5):801-9.
  75. Paunkovic N, Paunkovic J, Pavlovic O, Paunovic Z. The significant increase in incidence of Graves' disease in eastern Serbia during the civil war in the former Yugoslavia (1992 to 1995). Thyroid. 1998;8(1):37-41.
  76. Wang J, Chen Z, Carru C, Capobianco G, Sedda S, Li Z. What is the impact of stress on the onset and anti-thyroid drug therapy in patients with graves' disease: a systematic review and meta-analysis. BMC Endocr Disord. 2023;23(1):194.
  77. Chiovato L, Pinchera A. Stressful life events and Graves' disease. Eur J Endocrinol. 1996;134(6):680-2.
  78. Radosavljević VR, Janković SM, Marinković JM. Stressful life events in the pathogenesis of Graves' disease. Eur J Endocrinol. 1996;134(6):699-701.
  79. Winsa B, Adami HO, Bergström R, Gamstedt A, Dahlberg PA, Adamson U, et al. Stressful life events and Graves' disease. Lancet. 1991;338(8781):1475-9.
  80. Matos-Santos A, Nobre EL, Costa JG, Nogueira PJ, Macedo A, Galvão-Teles A, et al. Relationship between the number and impact of stressful life events and the onset of Graves' disease and toxic nodular goitre. Clin Endocrinol (Oxf). 2001;55(1):15-9.
  81. Landsberg L. Catecholamines and hyperthyroidism. Clin Endocrinol Metab. 1977;6(3):697-718.
  82. Bruun E. Exophthalmic goiter developing after treatment with thyroid preparations. Acta Med Scand. 1945;122:13-29.
  83. Takaba H, Takayanagi H. The Mechanisms of T Cell Selection in the Thymus. Trends Immunol. 2017;38(11):805-16.
  84. Strominger JL. Developmental biology of T cell receptors. Science. 1989;244(4907):943-50.
  85. Spitzweg C, Joba W, Heufelder AE. Expression of thyroid-related genes in human thymus. Thyroid. 1999;9(2):133-41.
  86. Sospedra M, Ferrer-Francesch X, Domínguez O, Juan M, Foz-Sala M, Pujol-Borrell R. Transcription of a broad range of self-antigens in human thymus suggests a role for central mechanisms in tolerance toward peripheral antigens. J Immunol. 1998;161(11):5918-29.
  87. Giménez-Barcons M, Casteràs A, Armengol Mdel P, Porta E, Correa PA, Marín A, et al. Autoimmune predisposition in Down syndrome may result from a partial central tolerance failure due to insufficient intrathymic expression of AIRE and peripheral antigens. J Immunol. 2014;193(8):3872-9.
  88. Miyara M, Sakaguchi S. Natural regulatory T cells: mechanisms of suppression. Trends Mol Med. 2007;13(3):108-16.
  89. Lahl K, Loddenkemper C, Drouin C, Freyer J, Arnason J, Eberl G, et al. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J Exp Med. 2007;204(1):57-63.
  90. Li Q, Wang B, Mu K, Zhang JA. The pathogenesis of thyroid autoimmune diseases: New T lymphocytes - Cytokines circuits beyond the Th1-Th2 paradigm. J Cell Physiol. 2019;234(3):2204-16.
  91. Nakano A, Watanabe M, Iida T, Kuroda S, Matsuzuka F, Miyauchi A, et al. Apoptosis-induced decrease of intrathyroidal CD4(+)CD25(+) regulatory T cells in autoimmune thyroid diseases. Thyroid. 2007;17(1):25-31.
  92. Klatka M, Grywalska E, Partyka M, Charytanowicz M, Kiszczak-Bochynska E, Rolinski J. Th17 and Treg cells in adolescents with Graves' disease. Impact of treatment with methimazole on these cell subsets. Autoimmunity. 2014;47(3):201-11.
  93. Mao C, Wang S, Xiao Y, Xu J, Jiang Q, Jin M, et al. Impairment of regulatory capacity of CD4+CD25+ regulatory T cells mediated by dendritic cell polarization and hyperthyroidism in Graves' disease. J Immunol. 2011;186(8):4734-43.
  94. Sridama V, Pacini F, DeGroot LJ. Decreased suppressor T-lymphocytes in autoimmune thyroid diseases detected by monoclonal antibodies. J Clin Endocrinol Metab. 1982;54(2):316-9.
  95. Wang Y, Fang S, Zhou H. Pathogenic role of Th17 cells in autoimmune thyroid disease and their underlying mechanisms. Best Pract Res Clin Endocrinol Metab. 2023;37(2):101743.
  96. Glick AB, Wodzinski A, Fu P, Levine AD, Wald DN. Impairment of regulatory T-cell function in autoimmune thyroid disease. Thyroid. 2013;23(7):871-8.
  97. Pan D, Shin YH, Gopalakrishnan G, Hennessey J, De Groot LJ. Regulatory T cells in Graves' disease. Clin Endocrinol (Oxf). 2009;71(4):587-93.
  98. Pacini F, DeGroot LJ. Studies of immunoglobulin synthesis in cultures of peripheral T and B lymphocytes: reduced T-suppressor cell activity in Graves' disease. Clin Endocrinol (Oxf). 1983;18(3):219-32.
  99. Topliss DJ, Okita N, Lewis M, Row VV, Volpé R. Allosuppressor T lymphocytes abolish migration inhibition factor production in autoimmune thyroid disease: evidence from radiosensitivity experiments. Clin Endocrinol (Oxf). 1981;15(4):335-41.
  100. Topliss D, How J, Lewis M, Row V, Volpé R. Evidence for cell-mediated immunity and specific suppressor T lymphocyte dysfunction in Graves' disease and diabetes mellitus. J Clin Endocrinol Metab. 1983;57(4):700-5.
  101. Marazuela M, García-López MA, Figueroa-Vega N, de la Fuente H, Alvarado-Sánchez B, Monsiváis-Urenda A, et al. Regulatory T cells in human autoimmune thyroid disease. J Clin Endocrinol Metab. 2006;91(9):3639-46.
  102. Gangi E, Vasu C, Cheatem D, Prabhakar BS. IL-10-producing CD4+CD25+ regulatory T cells play a critical role in granulocyte-macrophage colony-stimulating factor-induced suppression of experimental autoimmune thyroiditis. J Immunol. 2005;174(11):7006-13.
  103. Molteni M, Rossetti C, Scrofani S, Bonara P, Scorza R, Kohn LD. Regulatory CD8+ T cells control thyrotropin receptor-specific CD4+ clones in healthy subjects. Cancer Detect Prev. 2003;27(3):167-74.
  104. Vaidya B, Shenton BK, Stamp S, Miller M, Baister E, Andrews CD, et al. Analysis of peripheral blood T-cell subsets in active thyroid-associated ophthalmopathy: absence of effect of octreotide-LAR on T-cell subsets in patients with thyroid-associated ophthalmopathy. Thyroid. 2005;15(9):1073-8.
  105. Migita K, Eguchi K, Tezuka H, Otsubo T, Kawakami A, Nakao H, et al. Cytotoxic activity of interleukin-2 (IL-2) activated killer cells toward thyroid epithelial cells. Clin Exp Immunol. 1989;77(2):196-201.
  106. Piccinini LA, Mackenzie WA, Platzer M, Davies TF. Lymphokine regulation of HLA-DR gene expression in human thyroid cell monolayers. J Clin Endocrinol Metab. 1987;64(3):543-8.
  107. Eguchi K, Otsubo T, Kawabe Y, Shimomura C, Ueki Y, Nakao H, et al. Synergy in antigen presentation by thyroid epithelial cells and monocytes from patients with Graves' disease. Clin Exp Immunol. 1988;72(1):84-90.
  108. Bottazzo GF, Pujol-Borrell R, Hanafusa T, Feldmann M. Role of aberrant HLA-DR expression and antigen presentation in induction of endocrine autoimmunity. Lancet. 1983;2(8359):1115-9.
  109. Mukuta T, Arreaza G, Nishikawa M, Resetkova E, Jamieson C, Tamai H, et al. Thyroid xenografts from patients with Graves' disease in severe combined immunodeficient mice and NIH-beige-nude-xid mice. Clin Invest Med. 1997;20(1):5-15.
  110. Leclere J, Bene MC, Duprez A, Faure G, Thomas JL, Vignaud JM, et al. Behaviour of thyroid tissue from patients with Graves' disease in nude mice. J Clin Endocrinol Metab. 1984;59(1):175-7.
  111. Martin L. The hereditary and familial aspects of exophthalmic goitre and nodular goitre. Q J Med. 1945;14:207-19.
  112. Harvald B, Hauge M. A catamnestic investigation of Danish twins; a preliminary report. Dan Med Bull. 1956;3(5):150-8.
  113. Brix TH, Kyvik KO, Christensen K, Hegedüs L. Evidence for a major role of heredity in Graves' disease: a population-based study of two Danish twin cohorts. J Clin Endocrinol Metab. 2001;86(2):930-4.
  114. Hansen PS, Brix TH, Iachine I, Kyvik KO, Hegedüs L. The relative importance of genetic and environmental effects for the early stages of thyroid autoimmunity: a study of healthy Danish twins. Eur J Endocrinol. 2006;154(1):29-38.
  115. Berisso GA, van Lint MT, Bacigalupo A, Marmont AM. Adoptive autoimmune hyperthyroidism following allogeneic stem cell transplantation from an HLA-identical sibling with Graves' disease. Bone Marrow Transplant. 1999;23(10):1091-2.
  116. Taylor JC, Gough SC, Hunt PJ, Brix TH, Chatterjee K, Connell JM, et al. A genome-wide screen in 1119 relative pairs with autoimmune thyroid disease. J Clin Endocrinol Metab. 2006;91(2):646-53.
  117. Lee HJ, Stefan-Lifshitz M, Li CW, Tomer Y. Genetics and epigenetics of autoimmune thyroid diseases: Translational implications. Best Pract Res Clin Endocrinol Metab. 2023;37(2):101661.
  118. Campbell RD, Trowsdale J. Map of the human MHC. Immunol Today. 1993;14(7):349-52.
  119. Geluk A, Van Meijgaarden KE, Janson AA, Drijfhout JW, Meloen RH, De Vries RR, et al. Functional analysis of DR17(DR3)-restricted mycobacterial T cell epitopes reveals DR17-binding motif and enables the design of allele-specific competitor peptides. J Immunol. 1992;149(9):2864-71.
  120. Grumet FC, Payne RO, Konishi J, Kriss JP. HL-A antigens as markers for disease susceptibility and autoimmunity in Graves' disease. J Clin Endocrinol Metab. 1974;39(6):1115-9.
  121. Farid NR, Stone E, Johnson G. Graves' disease and HLA: clinical and epidemiologic associations. Clin Endocrinol (Oxf). 1980;13(6):535-44.
  122. Mangklabruks A, Cox N, DeGroot LJ. Genetic factors in autoimmune thyroid disease analyzed by restriction fragment length polymorphisms of candidate genes. J Clin Endocrinol Metab. 1991;73(2):236-44.
  123. Pichurin P, Chen CR, Pichurina O, David C, Rapoport B, McLachlan SM. Thyrotropin receptor-DNA vaccination of transgenic mice expressing HLA-DR3 or HLA-DQ6b. Thyroid. 2003;13(10):911-7.
  124. Yanagawa T, Mangklabruks A, Chang YB, Okamoto Y, Fisfalen ME, Curran PG, et al. Human histocompatibility leukocyte antigen-DQA1*0501 allele associated with genetic susceptibility to Graves' disease in a Caucasian population. J Clin Endocrinol Metab. 1993;76(6):1569-74.
  125. Yanagawa T, Mangklabruks A, DeGroot LJ. Strong association between HLA-DQA1*0501 and Graves' disease in a male Caucasian population. J Clin Endocrinol Metab. 1994;79(1):227-9.
  126. Sawai Y, DeGroot LJ. Binding of human thyrotropin receptor peptides to a Graves' disease-predisposing human leukocyte antigen class II molecule. J Clin Endocrinol Metab. 2000;85(3):1176-9.
  127. Kong YC, Lomo LC, Motte RW, Giraldo AA, Baisch J, Strauss G, et al. HLA-DRB1 polymorphism determines susceptibility to autoimmune thyroiditis in transgenic mice: definitive association with HLA-DRB1*0301 (DR3) gene. J Exp Med. 1996;184(3):1167-72.
  128. Ramgopal S, Rathika C, Padma MR, Murali V, Arun K, Kamaludeen MN, et al. Interaction of HLA-DRB1* alleles and CTLA4 (+49 AG) gene polymorphism in Autoimmune Thyroid Disease. Gene. 2018;642:430-8.
  129. Chen QY, Huang W, She JX, Baxter F, Volpe R, Maclaren NK. HLA-DRB1*08, DRB1*03/DRB3*0101, and DRB3*0202 are susceptibility genes for Graves' disease in North American Caucasians, whereas DRB1*07 is protective. J Clin Endocrinol Metab. 1999;84(9):3182-6.
  130. Thompson CB. Distinct roles for the costimulatory ligands B7-1 and B7-2 in T helper cell differentiation? Cell. 1995;81(7):979-82.
  131. Karandikar NJ, Vanderlugt CL, Walunas TL, Miller SD, Bluestone JA. CTLA-4: a negative regulator of autoimmune disease. J Exp Med. 1996;184(2):783-8.
  132. Donner H, Rau H, Walfish PG, Braun J, Siegmund T, Finke R, et al. CTLA4 alanine-17 confers genetic susceptibility to Graves' disease and to type 1 diabetes mellitus. J Clin Endocrinol Metab. 1997;82(1):143-6.
  133. Yanagawa T, Hidaka Y, Guimaraes V, Soliman M, DeGroot LJ. CTLA-4 gene polymorphism associated with Graves' disease in a Caucasian population. J Clin Endocrinol Metab. 1995;80(1):41-5.
  134. Yanagawa T, Taniyama M, Enomoto S, Gomi K, Maruyama H, Ban Y, et al. CTLA4 gene polymorphism confers susceptibility to Graves' disease in Japanese. Thyroid. 1997;7(6):843-6.
  135. Heward JM, Allahabadia A, Armitage M, Hattersley A, Dodson PM, Macleod K, et al. The development of Graves' disease and the CTLA-4 gene on chromosome 2q33. J Clin Endocrinol Metab. 1999;84(7):2398-401.
  136. Vaidya B, Imrie H, Perros P, Young ET, Kelly WF, Carr D, et al. The Cytotoxic T Lymphocyte Antigen-4 is a Major Graves' Disease Locus. Human Molecular Genetics. 1999;8(7):1195-9.
  137. Kouki T, Sawai Y, Gardine CA, Fisfalen ME, Alegre ML, DeGroot LJ. CTLA-4 gene polymorphism at position 49 in exon 1 reduces the inhibitory function of CTLA-4 and contributes to the pathogenesis of Graves' disease. J Immunol. 2000;165(11):6606-11.
  138. Zaletel K, Krhin B, Gaberscek S, Pirnat E, Hojker S. The influence of the exon 1 polymorphism of the cytotoxic T lymphocyte antigen 4 gene on thyroid antibody production in patients with newly diagnosed Graves' disease. Thyroid. 2002;12(5):373-6.
  139. Adams BD, Parsons C, Walker L, Zhang WC, Slack FJ. Targeting noncoding RNAs in disease. J Clin Invest. 2017;127(3):761-71.
  140. Chen X, Huang F, Qi Y, Zhou M, Yin Q, Peng Y, et al. Serum and thyroid tissue level of let-7b and their correlation with TRAb in Graves’ disease. Journal of Translational Medicine. 2018;16(1):188.
  141. Al-Heety RA, Al-Hadithi HS, Turki KM. Correlation of circulating miRNA-146a-5p and let-7b expression with thyroid-stimulating hormone receptor antibody in patients with graves disease. Gene Reports. 2020;19:100608.
  142. Martínez-Hernández R, Sampedro-Núñez M, Serrano-Somavilla A, Ramos-Leví AM, de la Fuente H, Triviño JC, et al. A MicroRNA Signature for Evaluation of Risk and Severity of Autoimmune Thyroid Diseases. The Journal of Clinical Endocrinology & Metabolism. 2018;103(3):1139-50.
  143. Yin L, Zeng C, Yao J, Shen J. Emerging Roles for Noncoding RNAs in Autoimmune Thyroid Disease. Endocrinology. 2020;161(8).
  144. Zou J, Peng H, Liu Y. The Roles of Exosomes in Immunoregulation and Autoimmune Thyroid Diseases. Front Immunol. 2021;12:757674.
  145. Wang Y, Xu F, Zhong JY, Lin X, Shan SK, Guo B, et al. Exosomes as Mediators of Cell-to-Cell Communication in Thyroid Disease. Int J Endocrinol. 2020;2020:4378345.
  146. Rossi M, Taddei AR, Fasciani I, Maggio R, Giorgi F. The cell biology of the thyroid-disrupting mechanism of dichlorodiphenyltrichloroethane (DDT). J Endocrinol Invest. 2018;41(1):67-73.
  147. Edo N, Kawakami K, Fujita Y, Morita K, Uno K, Tsukamoto K, et al. Exosomes Expressing Thyrotropin Receptor Attenuate Autoantibody-Mediated Stimulation of Cyclic Adenosine Monophosphate Production. Thyroid. 2019;29(7):1012-7.
  148. Hiratsuka I, Yamada H, Munetsuna E, Hashimoto S, Itoh M. Circulating MicroRNAs in Graves' Disease in Relation to Clinical Activity. Thyroid. 2016;26(10):1431-40.
  149. Hodgson NM, Rajaii F. Current Understanding of the Progression and Management of Thyroid Associated Orbitopathy: A Systematic Review. Ophthalmol Ther. 2020;9(1):21-33.
  150. Han J-S, Kim SE, Jin J-Q, Park NR, Lee J-Y, Kim HL, et al. Tear-Derived Exosome Proteins Are Increased in Patients with Thyroid Eye Disease. International Journal of Molecular Sciences. 2021;22(3):1115.
  151. Cuddihy RM, Dutton CM, Bahn RS. A polymorphism in the extracellular domain of the thyrotropin receptor is highly associated with autoimmune thyroid disease in females. Thyroid. 1995;5(2):89-95.
  152. Kotsa KD, Watson PF, Weetman AP. No association between a thyrotropin receptor gene polymorphism and Graves' disease in the female population. Thyroid. 1997;7(1):31-3.
  153. Tomer Y. Genetic susceptibility to autoimmune thyroid disease: past, present, and future. Thyroid. 2010;20(7):715-25.
  154. Hiratani H, Bowden DW, Ikegami S, Shirasawa S, Shimizu A, Iwatani Y, et al. Multiple SNPs in intron 7 of thyrotropin receptor are associated with Graves' disease. J Clin Endocrinol Metab. 2005;90(5):2898-903.
  155. Kuś A, Szymański K, Jurecka-Lubieniecka B, Pawlak-Adamska E, Kula D, Miśkiewicz P, et al. Gender-dependent and age-of-onset-specific association of the rs11675434 single-nucleotide polymorphism near TPO with susceptibility to Graves’ ophthalmopathy. Journal of Human Genetics. 2017;62(3):373-7.
  156. Huang C-J, Jap T-S. A systematic review of genetic studies of thyroid disorders in Taiwan. Journal of the Chinese Medical Association. 2015;78(3).
  157. Begum MN, Islam MT, Hossain SR, Bhuyan GS, Halim MA, Shahriar I, et al. Mutation Spectrum in TPO Gene of Bangladeshi Patients with Thyroid Dyshormonogenesis and Analysis of the Effects of Different Mutations on the Structural Features and Functions of TPO Protein through<i> In Silico</i> Approach. BioMed Research International. 2019;2019:9218903.
  158. Razmara E, Salehi M, Aslani S, Bitaraf A, Yousefi H, Colón JR, et al. Graves' disease: introducing new genetic and epigenetic contributors. J Mol Endocrinol. 2021;66(2):R33-r55.
  159. Citterio CE, Rivolta CM, Targovnik HM. Structure and genetic variants of thyroglobulin: Pathophysiological implications. Molecular and Cellular Endocrinology. 2021;528:111227.
  160. Ban Y, Taniyama M, Ban Y. Vitamin D receptor gene polymorphism is associated with Graves' disease in the Japanese population. J Clin Endocrinol Metab. 2000;85(12):4639-43.
  161. Vaidya B, Imrie H, Perros P, Young ET, Kelly WF, Carr D, et al. Evidence for a new Graves disease susceptibility locus at chromosome 18q21. Am J Hum Genet. 2000;66(5):1710-4.
  162. Tomer Y, Concepcion E, Greenberg DA. A C/T single-nucleotide polymorphism in the region of the CD40 gene is associated with Graves' disease. Thyroid. 2002;12(12):1129-35.
  163. Kurylowicz A, Kula D, Ploski R, Skorka A, Jurecka-Lubieniecka B, Zebracka J, et al. Association of CD40 gene polymorphism (C-1T) with susceptibility and phenotype of Graves' disease. Thyroid. 2005;15(10):1119-24.
  164. Shirasawa S, Harada H, Furugaki K, Akamizu T, Ishikawa N, Ito K, et al. SNPs in the promoter of a B cell-specific antisense transcript, SAS-ZFAT, determine susceptibility to autoimmune thyroid disease. Hum Mol Genet. 2004;13(19):2221-31.
  165. Lopez ER, Zwermann O, Segni M, Meyer G, Reincke M, Seissler J, et al. A promoter polymorphism of the CYP27B1 gene is associated with Addison's disease, Hashimoto's thyroiditis, Graves' disease and type 1 diabetes mellitus in Germans. Eur J Endocrinol. 2004;151(2):193-7.
  166. Imani D, Rezaei R, Razi B, Alizadeh S, Mahmoudi M. Association Between IL6-174 G/C Polymorphism and Graves' Disease: A Systematic Review and Meta-Analysis. Acta Med Iran. 2017;55(11):665-71.
  167. Hiromatsu Y, Fukutani T, Ichimura M, Mukai T, Kaku H, Nakayama H, et al. Interleukin-13 gene polymorphisms confer the susceptibility of Japanese populations to Graves' disease. J Clin Endocrinol Metab. 2005;90(1):296-301.
  168. Liu N, Li X, Liu C, Zhao Y, Cui B, Ning G. The association of interleukin-1alpha and interleukin-1beta polymorphisms with the risk of Graves' disease in a case-control study and meta-analysis. Hum Immunol. 2010;71(4):397-401.
  169. Zeitlin AA, Simmonds MJ, Gough SC. Genetic developments in autoimmune thyroid disease: an evolutionary process. Clin Endocrinol (Oxf). 2008;68(5):671-82.
  170. Tu Y, Fan G, Zeng T, Cai X, Kong W. Association of TNF-α promoter polymorphism and Graves' disease: an updated systematic review and meta-analysis. Biosci Rep. 2018;38(2).
  171. Ban Y, Tozaki T, Taniyama M, Tomita M, Ban Y. The codon 620 single nucleotide polymorphism of the protein tyrosine phosphatase-22 gene does not contribute to autoimmune thyroid disease susceptibility in the Japanese. Thyroid. 2005;15(10):1115-8.
  172. Zhang Q, Liu S, Guan Y, Chen Q, Zhang Q, Min X. RNASET2, GPR174, and PTPN22 gene polymorphisms are related to the risk of liver damage associated with the hyperthyroidism in patients with Graves’ disease. Journal of Clinical Laboratory Analysis. 2018;32(2):e22258.
  173. Wawrusiewicz-Kurylonek N, Koper-Lenkiewicz OM, Gościk J, Myśliwiec J, Pawłowski P, Krętowski AJ. Association of PTPN22 polymorphism and its correlation with Graves' disease susceptibility in Polish adult population—A preliminary study. Molecular Genetics & Genomic Medicine. 2019;7(6):e661.
  174. Valta M, Gazali AM, Viisanen T, Ihantola E-L, Ekman I, Toppari J, et al. Type 1 diabetes linked PTPN22 gene polymorphism is associated with the frequency of circulating regulatory T cells. European Journal of Immunology. 2020;50(4):581-8.
  175. Yuan M, Wei L, Zhou R, Bai Q, Wei Y, Zhang W, et al. Four FCRL3 Gene Polymorphisms (FCRL3_3, _5, _6, _8) Confer Susceptibility to Multiple Sclerosis: Results from a Case-Control Study. Molecular Neurobiology. 2016;53(3):2029-35.
  176. Fang Y, Li Y, Zeng J, Wang J, Liu R, Cao C. Genetic association of Fc receptor-like glycoprotein with susceptibility to Graves' disease in a Chinese Han population. Immunobiology. 2016;221(1):56-62.
  177. Schoenmakers EFPM, Wanschura S, Mols R, Bullerdiek J, Van den Berghe H, Van de Ven WJM. Recurrent rearrangements in the high mobility group protein gene, HMGI-C, in benign mesenchymal tumours. Nature Genetics. 1995;10(4):436-44.
  178. Yamada H, Watanabe M, Nanba T, Akamizu T, Iwatani Y. The +869T/C polymorphism in the transforming growth factor-beta1 gene is associated with the severity and intractability of autoimmune thyroid disease. Clin Exp Immunol. 2008;151(3):379-82.
  179. Sutherland A, Davies J, Owen CJ, Vaikkakara S, Walker C, Cheetham TD, et al. Genomic polymorphism at the interferon-induced helicase (IFIH1) locus contributes to Graves' disease susceptibility. J Clin Endocrinol Metab. 2007;92(8):3338-41.
  180. Vejrazkova D, Vcelak J, Vaclavikova E, Vankova M, Zajickova K, Duskova M, et al. Genetic predictors of the development and recurrence of Graves' disease. Physiol Res. 2018;67(Suppl 3):S431-s9.
  181. Calder EA, Penhale WJ, Barnes EW, Irvine WJ. Evidence for circulating immune complexes in thyroid disease. Br Med J. 1974;2(5909):30-1.
  182. Marinò M, Chiovato L, Friedlander JA, Latrofa F, Pinchera A, McCluskey RT. Serum antibodies against megalin (GP330) in patients with autoimmune thyroiditis. J Clin Endocrinol Metab. 1999;84(7):2468-74.
  183. Morris JC, Bergert ER, Bryant WP. Binding of immunoglobulin G from patients with autoimmune thyroid disease to rat sodium-iodide symporter peptides: evidence for the iodide transporter as an autoantigen. Thyroid. 1997;7(4):527-34.
  184. Kubota S, Gunji K, Stolarski C, Kennerdell JS, Wall J. Reevaluation of the prevalences of serum autoantibodies reactive with "64-kd eye muscle proteins" in patients with thyroid-associated ophthalmopathy. Thyroid. 1998;8(2):175-9.
  185. Mori T, Kriss JP. Measurements by competitive binding radioassay of serum anti-microsomal and anti-thyroglobulin antibodies in Graves' disease and other thyroid disorders. J Clin Endocrinol Metab. 1971;33(4):688-98.
  186. Wang PW, Huang MJ, Liu RT, Chen CD. Triiodothyronine autoantibodies in Graves' disease: their changes after antithyroid therapy and relationship with the thyroglobulin antibodies. Acta Endocrinol (Copenh). 1990;122(1):22-8.
  187. Nakamura S, Sakata S, Shima H, Komaki T, Kojima N, Kamikubo K, et al. Thyroid hormone autoantibodies (THAA) in two cases of Graves' disease: effects of antithyroid drugs, prednisolone, and subtotal thyroidectomy. Endocrinol Jpn. 1986;33(6):751-9.
  188. Kasagi K, Kousaka T, Higuchi K, Iida Y, Misaki T, Alam MS, et al. Clinical significance of measurements of antithyroid antibodies in the diagnosis of Hashimoto's thyroiditis: comparison with histological findings. Thyroid. 1996;6(5):445-50.
  189. Amino N, Hagen SR, Yamada N, Refetoff S. Measurement of circulating thyroid microsomal antibodies by the tanned red cell haemagglutination technique: its usefulness in the diagnosis of autoimmune thyroid diseases. Clin Endocrinol (Oxf). 1976;5(2):115-25.
  190. Mullins RJ, Cohen SB, Webb LM, Chernajovsky Y, Dayan CM, Londei M, et al. Identification of thyroid stimulating hormone receptor-specific T cells in Graves' disease thyroid using autoantigen-transfected Epstein-Barr virus-transformed B cell lines. J Clin Invest. 1995;96(1):30-7.
  191. Weetman AP, Gunn C, Hall R, McGregor AM. Thyroid autoantigen-induced lymphocyte proliferation in Graves' disease and Hashimoto's thyroiditis. J Clin Lab Immunol. 1985;17(1):1-6.
  192. Fisfalen ME, DeGroot LJ, Quintans J, Franklin WA, Soltani K. Microsomal antigen-reactive lymphocyte lines and clones derived from thyroid tissue of patients with Graves' disease. J Clin Endocrinol Metab. 1988;66(4):776-84.
  193. Soliman M, Kaplan E, Fisfalen ME, Okamoto Y, DeGroot LJ. T-cell reactivity to recombinant human thyrotropin receptor extracellular domain and thyroglobulin in patients with autoimmune and nonautoimmune thyroid diseases. J Clin Endocrinol Metab. 1995;80(1):206-13.
  194. Aoki N, DeGroot J. Lymphocyte blastogenic response to human thyroglobulin in Graves' disease, Hashimoto's thyroiditis, and metastatic thyroid cancer. Clin Exp Immunol. 1979;38(3):523-30.
  195. Inaba H, De Groot LJ, Akamizu T. Thyrotropin Receptor Epitope and Human Leukocyte Antigen in Graves' Disease. Front Endocrinol (Lausanne). 2016;7:120.
  196. Soliman M, Kaplan E, Yanagawa T, Hidaka Y, Fisfalen ME, DeGroot LJ. T-cells recognize multiple epitopes in the human thyrotropin receptor extracellular domain. J Clin Endocrinol Metab. 1995;80(3):905-14.
  197. Kula D, Bednarczuk T, Jurecka-Lubieniecka B, Polanska J, Hasse-Lazar K, Jarzab M, et al. Interaction of HLA-DRB1 alleles with CTLA-4 in the predisposition to Graves' disease: the impact of DRB1*07. Thyroid. 2006;16(5):447-53.
  198. Tandon N, Freeman M, Weetman AP. T cell responses to synthetic thyroid peroxidase peptides in autoimmune thyroid disease. Clin Exp Immunol. 1991;86(1):56-60.
  199. Fisfalen ME, Soliman M, Okamoto Y, Soltani K, DeGroot LJ. Proliferative responses of T-cells to thyroid antigens and synthetic thyroid peroxidase peptides in autoimmune thyroid disease. J Clin Endocrinol Metab. 1995;80(5):1597-604.
  200. Chiovato L, Bassi P, Santini F, Mammoli C, Lapi P, Carayon P, et al. Antibodies producing complement-mediated thyroid cytotoxicity in patients with atrophic or goitrous autoimmune thyroiditis. J Clin Endocrinol Metab. 1993;77(6):1700-5.
  201. 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(3):929-34.
  202. Ploth DW, Fitz A, Schnetzler D, Seidenfeld J, Wilson CB. Thyroglobulin-anti-thyroglobulin immune complex glomerulonephritis complicating radioiodine therapy. Clin Immunol Immunopathol. 1978;9(3):327-34.
  203. Matsuura M, Kikkawa Y, Akashi K, Kitagawa T, Inage Z, Iwamori M, et al. Thyroid antigen-antibody nephritis: possible involvement of fucosyl-GM1 as the antigen. Endocrinol Jpn. 1987;34(4):587-93.
  204. Carvalho GAd, Perez CLS, Ward LS. Utilização dos testes de função tireoidiana na prática clínica. Arquivos Brasileiros de Endocrinologia & Metabologia. 2013;57.
  205. Fröhlich E, Wahl R. Thyroid Autoimmunity: Role of Anti-thyroid Antibodies in Thyroid and Extra-Thyroidal Diseases. Front Immunol. 2017;8:521.
  206. Tada H, Izumi Y, Watanabe Y, Takano T, Fukata S, Kuma K, et al. Blocking Type Anti-TSH Receptor Antibodies Detected by Radioreceptor Assay in Graves' Disease. Endocrine Journal. 2001;48(6):703-10.
  207. Kim WB, Chung HK, Park YJ, Park DJ, Tahara K, Kohn LD, et al. The Prevalence and Clinical Significance of Blocking Thyrotropin Receptor Antibodies in Untreated Hyperthyroid Graves' Disease. Thyroid®. 2000;10(7):579-86.
  208. Roti E, Braverman LE, DeGroot LJ. TSH Receptor Antibody Measurement in the Diagnosis and Management of Graves’ Disease Is Rarely Necessaryb. The Journal of Clinical Endocrinology & Metabolism. 1998;83(11):3781-4.
  209. Kotwal A, Stan M. Thyrotropin Receptor Antibodies-An Overview. Ophthalmic Plast Reconstr Surg. 2018;34(4S Suppl 1):S20-s7.
  210. Cárdenas Roldán J, Amaya-Amaya J, Castellanos-de la Hoz J, Giraldo-Villamil J, Montoya-Ortiz G, Cruz-Tapias P, et al. Autoimmune thyroid disease in rheumatoid arthritis: a global perspective. Arthritis. 2012;2012:864907.
  211. Yavasoglu I, Senturk T, Coskun A, Bolaman Z. Rheumatoid arthritis and anti-thyroid antibodies. Autoimmunity. 2009;42(2):168-9.
  212. Jenkins RC, Weetman AP. Disease associations with autoimmune thyroid disease. Thyroid. 2002;12(11):977-88.
  213. Ochi Y, DeGroot LJ. Vitiligo in Graves' disease. Ann Intern Med. 1969;71(5):935-40.
  214. Welti H. [Skin manifestations associated with severe forms of Basedow's disease]. Bull Schweiz Akad Med Wiss. 1968;23(5):476-82.
  215. Amoroso A, Garzia P, Pasquarelli C, Sportelli G, Afeltra A. Hashimoto's thyroiditis associated with urticaria and angio-oedema: disappearance of cutaneous and mucosal manifestations after thyroidectomy. J Clin Pathol. 1997;50(3):254-6.
  216. Marinó M, Ricciardi R, Pinchera A, Barbesino G, Manetti L, Chiovato L, et al. Mild clinical expression of myasthenia gravis associated with autoimmune thyroid diseases. J Clin Endocrinol Metab. 1997;82(2):438-43.
  217. Sonnet E, Massart C, Gibassier J, Allannic H, Maugendre D. Longitudinal study of soluble intercellular adhesion molecule-1 (ICAM-1) in sera of patients with Graves' disease. J Endocrinol Invest. 1999;22(6):430-5.
  218. Marshall JS, Weisberger AS, Levy RP, Breckenridge RT. Coexistent Idiopathic Thrombocytopenic Purpura and Hyperthyroidism. Annals of Internal Medicine. 1967;67(2):411-4.
  219. White RG, Bass BH, Williams E. Lymphadenoid goitre and the syndrome of systemic lupus erythematosus. Lancet. 1961;1(7173):368-73.
  220. Tektonidou MG, Anapliotou M, Vlachoyiannopoulos P, Moutsopoulos HM. Presence of systemic autoimmune disorders in patients with autoimmune thyroid diseases. Ann Rheum Dis. 2004;63(9):1159-61.
  221. Irvine WJ, Davies SH, Teitelbaum S, Delamore IW, Williams AW. The clinical and pathological significance of gastric parietal cell antibody. Ann N Y Acad Sci. 1965;124(2):657-91.
  222. Dittmar M, Kahaly GJ. Polyglandular Autoimmune Syndromes: Immunogenetics and Long-Term Follow-Up. The Journal of Clinical Endocrinology & Metabolism. 2003;88(7):2983-92.
  223. Katakura M, Yamada T, Aizawa T, Hiramatsu K, Yukimura Y, Ishihara M, et al. Presence of antideoxyribonucleic acid antibody in patients with hyperthyroidism of Graves' disease. J Clin Endocrinol Metab. 1987;64(3):405-8.
  224. Cowling DC, Mackay IR, Taft LI. Lupoid hepatitis. Lancet. 1956;271(6957):1323-6.
  225. Paggi A, Caccavo D, Ferri GM, Di Prima MA, Amoroso A, Vaccaro F, et al. Anti-cardiolipin antibodies in autoimmune thyroid diseases. Clin Endocrinol (Oxf). 1994;40(3):329-33.
  226. Siddiqi A, Monson JP, Wood DF, Besser GM, Burrin JM. Serum cytokines in thyrotoxicosis. J Clin Endocrinol Metab. 1999;84(2):435-9.
  227. Orgiazzi J, Williams DE, Chopra IJ, Solomon DH. Human thyroid adenyl cyclase-stimulating activity in immunoglobulin G of patients with Graves' disease. J Clin Endocrinol Metab. 1976;42(2):341-54.
  228. Evans C, Morgenthaler NG, Lee S, Llewellyn DH, Clifton-Bligh R, John R, et al. Development of a luminescent bioassay for thyroid stimulating antibodies. J Clin Endocrinol Metab. 1999;84(1):374-7.
  229. Di Cerbo A, Di Paola R, Menzaghi C, De Filippis V, Tahara K, Corda D, et al. Graves' immunoglobulins activate phospholipase A2 by recognizing specific epitopes on thyrotropin receptor. J Clin Endocrinol Metab. 1999;84(9):3283-92.
  230. Smyth PP, McMullan NM, Grubeck-Loebenstein B, O'Donovan DK. Thyroid growth-stimulating immunoglobulins in goitrous disease: relationship to thyroid-stimulating immunoglobulins. Acta Endocrinol (Copenh). 1986;111(3):321-30.
  231. Endo K, Kasagi K, Konishi J, Ikekubo K, Okuno T, Takeda Y, et al. Detection and properties of TSH-binding inhibitor immunoglobulins in patients with Graves' disease and Hashimoto's thyroiditis. J Clin Endocrinol Metab. 1978;46(5):734-9.
  232. Irvine WJ, Lamberg BA, Cullen DR, Gordin R. Primary hypothyroidism preceding thyrotoxicosis: a report of 2 cases and a review of the literature. J Clin Lab Immunol. 1979;2(4):349-52.
  233. Eckstein AK, Plicht M, Lax H, Neuhäuser M, Mann K, Lederbogen S, et al. 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(9):3464-70.
  234. Morshed SA, Davies TF. Graves' Disease Mechanisms: The Role of Stimulating, Blocking, and Cleavage Region TSH Receptor Antibodies. Horm Metab Res. 2015;47(10):727-34.
  235. 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. 2010;151(11):5537-49.
  236. Ando T, Latif R, Daniel S, Eguchi K, Davies TF. Dissecting linear and conformational epitopes on the native thyrotropin receptor. Endocrinology. 2004;145(11):5185-93.
  237. Misrahi M, Milgrom E. Cleavage and shedding of the TSH receptor. Eur J Endocrinol. 1997;137(6):599-602.
  238. McKenzie JM. FURTHER EVIDENCE FOR A THYROID ACTIVATOR IN HYPERTHYROIDISM*. The Journal of Clinical Endocrinology & Metabolism. 1960;20(3):380-8.
  239. Takasu N, Oshiro C, Akamine H, Komiya I, Nagata A, Sato Y, et al. Thyroid-stimulating antibody and TSH-binding inhibitor immunoglobulin in 277 Graves' patients and in 686 normal subjects. J Endocrinol Invest. 1997;20(8):452-61.
  240. Stan MN, Algeciras-Schimnich A, Murthy V, Thapa P, Araki N. Diagnostic Utility of a New Assay for Thyroid Stimulating Immunoglobulins in Graves' Disease and Thyroid Eye Disease. Thyroid®. 2021;32(2):170-6.
  241. 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(1):14-24.
  242. Ahmad E, Hafeez K, Arshad MF, Isuga J, Vrettos A. Hypothyroidism conversion to hyperthyroidism: it's never too late. Endocrinol Diabetes Metab Case Rep. 2018;2018.
  243. Kuzuya N, Chiu SC, Ikeda H, Uchimura H, Ito K, Nagataki S. Correlation between thyroid stimulators and 3,5,3'-triiodothyronine suppressibility in patients during treatment for hyperthyroidism with thionamide drugs: comparison of assays by thyroid-stimulating and thyrotropin-displacing activities. J Clin Endocrinol Metab. 1979;48(4):706-11.
  244. Visscher SL, Naessens JM, Yawn BP, Reinalda MS, Anderson SS, Borah BJ. Developing a standardized healthcare cost data warehouse. BMC Health Serv Res. 2017;17(1):396.
  245. Soliman M, Kaplan E, Abdel-Latif A, Scherberg N, DeGroot LJ. Does thyroidectomy, radioactive iodine therapy, or antithyroid drug treatment alter reactivity of patients' T cells to epitopes of thyrotropin receptor in autoimmune thyroid diseases? J Clin Endocrinol Metab. 1995;80(8):2312-21.
  246. Fenzi G, Hashizume K, Roudebush CP, DeGroot LJ. Changes in thyroid-stimulating immunoglobulins during antithyroid therapy. J Clin Endocrinol Metab. 1979;48(4):572-6.
  247. Mukhtar ED, Smith BR, Pyle GA, Hall R, Vice P. Relation of thyroid-stimulating immunoglobulins to thyroid function and effects of surgery, radioiodine, and antithyroid drugs. Lancet. 1975;1(7909):713-5.
  248. Nygaard B, Metcalfe RA, Phipps J, Weetman AP, Hegedüs L. Graves' disease and thyroid associated ophthalmopathy triggered by 131I treatment of non-toxic goiter. J Endocrinol Invest. 1999;22(6):481-5.
  249. Pichurin P, Pham N, David CS, Rapoport B, McLachlan SM. HLA-DR3 transgenic mice immunized with adenovirus encoding the thyrotropin receptor: T cell epitopes and functional analysis of the CD40 Graves' polymorphism. Thyroid. 2006;16(12):1221-7.
  250. Inaba H, Pan D, Shin YH, Martin W, Buchman G, De Groot LJ. Immune response of mice transgenic for human histocompatibility leukocyte Antigen-DR to human thyrotropin receptor-extracellular domain. Thyroid. 2009;19(11):1271-80.
  251. Pearce SHS, Dayan C, Wraith DC, Barrell K, Olive N, Jansson L, et al. Antigen-Specific Immunotherapy with Thyrotropin Receptor Peptides in Graves' Hyperthyroidism: A Phase I Study. Thyroid. 2019;29(7):1003-11.
  252. Jansson L, Vrolix K, Jahraus A, Martin KF, Wraith DC. Immunotherapy With Apitopes Blocks the Immune Response to TSH Receptor in HLA-DR Transgenic Mice. Endocrinology. 2018;159(9):3446-57.
  253. Diana T, Olivo PD, Kahaly GJ. Thyrotropin Receptor Blocking Antibodies. Horm Metab Res. 2018;50(12):853-62.
  254. Núñez Miguel R, Sanders P, Allen L, Evans M, Holly M, Johnson W, et al. Structure of full-length TSH receptor in complex with antibody K1-70™. J Mol Endocrinol. 2023;70(1).
  255. Fisfalen ME, Palmer EM, Van Seventer GA, Soltani K, Sawai Y, Kaplan E, et al. Thyrotropin-receptor and thyroid peroxidase-specific T cell clones and their cytokine profile in autoimmune thyroid disease. J Clin Endocrinol Metab. 1997;82(11):3655-63.
  256. Ferrari SM, Paparo SR, Ragusa F, Elia G, Mazzi V, Patrizio A, et al. Chemokines in thyroid autoimmunity. Best Pract Res Clin Endocrinol Metab. 2023;37(2):101773.
  257. Hiromatsu Y, Yang D, Bednarczuk T, Miyake I, Nonaka K, Inoue Y. Cytokine Profiles in Eye Muscle Tissue and Orbital Fat Tissue from Patients with Thyroid-Associated Ophthalmopathy*. The Journal of Clinical Endocrinology & Metabolism. 2000;85(3):1194-9.
  258. Wakelkamp IM, Gerding MN, Van Der Meer JW, Prummel MF, Wiersinga WM. Both Th1- and Th2-derived cytokines in serum are elevated in Graves' ophthalmopathy. Clin Exp Immunol. 2000;121(3):453-7.
  259. Zheng L, Ye P, Liu C. The role of the IL-23/IL-17 axis in the pathogenesis of Graves' disease. Endocr J. 2013;60(5):591-7.
  260. Kościuszko M, Popławska-Kita A, Pawłowski P, Lipińska D, Hryniewicka J, Jankowska D, et al. Clinical relevance of estimating circulating interleukin-17 and interleukin-23 during methylprednisolone therapy in Graves’ orbitopathy: A preliminary study. Advances in Medical Sciences. 2021;66(2):315-20.
  261. Inaba H, Martin W, Ardito M, De Groot AS, De Groot LJ. The role of glutamic or aspartic acid in position four of the epitope binding motif and thyrotropin receptor-extracellular domain epitope selection in Graves' disease. J Clin Endocrinol Metab. 2010;95(6):2909-16.
  262. Crisp M, Starkey KJ, Lane C, Ham J, Ludgate M. Adipogenesis in thyroid eye disease. Invest Ophthalmol Vis Sci. 2000;41(11):3249-55.
  263. Bell A, Gagnon A, Grunder L, Parikh SJ, Smith TJ, Sorisky A. Functional TSH receptor in human abdominal preadipocytes and orbital fibroblasts. Am J Physiol Cell Physiol. 2000;279(2):C335-40.
  264. Haraguchi K, Shimura H, Kawaguchi A, Ikeda M, Endo T, Onaya T. Effects of thyrotropin on the proliferation and differentiation of cultured rat preadipocytes. Thyroid. 1999;9(6):613-9.
  265. Kasagi K, Konishi J, Endo K, Mori T, Nagahara K, Makimoto K, et al. Adenylate cyclase activity in thyroid tissue from patients with untreated Graves' disease. J Clin Endocrinol Metab. 1980;51(3):492-9.
  266. Intenzo CM, dePapp AE, Jabbour S, Miller JL, Kim SM, Capuzzi DM. Scintigraphic manifestations of thyrotoxicosis. Radiographics. 2003;23(4):857-69.
  267. Acland JD. The interpretation of the serum protein-bound iodine: A review. J Clin Pathol. 1971;24(3):187-218.
  268. Farran HE, Shalom ES. Effect of L-tyrosine upon the protein bound iodine in thyrotoxicosis. J Clin Endocrinol Metab. 1966;26(8):918-20.
  269. Sterling K, Chodos RB. Radiothyroxine turnover studies in myxedema, thyrotoxicosis, and hypermetabolism without endocrine disease. J Clin Invest. 1956;35(7):806-13.
  270. Ingbar SH, Freinkel N. Studies of thyroid function and the peripheral metabolism of I 131-labeled thyroxine in patients with treated Graves disease. J Clin Invest. 1958;37(11):1603-14.
  271. Uller RP, Van Herle AJ. Effect of therapy on serum thyroglobulin levels in patients with Graves' disease. J Clin Endocrinol Metab. 1978;46(5):747-55.
  272. Greer MA, Smith GE. Method for increasing the accuracy of the radioiodine uptake as a test for thyroid function by the use of desiccated thyroid. J Clin Endocrinol Metab. 1954;14(11):1374-84.
  273. Werner SC, Spooner M. A new and simple test for hyperthyroidism employing L-triiodothyronine and the twenty-four hour I-131 uptake method. Bull N Y Acad Med. 1955;31(2):137-45.
  274. Raben MS. The paradoxical effects of thiocyanate and of thyrotropin on the organic binding of iodine by the thyroid in the presence of large amounts of iodide. Endocrinology. 1949;45(3):296-304.
  275. Suzuki H, Mashimo K. Significance of the iodide-perchlorate discharge test in patients with 131 I-treated and untreated hyperthyroidism. J Clin Endocrinol Metab. 1972;34(2):332-8.
  276. Uchida T, Shimamura M, Taka H, Kaga N, Miura Y, Nishida Y, et al. The Effect of Long-Term Inorganic Iodine on Intrathyroidal Iodothyronine Content and Gene Expression in Mice with Graves' Hyperthyroidism. Thyroid. 2023;33(3):330-7.
  277. Ochi Y, DeGroot LJ. TSH- or LATS-stimulated thyroid hormone release is inhibited by iodide. Endocrinology. 1969;84(6):1305-9.
  278. Grollman EF, Smolar A, Ommaya A, Tombaccini D, Santisteban P. Iodine Suppression of Iodide Uptake in FRTL-5 Thyroid Cells. Endocrinology. 1986;118(6):2477-82.
  279. Granner DK, Scranton JR, Curtis SJ. Kinetic analysis of thyroidal iodide concentration in hypophysectomized rats fed high or low iodine diets. Endocrinology. 1963;72:503-4.
  280. Sherwin JR, Tong W. The actions of iodide and TSH on thyroid cells showing a dual control system for the iodide pump. Endocrinology. 1974;94(5):1465-74.
  281. Sato S, Noh JY, Sato S, Suzuki M, Yasuda S, Matsumoto M, et al. Comparison of efficacy and adverse effects between methimazole 15 mg+inorganic iodine 38 mg/day and methimazole 30 mg/day as initial therapy for Graves' disease patients with moderate to severe hyperthyroidism. Thyroid. 2015;25(1):43-50.
  282. Takata K, Amino N, Kubota S, Sasaki I, Nishihara E, Kudo T, et al. Benefit of short-term iodide supplementation to antithyroid drug treatment of thyrotoxicosis due to Graves' disease. Clin Endocrinol (Oxf). 2010;72(6):845-50.
  283. Krieger CC, Neumann S, Place RF, Marcus-Samuels B, Gershengorn MC. Bidirectional TSH and IGF-1 receptor cross talk mediates stimulation of hyaluronan secretion by Graves' disease immunoglobins. J Clin Endocrinol Metab. 2015;100(3):1071-7.
  284. Neumann S, Krieger Christine C, Gershengorn Marvin C. Targeting TSH and IGF-1 Receptors to Treat Thyroid Eye Disease. European Thyroid Journal. 2020;9(Suppl. 1):59-65.
  285. Minich WB, Dehina N, Welsink T, Schwiebert C, Morgenthaler NG, Köhrle J, et al. Autoantibodies to the IGF1 receptor in Graves' orbitopathy. J Clin Endocrinol Metab. 2013;98(2):752-60.
  286. Heimann P. ULTRASTRUCTURE OF HUMAN THYROID. Acta Endocrinologica (Norway). 1966;53(2_Supplement):S6-S102.
  287. LiVolsi VA, Baloch ZW. The Pathology of Hyperthyroidism. Frontiers in Endocrinology. 2018;9.
  288. Bostrom H, Hed R. Thyrotoxic myopathy and polymyositis in elderly patients; differential-diagnostic viewpoints. Acta Med Scand. 1958;162(3):225-30.
  289. McEachern D, Ross WD. CHRONIC THYROTOXIC MYOPATHY1: A REPORT OF THREE CASES WITH A REVIEW OF PREVIOUSLY REPORTED CASES. Brain. 1942;65(2):181-92.
  290. Rundle FF, Finlay-Jones LR, Noad KB. MALIGNANT EXOPHTHALMOS: A QUANTITATIVE ANALYSIS OF THE ORBITAL TISSUES. Australasian Annals of Medicine. 1953;2(2):128-35.
  291. Dudgeon LS, Urquhart AL. LYMPHORRHAGES IN THE MUSCLES IN EXOPHTHALMIC GOITRE. Brain. 1926;49(2):182-6.
  292. Ezrin C, Swanson HE, Humphrey JG, Dawson JW, Hill FM. THE CELLS OF THE HUMAN ADENOHYPOPHYSIS IN THYROID DISORDERS*. The Journal of Clinical Endocrinology & Metabolism. 1959;19(8):958-66.
  293. Scheithauer BW, Kovacs KT, Young WF, Jr., Randall RV. The pituitary gland in hyperthyroidism. Mayo Clin Proc. 1992;67(1):22-6.
  294. Movitt ER, Gerstl B, Davis AE. NEEDLE LIVER BIOPSY IN THYROTOXICOSIS. AMA Archives of Internal Medicine. 1953;91(6):729-39.
  295. Piper J, Poulsen E. Liver biopsy in thyrotoxicosis. Acta Med Scand. 1947;127(4):439-47.
  296. Okosieme OE, Taylor PN, Evans C, Thayer D, Chai A, Khan I, et al. Primary therapy of Graves' disease and cardiovascular morbidity and mortality: a linked-record cohort study. Lancet Diabetes Endocrinol. 2019;7(4):278-87.
  297. Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N Engl J Med. 2024;390(6):497-509.
  298. Fallon MD, Perry HM, 3rd, Bergfeld M, Droke D, Teitelbaum SL, Avioli LV. Exogenous hyperthyroidism with osteoporosis. Arch Intern Med. 1983;143(3):442-4.
  299. Follis RH, Jr. Skeletal changes associated with hyperthyroidism. Bull Johns Hopkins Hosp. 1953;92(6):405-21.
  300. Hershman JM. Human chorionic gonadotropin and the thyroid: hyperemesis gravidarum and trophoblastic tumors. Thyroid. 1999;9(7):653-7.
  301. Nguyen CT, Sasso EB, Barton L, Mestman JH. Graves' hyperthyroidism in pregnancy: a clinical review. Clin Diabetes Endocrinol. 2018;4:4.
  302. White WH. On the Prognosis of Secondary Symptoms and Conditions of Exophthalmic Goitre. Br Med J. 1886;2(1334):151-3.
  303. Sattler H. Basedow's disease. New York: Grune & Stratton; 1952.
  304. Arnold BM, Casal G, Higgins HP. Apathetic thyrotoxicosis. Can Med Assoc J. 1974;111(9):957-8.
  305. Leblond CP, Fertman MB, et al. Radio-iodine autography in studies of human goitrous thyroid glands. Arch Pathol (Chic). 1946;41:510-5.
  306. Collazo-Clavell ML, Gharib H, Maragos NE. Relationship between vocal cord paralysis and benign thyroid disease. Head Neck. 1995;17(1):24-30.
  307. Mahaux JE, Chamla-Soumenkoff J, Delcourt R, Levin S. Painful enlargement of left subtrapezoid lymph nodes in Graves's disease. Br Med J. 1971;1(5745):384.
  308. Rumbyrt JS, Schocket AL. Chronic urticaria and thyroid disease. Immunol Allergy Clin North Am. 2004;24(2):215-23, vi.
  309. Fatourechi V, Garrity JA, Bartley GB, Bergstralh EJ, Gorman CA. Orbital decompression in Graves' ophthalmopathy associated with pretibial myxedema. J Endocrinol Invest. 1993;16(6):433-7.
  310. Bartley GB, Fatourechi V, Kadrmas EF, Jacobsen SJ, Ilstrup DM, Garrity JA, et al. The incidence of Graves' ophthalmopathy in Olmsted County, Minnesota. Am J Ophthalmol. 1995;120(4):511-7.
  311. Swanson JW, Kelly JJ, Jr., McConahey WM. Neurologic aspects of thyroid dysfunction. Mayo Clin Proc. 1981;56(8):504-12.
  312. Brownlie BE, Rae AM, Walshe JW, Wells JE. Psychoses associated with thyrotoxicosis - 'thyrotoxic psychosis.' A report of 18 cases, with statistical analysis of incidence. Eur J Endocrinol. 2000;142(5):438-44.
  313. Adams R, Oh ES, Yasar S, Lyketsos CG, Mammen JS. Endogenous and Exogenous Thyrotoxicosis and Risk of Incident Cognitive Disorders in Older Adults. JAMA Intern Med. 2023;183(12):1324-31.
  314. Gold HK, Spann JF, Jr., Braunwald E. Effect of alterations in the thyroid state on the intrinsic contractile properties of isolated rat skeletal muscle. J Clin Invest. 1970;49(4):849-54.
  315. Woodbury DM, Hurley RE, Lewis NG, McArthur MW, Copeland WW, Kirschvink JF, et al. Effect of thyroxin, thyroidectomy and 6-n-propyl-2-thiouracil on brain function. J Pharmacol Exp Ther. 1952;106(3):331-40.
  316. Ravera JJ, Cervino JM, Fernandez G, Ferrari Forcade A, Malosetti H, Muxi F, et al. Two cases of Graves' disease with signs of a pyramidal lesion. Improvement in neurologic signs during treatment with antithyroid drugs. J Clin Endocrinol Metab. 1960;20:876-80.
  317. Feibel JH, Campa JF. Thyrotoxic neuropathy (Basedow's paraplegia). J Neurol Neurosurg Psychiatry. 1976;39(5):491-7.
  318. WaldenstrÖM JAN. Acute Thyrotoxic Encephalo- or Myopathy, its cause and treatment. Acta Medica Scandinavica. 1945;121(2-3):251-94.
  319. Condon JV, Becka DR, Gibbs FA. Electroencephalographic abnormalities in hyperthyroidism. J Clin Endocrinol Metab. 1954;14(12):1511-8.
  320. Cantón A, de Fàbregas O, Tintoré M, Mesa J, Codina A, Simó R. Encephalopathy associated to autoimmune thyroid disease: a more appropriate term for an underestimated condition? J Neurol Sci. 2000;176(1):65-9.
  321. Priest WM. Chronic thyrotoxic myopathy. Br Med J. 1967;4(5574):295-6.
  322. Whitfield AGW, Hudson WA. CHRONIC THYROTOXIC MYOPATHY. QJM: An International Journal of Medicine. 1961;30(3):257-67.
  323. Sanderson KV, Adey WR. Electromyographic and endocrine studies in chronic thyrotoxic myopathy. J Neurol Neurosurg Psychiatry. 1952;15(3):200-5.
  324. Zürcher RM, Horber FF, Grünig BE, Frey FJ. Effect of thyroid dysfunction on thigh muscle efficiency. J Clin Endocrinol Metab. 1989;69(5):1082-6.
  325. Erkintalo M, Bendahan D, Mattéi JP, Fabreguettes C, Vague P, Cozzone PJ. Reduced metabolic efficiency of skeletal muscle energetics in hyperthyroid patients evidenced quantitatively by in vivo phosphorus-31 magnetic resonance spectroscopy. Metabolism. 1998;47(7):769-76.
  326. Fitch CD, Coker R, Dinning JS. Metabolism of creatine-1-C14 by vitamin E-deficient and hyperthyroid rats. Am J Physiol. 1960;198:1232-4.
  327. Thorn GW. CREATINE STUDIES IN THYROID DISORDERS. Endocrinology. 1936;20:628-34.
  328. Drachman DB. Myasthenia gravis. N Engl J Med. 1994;330(25):1797-810.
  329. Engel AG. Thyroid function and periodic paralysis. Am J Med. 1961;30:327-33.
  330. Ober KP. Thyrotoxic periodic paralysis in the United States. Report of 7 cases and review of the literature. Medicine (Baltimore). 1992;71(3):109-20.
  331. Kelley DE, Gharib H, Kennedy FP, Duda RJ, Jr., McManis PG. Thyrotoxic periodic paralysis. Report of 10 cases and review of electromyographic findings. Arch Intern Med. 1989;149(11):2597-600.
  332. Hsieh MJ, Lyu RK, Chang WN, Chang KH, Chen CM, Chang HS, et al. Hypokalemic thyrotoxic periodic paralysis: clinical characteristics and predictors of recurrent paralytic attacks. Eur J Neurol. 2008;15(6):559-64.
  333. Lin SH, Huang CL. Mechanism of thyrotoxic periodic paralysis. J Am Soc Nephrol. 2012;23(6):985-8.
  334. Gbefon CY, Sobral CPS, Caldas A, Rocha VCC, Azulay RSS, Nascimento GC, et al. Genetic Screening of Patients with Thyrotoxic Hypokalemic Periodic Paralysis: An Experience from a Tertiary Care Hospital in the Northeast of Brazil. Endocr Metab Immune Disord Drug Targets. 2021;21(12):2231-7.
  335. Brugnoni R, Canioni E, Filosto M, Pini A, Tonin P, Rossi T, et al. Mutations associated with hypokalemic periodic paralysis: from hotspot regions to complete analysis of CACNA1S and SCN4A genes. Neurogenetics. 2022;23(1):19-25.
  336. Rhee EP, Scott JA, Dighe AS. Case records of the Massachusetts General Hospital. Case 4-2012. A 37-year-old man with muscle pain, weakness, and weight loss. N Engl J Med. 2012;366(6):553-60.
  337. Fisher J. Thyrotoxic periodic paralysis with ventricular fibrillation. Arch Intern Med. 1982;142(7):1362-4.
  338. Siafakas NM, Milona I, Salesiotou V, Filaditaki V, Tzanakis N, Bouros D. Respiratory muscle strength in hyperthyroidism before and after treatment. Am Rev Respir Dis. 1992;146(4):1025-9.
  339. Stein M, Kimbel P, Johnson RL. PULMONARY FUNCTION IN HYPERTHYROIDISM. J Clin Invest. 1961;40(2):348-63.
  340. Massey DG, Becklake MR, McKenzie JM, Bates DV. Circulatory and ventilatory response to exercise in thyrotoxicosis. N Engl J Med. 1967;276(20):1104-12.
  341. Sandler G, Wilson GM. The nature and prognosis of heart disease in thyrotoxicosis. A review of 150 patients treated with 131 I. Q J Med. 1959;28:347-69.
  342. McDevitt DG, Shanks RG, Hadden DR, Montgomery DA, Weaver JA. The role of the thyroid in the control of heart-rate. Lancet. 1968;1(7550):998-1000.
  343. Pietras RJ, Real MA, Poticha GS, Bronsky D, Waldstein SS. Cardiovascular response in hyperthyroidism. The influence of adrenergic-receptor blockade. Arch Intern Med. 1972;129(3):426-9.
  344. DeGroot W, Leonard J, Paley H, Johnson J, Warren J. The importance of autonomic integrity in maintaining the hyperkinetic circulatory dynamics of human hyperthyroidism. J Clin Invest. 1961;40(June):1033.
  345. Auer J, Scheibner P, Mische T, Langsteger W, Eber O, Eber B. Subclinical hyperthyroidism as a risk factor for atrial fibrillation. Am Heart J. 2001;142(5):838-42.
  346. Blizzard JJ, Rupp JJ. Prolongation of the P-R interval as a manifestation of thyrotoxicosis. Jama. 1960;173:1845.
  347. Tayal B, Graff C, Selmer C, Kragholm KH, Kihlstrom M, Nielsen JB, et al. Thyroid dysfunction and electrocardiographic changes in subjects without arrhythmias: a cross-sectional study of primary healthcare subjects from Copenhagen. BMJ Open. 2019;9(6):e023854.
  348. Buccino RA, Spann JFJ, Sonnenblick EH, Braunwald E. Effect of Thyroid State on Myocardial Contractility. Endocrinology. 1968;82(1):191-2.
  349. Reynolds JL, Woody HB. Thyrotoxic mitral regurgitation: a probable form of intrinsic papillary muscle dysfunction. Am J Dis Child. 1971;122(6):544-8.
  350. Rodbard D, Fujita T, Rodbard S. Estimation of thyroid function by timing the arterial sounds. Jama. 1967;201(11):884-7.
  351. Klein I, Danzi S. Thyroid disease and the heart. Circulation. 2007;116(15):1725-35.
  352. Rowe GG, Huston JH, Weinstein AB, Tuchman H, Brown JF, Crumpton CW. The hemodynamics of thyrotoxicosis in man with special reference to coronary blood flow and myocardial oxygen metabolism. J Clin Invest. 1956;35(3):272-6.
  353. Biondi B, Fazio S, Cuocolo A, Sabatini D, Nicolai E, Lombardi G, et al. Impaired cardiac reserve and exercise capacity in patients receiving long-term thyrotropin suppressive therapy with levothyroxine. J Clin Endocrinol Metab. 1996;81(12):4224-8.
  354. Graettinger JS, Muenster JJ, Selverstone LA, Campbell JA. A correlation of clinical and hemodynamic studies in patients with hyperthyroidism with and without congestive heart failure. J Clin Invest. 1959;38(8):1316-27.
  355. Smit JW, Eustatia-Rutten CF, Corssmit EP, Pereira AM, Frölich M, Bleeker GB, et al. Reversible diastolic dysfunction after long-term exogenous subclinical hyperthyroidism: a randomized, placebo-controlled study. J Clin Endocrinol Metab. 2005;90(11):6041-7.
  356. Resnekov L, Falicov RE. Thyrotoxicosis and lactate-producing angina pectoris with normal coronary arteries. Br Heart J. 1977;39(10):1051-7.
  357. Choi YH, Chung JH, Bae SW, Lee WH, Jeong EM, Kang MG, et al. Severe coronary artery spasm can be associated with hyperthyroidism. Coron Artery Dis. 2005;16(3):135-9.
  358. Martí V, Ballester M, Rigla M, Narula J, Bernà L, Pons-Lladó G, et al. Myocardial damage does not occur in untreated hyperthyroidism unless associated with congestive heart failure. Am Heart J. 1997;134(6):1133-7.
  359. Kotler MN, Michaelides KM, Bouchard RJ, Warbasse JR. Myocardial infarction associated with thyrotoxicosis. Arch Intern Med. 1973;132(5):723-8.
  360. Levey GS, Klein I. Catecholamine-thyroid hormone interactions and the cardiovascular manifestations of hyperthyroidism. Am J Med. 1990;88(6):642-6.
  361. Mintz G, Pizzarello R, Klein I. Enhanced left ventricular diastolic function in hyperthyroidism: noninvasive assessment and response to treatment. J Clin Endocrinol Metab. 1991;73(1):146-50.
  362. Valcavi R, Menozzi C, Roti E, Zini M, Lolli G, Roti S, et al. Sinus node function in hyperthyroid patients. J Clin Endocrinol Metab. 1992;75(1):239-42.
  363. Smolenski RT, Yacoub MH, Seymour A-ML. Hyperthyroidism increases adenosine transport and metabolism in the rat heart. Molecular and Cellular Biochemistry. 1995;143(2):143-9.
  364. Brent GA. The molecular basis of thyroid hormone action. N Engl J Med. 1994;331(13):847-53.
  365. Klein I, Ojamaa K. Thyroid hormone and the cardiovascular system. N Engl J Med. 2001;344(7):501-9.
  366. Schmidt BM, Martin N, Georgens AC, Tillmann HC, Feuring M, Christ M, et al. Nongenomic cardiovascular effects of triiodothyronine in euthyroid male volunteers. J Clin Endocrinol Metab. 2002;87(4):1681-6.
  367. Davis PJ, Davis FB. Nongenomic actions of thyroid hormone on the heart. Thyroid. 2002;12(6):459-66.
  368. Hönes GS, Rakov H, Logan J, Liao XH, Werbenko E, Pollard AS, et al. Noncanonical thyroid hormone signaling mediates cardiometabolic effects in vivo. Proc Natl Acad Sci U S A. 2017;114(52):E11323-e32.
  369. Dillmann WH. Biochemical basis of thyroid hormone action in the heart. Am J Med. 1990;88(6):626-30.
  370. Tse J, Wrenn RW, Kuo JF. Thyroxine-induced changes in characteristics and activities of beta-adrenergic receptors and adenosine 3',5'-monophosphate and guanosine 3',5'-monophosphate systems in the heart may be related to reputed catecholamine supersensitivity in hyperthyroidism. Endocrinology. 1980;107(1):6-16.
  371. Williams LT, Lefkowitz RJ, Watanabe AM, Hathaway DR, Besch HR, Jr. Thyroid hormone regulation of beta-adrenergic receptor number. J Biol Chem. 1977;252(8):2787-9.
  372. Fatourechi V, Edwards WD. Graves' disease and low-output cardiac dysfunction: implications for autoimmune disease in endomyocardial biopsy tissue from eleven patients. Thyroid. 2000;10(7):601-5.
  373. Nightingale S, Vitek PJ, Himsworth RL. The haematology of hyperthyroidism. Q J Med. 1978;47(185):35-47.
  374. Viherkoski M, Lamberg BA. The Glucose-6-Phosphate Dehydrogenase Activity (G-6-PD) of the Red Blood Cells in Hyperthyroidism and Hypothyroidism. Scandinavian Journal of Clinical and Laboratory Investigation. 1970;25(2):137-43.
  375. Popovic WJ, Brown JE, Adamson JW. The influence of thyroid hormones on in vitro erythropoiesis. Mediation by a receptor with beta adrenergic properties. J Clin Invest. 1977;60(4):907-13.
  376. Rivlin RS, Wagner HN, Jr. Anemia in hyperthyroidism. Ann Intern Med. 1969;70(3):507-16.
  377. Bergman TA, Mariash CN, Oppenheimer JH. Anterior mediastinal mass in a patient with Graves' disease. J Clin Endocrinol Metab. 1982;55(3):587-8.
  378. Huang W, Molitch ME. Enlarged thymus in a patient with dyspnea and weight loss. Jama. 2015;313(21):2174-5.
  379. Irvine WJ, Wu FC, Urbaniak SJ, Toolis F. Peripheral blood leucocytes in thyrotoxicosis (Graves' disease) as studied by conventional light microscopy. Clin Exp Immunol. 1977;27(2):216-21.
  380. Hertz S, Lerman J. THE BLOOD PICTURE IN EXOPHTHALMIC GOITRE AND ITS CHANGES RESULTING FROM IODINE AND OPERATION. A STUDY BY MEANS OF THE SUPRAVITAL TECHNIQUE. J Clin Invest. 1932;11(6):1179-96.
  381. Lima CS, Zantut Wittmann DE, Castro V, Tambascia MA, Lorand-Metze I, Saad ST, et al. Pancytopenia in untreated patients with Graves' disease. Thyroid. 2006;16(4):403-9.
  382. Lamberg BA, Kivikangas V, Pelkonen R, Vuopio P. Thrombocytopenia and decreased life-span of thrombocytes in hyperthyroidism. Ann Clin Res. 1971;3(2):98-102.
  383. Adrouny A, Sandler RM, Carmel R. Variable presentation of thrombocytopenia in Graves' disease. Arch Intern Med. 1982;142(8):1460-4.
  384. Verberne HJ, Fliers E, Prummel MF, Stam J, Brandjes DP, Wiersinga WM. Thyrotoxicosis as a predisposing factor for cerebral venous thrombosis. Thyroid. 2000;10(7):607-10.
  385. Davis PJ, Mousa SA, Schechter GP. New Interfaces of Thyroid Hormone Actions With Blood Coagulation and Thrombosis. Clinical and Applied Thrombosis/Hemostasis. 2018;24(7):1014-9.
  386. Wegener M, Wedmann B, Langhoff T, Schaffstein J, Adamek R. Effect of hyperthyroidism on the transit of a caloric solid-liquid meal through the stomach, the small intestine, and the colon in man. J Clin Endocrinol Metab. 1992;75(3):745-9.
  387. Lerman J, Means JH. THE GASTRIC SECRETION IN EXOPHTHALMIC GOITRE AND MYXOEDEMA. J Clin Invest. 1932;11(1):167-82.
  388. Berryhill WR, Williams HA. A STUDY OF THE GASTRIC SECRETION IN HYPERTHYROIDISM BEFORE AND AFTER OPERATION. J Clin Invest. 1932;11(4):753-60.
  389. Siurala M, Lamberg BA. Stomach in thyrotoxicosis. Acta Med Scand. 1959;165:181-8.
  390. Seino Y, Matsukura S, Miyamoto Y, Goto Y, Taminato T, Imura H. Hypergastrinemia in hyperthyroidism. J Clin Endocrinol Metab. 1976;43(4):852-5.
  391. Scappaticcio L, Longo M, Maiorino MI, Pernice V, Caruso P, Esposito K, et al. Abnormal Liver Blood Tests in Patients with Hyperthyroidism: Systematic Review and Meta-Analysis. Thyroid. 2021;31(6):884-94.
  392. Lamberg BA, Gordin R. Liver Function in Thyrotoxicosis: Studies on the Cholesterol and Prothrombin Level in the Blood During the Treatment of Thyrotoxicosis. Acta Endocrinologica (Norway). 1954;15(1):82-96.
  393. Thompson P, Jr., Strum D, Boehm T, Wartofsky L. Abnormalities of liver function tests in tyrotoxicosis. Mil Med. 1978;143(8):548-51.
  394. Greenberger NJ, Milligan FD, Degroot LJ, Isselbacher KJ. JAUNDICE AND THYROTOXICOSIS IN THE ABSENCE OF CONGESTIVE HEART FAILURE. A STUDY OF FOUR CASES. Am J Med. 1964;36:840-6.
  395. Kubota S, Amino N, Matsumoto Y, Ikeda N, Morita S, Kudo T, et al. Serial changes in liver function tests in patients with thyrotoxicosis induced by Graves' disease and painless thyroiditis. Thyroid. 2008;18(3):283-7.
  396. Yorke E. Hyperthyroidism and Liver Dysfunction: A Review of a Common Comorbidity. Clin Med Insights Endocrinol Diabetes. 2022;15:11795514221074672.
  397. de Campos Mazo DF, de Vasconcelos GB, Pereira MA, de Mello ES, Bacchella T, Carrilho FJ, et al. Clinical spectrum and therapeutic approach to hepatocellular injury in patients with hyperthyroidism. Clin Exp Gastroenterol. 2013;6:9-17.
  398. Youssef WI, Mullen KD. The liver in other (nondiabetic) endocrine disorders. Clin Liver Dis. 2002;6(4):879-89, vii.
  399. Piantanida E, Ippolito S, Gallo D, Masiello E, Premoli P, Cusini C, et al. The interplay between thyroid and liver: implications for clinical practice. J Endocrinol Invest. 2020;43(7):885-99.
  400. Epstein FH, Rivera MJ. Renal concentrating ability in thyrotoxicosis. J Clin Endocrinol Metab. 1958;18(10):1135-7.
  401. Ford RV, Owens JC, Curd GW, Jr., Moyer JH, Spurr CL. Kidney function in various thyroid states. J Clin Endocrinol Metab. 1961;21:548-53.
  402. Sataline LR, Powell C, Hamwi GJ. Suppression of the hypercalcemia of thyrotoxicosis by corticosteroids. N Engl J Med. 1962;267:646-50.
  403. Sallin O. Hypercalcemic Nephropathy in Thyrotoxicosis. Acta Endocrinologica (Norway). 1958;29(3):425-34.
  404. Goldsmith RE, Sturgis SH, Lerman J, Stanbury JB. The menstrual pattern in thyroid disease. J Clin Endocrinol Metab. 1952;12(7):846-55.
  405. Sanjari M, Safi Z, Tahroodi KM. HYPERTHYROIDISM AND HYPERPROLACTINEMIA: IS THERE ANY ASSOCIATION? Endocr Pract. 2016;22(12):1377-82.
  406. Freedberg IM, Hamolsky MW, Freedberg AS. The thyroid gland in pregnancy. N Engl J Med. 1957;256(11):505-10; contd.
  407. De Groot L, Abalovich M, Alexander EK, Amino N, Barbour L, Cobin RH, et al. Management of thyroid dysfunction during pregnancy and postpartum: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2012;97(8):2543-65.
  408. Polak M, Le Gac I, Vuillard E, Guibourdenche J, Leger J, Toubert ME, et al. Fetal and neonatal thyroid function in relation to maternal Graves' disease. Best Pract Res Clin Endocrinol Metab. 2004;18(2):289-302.
  409. Anselmo J, Cao D, Karrison T, Weiss RE, Refetoff S. Fetal loss associated with excess thyroid hormone exposure. Jama. 2004;292(6):691-5.
  410. Southren AL, Olivo J, Gordon GG, Vittek J, Brener J, Rafii F. The conversion of androgens to estrogens in hyperthyroidism. J Clin Endocrinol Metab. 1974;38(2):207-14.
  411. Chopra IJ, Tulchinsky DAN. Status of Estrogen-Androgen Balance in Hyperthyroid Men with Graves' Disease. The Journal of Clinical Endocrinology & Metabolism. 1974;38(2):269-77.
  412. Chopra IJ, Abraham GE, Chopra U, Solomon DH, Odell WD. Alterations in circulating estradiol-17-beta in male patients with Graves's disease. N Engl J Med. 1972;286(3):124-9.
  413. Becker KL, Winnacker JL, Matthews MJ, Higgins GA, Jr. Gynecomastia and hyperthyroidism. An endocrine and histological investigation. J Clin Endocrinol Metab. 1968;28(2):277-85.
  414. Bercovici JP, Mauvais-Jarvis P. Hyperthyroidism and gynecomastia: metabolic studies. J Clin Endocrinol Metab. 1972;35(5):671-7.
  415. Kidd GS, Glass AR, Vigersky RA. The hypothalamic-pituitary-testicular axis in thyrotoxicosis. J Clin Endocrinol Metab. 1979;48(5):798-802.
  416. Abalovich M, Levalle O, Hermes R, Scaglia H, Aranda C, Zylbersztein C, et al. Hypothalamic-pituitary-testicular axis and seminal parameters in hyperthyroid males. Thyroid. 1999;9(9):857-63.
  417. Krassas GE, Perros P. Thyroid disease and male reproductive function. J Endocrinol Invest. 2003;26(4):372-80.
  418. Peterson RE. The influence of the thyroid on adrenal cortical function. J Clin Invest. 1958;37(5):736-43.
  419. Kenny FM, Iturzaeta N, Preeyasombat C, Taylor FH, Migeon CJ, Richards C. Cortisol Production Rate. VII. Hypothyroidism and Hyperthyroidism in Infants and Children. The Journal of Clinical Endocrinology & Metabolism. 1967;27(11):1616-22.
  420. Toro-Tobon D, Bancos I, Brito JP. Abstract #1408256: In Search of New Biomarkers of Thyroid Function: The Association Between Thyroid Dysfunction and 11-beta Hydroxysteroid Dehydrogenase 2 Activity. Endocrine Practice. 2023;29(5):S116.
  421. Hellman L, Bradlow HL, Zumoff B, Gallagher TF. The influence of thyroid hormone on hydrocortisone production and metabolism. J Clin Endocrinol Metab. 1961;21:1231-47.
  422. Gallagher TF, Hellman L, Finkelstein J, Yoshida K, Weitzman ED, Roffwarg HD, et al. Hyperthyroidism and cortisol secretion in man. J Clin Endocrinol Metab. 1972;34(6):919-27.
  423. Hilton JG, Black WC, Athos W, Mc HB, Westermann CD. Increased ACTH-like activity in plasma of patients with thyrotoxicosis. J Clin Endocrinol Metab. 1962;22:900-5.
  424. Felber J-P, Reddy WJ, Selenkow HA, Thorn GW. ADRENOCORTICAL RESPONSE TO THE 48-HOUR ACTH TEST IN MYXEDEMA AND HYPERTHYROIDISM*. The Journal of Clinical Endocrinology & Metabolism. 1959;19(8):895-906.
  425. Hellman L, Bradlow HL, Zumoff B, Fukushima DK, Gallagher TF. Thyroid-androgen interrelations and the hypocholesteremic effect of androsterone. J Clin Endocrinol Metab. 1959;19:936-48.
  426. Hoshiro M, Ohno Y, Masaki H, Iwase H, Aoki N. Comprehensive study of urinary cortisol metabolites in hyperthyroid and hypothyroid patients. Clinical Endocrinology. 2006;64(1):37-45.
  427. Fraser SA, Anderson JB, Smith DA, Wilson GM. Osteoporosis and fractures following thyrotoxicosis. Lancet. 1971;1(7707):981-3.
  428. Rosen CJ, Adler RA. Longitudinal changes in lumbar bone density among thyrotoxic patients after attainment of euthyroidism. J Clin Endocrinol Metab. 1992;75(6):1531-4.
  429. Langdahl BL, Loft AG, Eriksen EF, Mosekilde L, Charles P. Bone mass, bone turnover, body composition, and calcium homeostasis in former hyperthyroid patients treated by combined medical therapy. Thyroid. 1996;6(3):161-8.
  430. Jódar E, Martínez-Díaz-Guerra G, Azriel S, Hawkins F. Bone mineral density in male patients with L-thyroxine suppressive therapy and Graves disease. Calcif Tissue Int. 2001;69(2):84-7.
  431. Uzzan B, Campos J, Cucherat M, Nony P, Boissel JP, Perret GY. Effects on bone mass of long term treatment with thyroid hormones: a meta-analysis. J Clin Endocrinol Metab. 1996;81(12):4278-89.
  432. Krane SM, Brownell GL, Stanbury JB, Corrigan H. The effect of thyroid disease on calcium metabolism in man. J Clin Invest. 1956;35(8):874-87.
  433. Harvey RD, McHardy KC, Reid IW, Paterson F, Bewsher PD, Duncan A, et al. Measurement of bone collagen degradation in hyperthyroidism and during thyroxine replacement therapy using pyridinium cross-links as specific urinary markers. J Clin Endocrinol Metab. 1991;72(6):1189-94.
  434. Lukert BP, Higgins JC, Stoskopf MM. Serum osteocalcin is increased in patients with hyperthyroidism and decreased in patients receiving glucocorticoids. J Clin Endocrinol Metab. 1986;62(5):1056-8.
  435. Cooper DS, Kaplan MM, Ridgway EC, Maloof F, Daniels GH. Alkaline phosphatase isoenzyme patterns in hyperthyroidism. Ann Intern Med. 1979;90(2):164-8.
  436. Siddiqi A, Burrin JM, Noonan K, James I, Wood DF, Price CP, et al. A longitudinal study of markers of bone turnover in Graves' disease and their value in predicting bone mineral density. J Clin Endocrinol Metab. 1997;82(3):753-9.
  437. Garrel DR, Delmas PD, Malaval L, Tourniaire J. Serum bone Gla protein: a marker of bone turnover in hyperthyroidism. J Clin Endocrinol Metab. 1986;62(5):1052-5.
  438. Epstein FH, Freedman LR, Levitin H. Hypercalcemia, nephrocalcinosis and reversible renal insufficiency associated with hyperthyroidism. N Engl J Med. 1958;258(16):782-5.
  439. Peerenboom H, Keck E, Krüskemper HL, Strohmeyer G. The defect of intestinal calcium transport in hyperthyroidism and its response to therapy. J Clin Endocrinol Metab. 1984;59(5):936-40.
  440. Bouillon R, De Moor P. Parathyroid function in patients with hyper- or hypothyroidism. J Clin Endocrinol Metab. 1974;38(6):999-1004.
  441. Bouillon R, Muls E, De Moor P. Influence of thyroid function on the serum concentration of 1,25-dihydroxyvitamin D3. J Clin Endocrinol Metab. 1980;51(4):793-7.
  442. Bortz W, Eisenberg E, Bowers CY, Pont M. DIFFERENTIATION BETWEEN THYROID AND PARATHYROID CAUSES OF HYPERCALCEMIA. Annals of Internal Medicine. 1961;54(4):610-9.
  443. David NJ, Verner JV, Engel FL. The diagnostic spectrum of hypercalcemia. Case reports and discussion. Am J Med. 1962;33:88-110.
  444. Kleeman CR, Tuttle S, Bassett SH. METABOLIC OBSERVATIONS IN A CASE OF THYROTOXICOSIS WITH HYPERCALCEMIA*. The Journal of Clinical Endocrinology & Metabolism. 1958;18(5):477-91.
  445. Danforth E, Jr., Burger A. The role of thyroid hormones in the control of energy expenditure. Clin Endocrinol Metab. 1984;13(3):581-95.
  446. Silva JE. The thermogenic effect of thyroid hormone and its clinical implications. Ann Intern Med. 2003;139(3):205-13.
  447. Yavuz S, Salgado Nunez del Prado S, Celi FS. Thyroid Hormone Action and Energy Expenditure. Journal of the Endocrine Society. 2019;3(7):1345-56.
  448. Plummer H, Boothby W. The cost of work in exophthalmic goiter. Am J Physiol. 1923;63(406):1922-3.
  449. Briard SP, McClintock JT, Baldridge CW. COST OF WORK IN PATIENTS WITH HYPERMETABOLISM DUE TO LEUKEMIA AND TO EXOPHTHALMIC GOITER. Archives of Internal Medicine. 1935;56(1):30-7.
  450. Acheson K, Jéquier E, Burger A, Danforth E, Jr. Thyroid hormones and thermogenesis: the metabolic cost of food and exercise. Metabolism. 1984;33(3):262-5.
  451. Potenza M, Via MA, Yanagisawa RT. Excess Thyroid Hormone and Carbohydrate Metabolism. Endocrine Practice. 2009;15(3):254-62.
  452. Eom YS, Wilson JR, Bernet VJ. Links between Thyroid Disorders and Glucose Homeostasis. Diabetes Metab J. 2022;46(2):239-56.
  453. Mitrou P, Raptis SA, Dimitriadis G. Insulin Action in Hyperthyroidism: A Focus on Muscle and Adipose Tissue. Endocrine Reviews. 2010;31(5):663-79.
  454. Kabadi UM, Eisenstein AB. Impaired pancreatic alpha-cell response in hyperthyroidism. J Clin Endocrinol Metab. 1980;51(3):478-82.
  455. Wennlund A, Arner P, Ostman J. Changes in the effects of insulin on human adipose tissue metabolism in hyperthyroidism. J Clin Endocrinol Metab. 1981;53(3):631-5.
  456. Houssay BA. THYROID AND METATHYROID DIABETES. Endocrinology. 1944;35(3):158-72.
  457. Bratusch-Marrain PR, Komjati M, Waldhäusl WK. Glucose metabolism in noninsulin-dependent diabetic patients with experimental hyperthyroidism. J Clin Endocrinol Metab. 1985;60(6):1063-8.
  458. Elrick H, Hlad CJ, Jr., Arai Y. Influence of thyroid function on carbohydrate metabolism and a new method for assessing response to insulin. J Clin Endocrinol Metab. 1961;21:387-400.
  459. Kritchevsky D. Influence of thyroid hormones and related compounds on cholesterol biosynthesis and degradation: a review. Metabolism. 1960;9:984-94.
  460. Siperstein MD, Murray AW. Cholesterol metabolism in man. J Clin Invest. 1955;34(9):1449-53.
  461. O'Brien T, Katz K, Hodge D, Nguyen TT, Kottke BA, Hay ID. The effect of the treatment of hypothyroidism and hyperthyroidism on plasma lipids and apolipoproteins AI, AII and E. Clin Endocrinol (Oxf). 1997;46(1):17-20.
  462. Chait A, Bierman EL, Albers JJ. Regulatory role of triiodothyronine in the degradation of low density lipoprotein by cultured human skin fibroblasts. J Clin Endocrinol Metab. 1979;48(5):887-9.
  463. Tulloch BR, Vydelingum N, Lewis B, Fraser R. Triglyceride metabolism in thyroid disease. Clin Sci. 1973;44(2):6P passim.
  464. Cachefo A, Boucher P, Vidon C, Dusserre E, Diraison F, Beylot M. Hepatic lipogenesis and cholesterol synthesis in hyperthyroid patients. J Clin Endocrinol Metab. 2001;86(11):5353-7.
  465. Arons DL, Schreibman PH, Downs P, Braverman LE, Arky RA. Decreased post-heparin lipases in Graves's disease. N Engl J Med. 1972;286(5):233-7.
  466. Sachs BA, Danielson E, Isaacs MC, Weston RE. Effect of triiodothyronine on the serum lipids and lipoproteins of euthyroid and hyperthyroid subjects. J Clin Endocrinol Metab. 1958;18(5):506-15.
  467. Kung AW, Pang RW, Lauder I, Lam KS, Janus ED. Changes in serum lipoprotein(a) and lipids during treatment of hyperthyroidism. Clin Chem. 1995;41(2):226-31.
  468. Ozata M, Uckaya G, Bolu E, Corapcioglu D, Bingol N, Ozdemir IC. Plasma leptin concentrations in patients with Graves' disease with or without ophthalmopathy. Med Sci Monit. 2001;7(4):696-700.
  469. Strisower B, Elmlinger P, Gofman JW, Delalla O. The effect of 1-thyroxine on serum lipoprotein and cholesterol concentrations. J Clin Endocrinol Metab. 1959;19(1):117-26.
  470. Postel S. Total free tocopherols in the serum of patients with thyroid disease. J Clin Invest. 1956;35(12):1345-56.
  471. Rich C, Bierman EL, Schwartz IL. Plasma nonesterified fatty acids in hyperthyroid states. J Clin Invest. 1959;38(2):275-8.
  472. Riis AL, Jørgensen JO, Ivarsen P, Frystyk J, Weeke J, Møller N. Increased protein turnover and proteolysis is an early and primary feature of short-term experimental hyperthyroidism in healthy women. J Clin Endocrinol Metab. 2008;93(10):3999-4005.
  473. Lewallen CG, Rall JE, Berman M. Studies of iodoalbumin metabolism. II. The effects of thyroid hormone. J Clin Invest. 1959;38(1, Part 1):88-101.
  474. Kivirikko KI, Laitinen O, Aer J, Halme J. Metabolism of collagen in experimental hyperthyroidism and hypothyroidism in the rat. Endocrinology. 1967;80(6):1051-61.
  475. Crispell KR, Parson W, Hollifield G. A study of the rate of protein synthesis before and during the administration of L-triiodothyronine to patients with myxedema and healthy volunteers using N-15 glycine. J Clin Invest. 1956;35(2):164-9.
  476. Sokoloff L, Kaufman S. Effects of thyroxin on amino acid incorporation into protein. Science. 1959;129(3348):569-70.
  477. Sokoloff L, Roberts PA, Januska MM, Kline JE. Mechanisms of stimulation of protein synthesis by thyroid hormones in vivo. Proc Natl Acad Sci U S A. 1968;60(2):652-9.
  478. Aktuna D, Buchinger W, Langsteger W, Meister E, Sternad H, Lorenz O, et al. [Beta-carotene, vitamin A and carrier proteins in thyroid diseases]. Acta Med Austriaca. 1993;20(1-2):17-20.
  479. Wohl MG, Levy HA, Szutka A, Maldia G. Pyridoxine deficiency in hyperthyroidism. Proc Soc Exp Biol Med. 1960;105:523-7.