Archives

Exercise Treatment of Obesity

ABSTRACT

 

Surveillance data from the general US population indicate a continued increase in the prevalence of overweight and obesity that is consistent with weight gain trends observed globally among industrialized countries. Physical inactivity and obesity are closely linked conditions and they account for a large burden of chronic disease and impaired function. The underlying agent in the etiology of obesity is a long-term positive energy balance; however, the pathways determining the rate and extent of weight gain due to a positive energy are complex. Engaging in regular, moderate-intensity physical activity for at least 150 min/week can help maintain energy balance and prevent excessive weight gain; however, this minimum requirement may not be sufficient for reversing already-existing obesity and chronic disease. In fact, physical activity closer to 300 min/week may be necessary for successful weight loss and weight loss maintenance. Regimented exercise programs alone may not be the most effective treatment for people with obesity, however. Rather, lifestyle changes that increase total daily energy expenditure need to be accompanied by dietary counseling for reducing daily caloric intake. Also, accumulating the necessary exercise and lifestyle physical activity in intermittent bouts, rather than one long continuous bout, can improve adherence and the success of weight loss regimens. It is also important for both clinicians and patients to understand that a simple solution to obesity treatment does not exist due to the constellation of underlying mechanisms that drive energy balance. Indeed, physiological, behavioral, environmental, and genetic factors play both independent and interrelated roles that contribute to the complex etiology of obesity.  Research from numerous scientific disciplines has shaped our understanding of obesity. While the relative contributions of insufficient energy expenditure versus excessive energy intake to obesity development continue to be debated, there is general agreement that exercise is a key element for both prevention and treatment. Future research should focus on the prevention of excess weight gain over the life course. In addition to the behavioral and intervention studies of the past several decades, an understanding of the regulatory processes governing energy intake, energy storage, and energy expenditure and how the reinstatement of exercise can correct the disruption of neural pathways is vital to the future of obesity research. Finally, public health science needs to link with public health practice to better enable the translation of this knowledge into policies that can alter the environment in a way that promotes an active lifestyle for all.

 

INTRODUCTION

 

Surveillance data from the general US population indicate a continued increase in the prevalence of overweight and obesity that is consistent with weight gain trends observed globally among industrialized countries (1-3). Myriad environmental, behavioral, physiological, and genetic factors contribute to the development of human obesity. However, the common underlying feature leading to these conditions is a positive energy balance. Attenuated metabolic responses to environmental exposures combined with predisposing factors and overall low energy expenditure may contribute to this positive energy balance. Although exercise is most effective in the prevention of obesity (4, 5), it can also contribute to weight loss and to weight maintenance over the long-term. Numerous intervention studies have evaluated the role of exercise training of various modes and intensities on the reduction of body weight and adiposity (6), and there is little doubt about the established benefits of increasing physical activity to the attainment and the maintenance of healthy body weight throughout the life span. Moreover, since exercise itself improves metabolic, respiratory, and cardiovascular function independent of weight loss (6), it has special significance for people with obesity who are at increased risk for obesity-related chronic conditions. In this chapter, we will describe the importance of exercise for the prevention and treatment of obesity, as well as to the prevention of weight regain following weight-loss therapy. In addition, this chapter will address the contributions of the built environment to the onset and possible reversal of obesity at the population level.

 

THE ETIOLOGY OF OBESITY

 

Inactivity and obesity are closely linked conditions accounting for a large burden of chronic disease and impaired function. Over the past several decades, ever-decreasing levels of daily energy expenditure, along with a ready supply of calorie-dense foods, have resulted in a marked disruption to energy regulatory systems, which are still genetically programmed for the subsistence efficiency of our late-Paleolithic ancestors (7, 8). As stated previously, the underlying agent in the etiology of obesity is a long-term positive energy balance. However, the relative importance of excess energy intake over low energy expenditure to this imbalance is controversial. Ultimately, the pathways determining the rate and extent of a positive energy balance with weight gain are complex, and the unique and combined contributions of heredity, physiology, and behavior, to the development of obesity are not understood completely—especially since the influence of any one of these primary factors is usually modified by a constellation of other secondary factors endemic to our current obesogenic environment (Figure 1).

Figure 1. Public health model illustrating the multifactorial model etiology of obesity. The traditional public health of disease transmission applied to obesity etiology. In this model, the impact of the agent (positive energy balance) can be modified by a number of host (specific to the individual) and environment (specific to collective behaviors or conditions) factors. In addition, a variety of vehicles/vectors are responsible for transmitting the causal agent.

 

The 2018 Physical Activity Guidelines for Americans, 2nd Edition (6), along with the 2020 World Health Organization (WHO) Physical Activity and Sedentary Behavior Guidelines (9) recommend for all adults 150-300 min/week of moderate-intensity physical activity (e.g., brisk walking) or 75-150 min/week of vigorous-intensity activity for the prevention of excessive weight gain, cardiovascular and metabolic diseases, and functional decline. These recommendations also include muscle strengthening exercises on two days/week. Although specific recommendations pertaining to sedentary behavior have not been made thus far, the evidence linking extended sedentary time to morbidity and all-cause mortality is growing (6). Indeed, both the 2018 Guidelines for Americans and the WHO Guidelines stress that everyone should "move more and sit less" (6) and "every move counts" (9). Importantly, current guidelines now stress the joint association between physical activity and sedentary time. For example, the health impact of sedentary behavior (particularly television viewing) becomes especially detrimental when combined with low levels of physical activity (6, 10). People can compensate for large amounts of sedentary time during the day (i.e., 8–14 h) by increasing their physical activity to achieve at least 30 min of accumulated moderate-intensity activity throughout the day. However, the more sedentary one is, the more accumulated activity is necessary to compensate (Figure 2).

Figure 2. The joint effects of physical activity on health and function. The red zone is harmful, while the green zone is healthful, suggesting that the more sedentary one is, the more accumulated physical activity they need to compensate.

The Role of Exercise in Weight Loss and Weight Maintenance

 

Population physical activity guidelines may be more effective for health promotion and the primary prevention of chronic disease risk factors than they are for the reversal of already established chronic conditions. Although increasing physical activity and reducing sedentary time has demonstrated benefits to improved health and function, even among people with chronic disease or with disabilities (6, 9), it is important to note that the minimum recommendation of 150 min/week of moderate-intensity physical activity, may not be sufficient to reverse these chronic conditions. Indeed, the treatment or reversal of some established conditions may require a dose of physical activity closer to 300 min/week. This may be especially true for the reversal of obesity and for weight loss maintenance. Although population- and laboratory-based data are limited, it appears that about 45–60 min/day of moderate-intensity activity is necessary to transition from overweight to normal weight, and ≥ 60 min/day may be necessary to transition from obesity (11-14), at least for a large part of the population with overweight and obesity who spend considerable time sitting throughout the day. In addition, there is substantial individual heterogeneity regarding a person's weight loss responsiveness to an exercise regimen, and this responsiveness may vary by age, sex, degree of obesity, adipose tissue distribution, and even adipocyte size (15-17). Thus, the benefits of increased physical activity to cardiovascular and metabolic health notwithstanding, its effectiveness per se for substantial weight loss and in the reversal of obesity may be less so.

 

Weight loss of 1–2 pounds (0.5–1 kg) per week is generally recognized as safe and effective (18). Weight loss at this recommended rate, however, would require a negative energy balance of ~ 500–1000 kcal/day over an extended period of time. Such an energy deficit is difficult to achieve by lowering energy intake (dieting) alone. More importantly, such drastic decreases in caloric intake could result in nutritional deficiencies and the loss of lean mass, thereby lowering the metabolic rate (19). Also, adherence to such a degree of caloric restriction is difficult to maintain over long periods of time and, therefore, increases the likelihood of relapse and compensatory weight re-gain.

 

On the other hand, whether exercise alone (without coincident caloric restriction) significantly alters body weight in people with obesity is debatable. Assuming that 60 min/day of moderate-intensity activity is necessary for meaningful weight loss for people with obesity, a man would need to perform 68–136 min/day of moderate-intensity walking (7.9 kcal/min), and a woman may have to perform 72–145 min/day of the same activity (6.4 kcal/min) to achieve 500-1000 kcal/day deficit necessary for a weight loss of 1–2 pounds (0.5–1 kg)/week (20). Further, although this walking pace (3.5 mph or 3.8 METs) may be comfortable for most people, sustaining it for over 60 min on 7 days/week may not be feasible for people with obesity. Indeed, it may be quite difficult for people with obesity to perform the volume (i.e., intensity, frequency and duration) of exercise necessary for meaningful weight loss in the absence of caloric restriction. Therefore, most evidence currently indicates that both exercise and caloric restriction are necessary components of a successful weight loss program.

 

People who are successful in losing substantial amounts of body weight through diet alone often quickly regain it. Weight regain is often seen following exercise-, medication-, and even surgery-induced weight loss, indicating that adaptations to a negative energy balance contribute to the obesity epidemic. Laboratory findings report that the level of daily energy expenditure necessary to prevent the re-gain of body weight following obesity is also quite high relative to the modern-day lifestyle (17). This challenge may be the result of changes in body composition or the body's overall adaptive energy expenditure and metabolic response to exercise that limits weight loss to activity alone (8, 21) (Figure 3). The 2003 consensus statement from the International Association for the Study of Obesity (14) recommended 60–90 min/day of moderate-intensity activity or about 35 min/day of vigorous activity for successful weight maintenance following the reversal of obesity, which, again, exceeds the upper threshold of current physical activity recommendations (6, 9).

Figure 3. Changes in Total energy expenditure ADJ, resting metabolic rate ADJ, and activity energy (CPM/d), (right, Pontzer (8), with permission are consistent with the findings shown in the schematic of exercise impact on body weight demonstrating a new equilibrium after an initial weight loss (left (21)), with permission

In sum, caloric restriction without exercise may result in a loss of lean mass along with adipose tissue, thereby resulting in a drop in the metabolic rate and setting the stage for weight re-gain. The amount of daily exercise that is necessary to achieve a healthy weight loss without caloric restriction may not be feasible over time for people with obesity, thus again resulting in relapse. Most research now supports the conclusion that exercise combined with caloric restriction increases the net caloric deficit induced by a weight loss program and markedly attenuates the loss of both fat-free and total body mass (19). Finally, as is the case through the period of dynamic weight loss, those who combine caloric restriction with exercise are more successful in maintaining that weight loss over time, compared with those relying on either diet or exercise alone.

 

THE ROLE OF RESISTANCE TRAINING FOR WEIGHT LOSS AND MAINTENANCE

 

Both aerobic and resistance exercise will preserve lean tissue during the period of dynamic weight loss, and this is primarily a function of the volume of exercise performed over the weight-loss period (i.e., dose-response). Resistance training is especially effective at preserving lean body mass during dynamic weight loss, although the amount of protein in the diet may impact this effectiveness (22). A program that combines caloric restriction with both aerobic and resistance training generally leads to greater weight loss and improved overall health, compared to a program combining caloric restriction with only aerobic exercise (11, 13). Of note is that the benefits of strength training to health and function can be independent of weight loss. For example, one 5-month study in older men and women with obesity that used both caloric restriction and resistance training led to reduced abdominal obesity, reduced hypertension, and improved metabolic syndrome without any changes in body weight (23). This is likely due to the increase in lean mass with resistance training, as well as the resulting quantitative and qualitative improvements in vascular and muscle function. Another study of older adults with obesity combined caloric restriction with one of three other exercise interventions: 1) aerobic exercise alone; 2) resistance training alone and 3) aerobic exercise and resistance training. Total body weight loss was similar across the three different exercise groups. However, the greatest improvements in measures of physical function were observed in the combined aerobic exercise with resistance training group (24). Thus, the benefits of resistance training extend beyond fat loss to include improved metabolic and physical function—and this may be especially so for older people.

 

THE ROLE OF TOTAL DAILY ACTIVITY IN WEIGHT MAINTENANCE

 

Evidence suggests that total daily accumulated energy expenditure is the strongest predictor of weight loss in people with obesity (25-27). Therefore, an alternative to the typical recommendation of large continuous bouts of exercise may be intermittent exercise, which can result in a similar weight loss but with improved adherence over the long-term. Also, the integration of increased physical activity as part of an overall lifestyle change (e.g., more walking and stair climbing as part of the daily routine) may be as successful in promoting weight loss as is a structured exercise program. Given the high degree of negative energy balance required for weight loss, however, high levels of lifestyle activity combined with caloric restriction are now prescribed for both initial and long-term weight loss for people with overweight and obesity.

 

The Physical Activity Level (PAL) has become a method of expressing total daily energy expenditure (TEE) in multiples of the resting metabolic rate (RMR: PAL = TEE/RMR), and thus far, few studies have examined its relation to weight regulation at the population level. Data from men in the Aerobics Center Longitudinal Study cohort indicate that a daily PAL >1.60 METs·24 h-1 (i.e., an average daily TEE 60% above RMR) is optimal for preventing meaningful weight gain (~ 0.82–0.91 kg·y-1 (13)) through middle-age (4). Moreover, increasing daily activity from the low PAL category (<1.46 METs·24 h-1) to the moderate (1.46–1.60 METs·24 h-1) or high (>1.60 METs·24 h-1) categories resulted in a slight weight loss over time in this cohort (Figure 4).

Figure 4. Predicted weight change over time by PAL change category among men in the Aerobic Center Longitudinal Study (ACLS) cohort. PAL=average daily physical activity level expressed as the ratio of total energy expenditure to the resting metabolic rate (TEE/RMR). Models adjusted for age, sex, height, baseline weight, and smoking. DiPietro, et al. Int J Obesity. 28:1541-1547,2004 (4)

The most useful strategy for accomplishing this average level of daily physical activity is exchanging passive or very low intensity activities (i.e., those involving sitting) for moderate-intensity activities that have energy requirements of about 3–6 METs. Moderate-intensity activities may have a substantially greater impact on the PAL than vigorous activities since vigorous activity is usually performed for very short periods of time and then can be compensated for by reduced volitional activity throughout the remainder of the day (28). Therefore, the best way to increase the average daily PAL from sedentary (1.5 METs·24 h-1) to active (>1.6 METs·24 h-1) is to add about 45–60 minutes of moderate-intensity activity to the daily routine. As described above, using either a continuous or intermittent exercise routine is equally effective in increasing overall TEE.

 

THE IMPACT OF WEARABLE DEVICES

 

In 2014, 10% of adult Americans over the age of 18 years reported owning an activity tracking device, and by 2016, the Worldwide Survey of Fitness Trends identified wearable technology as the most popular growing fitness trend, estimating the market to be around $6 billion (29). This survey was recently updated, reporting that wearable technology remained the number one trend for 2020, and the market reached an estimated $95 billion (30). Most large technology companies have incorporated activity monitoring technology into cellular phones, while larger corporations, including Apple and Google, have continued to expand their product lines to feature new models of watches, wristbands and other clothing devices with activity tracking capabilities. The most popular and affordable devices remain somewhat restricted to measuring step count and distance traveled.

 

New products are constantly in development given the high demand. Even though technological advancements have reportedly improved these devices, debate among product engineers, research scientists and others involved in this industry regarding their accuracy still persists. Data indicate that these devices are less consistent with the measurement of overall activity duration, energy expenditure, and sleep quality, so they may require further testing and more advanced algorithms before being used in research (31). Advanced devices are in development that are capable of measuring biometric signs, such as stress, strain, impact forces, in addition to metabolic parameters (e.g., glucose and lactic acid) and the tracking of physical activity (32).

 

Despite some limitations, such devices are quite useful in helping people to monitor their own daily caloric intake, energy expenditure, sleep patterns, and overall health profile. These devices may also serve to increase motivation among those starting an exercise program because they can help to set goals and provide immediate feedback, although whether or not this is so for long-term weight loss programs is questionable (32). Ideally, such devices can sync with the electronic health record (EHR), thereby allowing health care providers a chance to objectively monitor a patient's lifestyle behaviors.

 

PERSONAL AND ONLINE TRAINING

 

Personal training has remained in the top 10 fitness trends reported since 2006, and popularity has increased as online training has become more accessible (30), especially during the strict quarantine policies imposed during the COVID-19 pandemic in 2020. It is reasonable to suspect that there will be a continued use of online training programs in 2021 and beyond. Unfortunately, like wearable devices, training fees and internet access may be luxuries not available to low-income households, and although some communities have facilities that provide free web access to the public (e.g., public libraries), they may not be feasible locations for virtual exercise training. Thus, virtual exercise solutions that consider the financial limitations of current fitness trends are needed.

 

PROMOTING AN ACTIVE LIFESTYLE THROUGH THE BUILT ENVIRONMENT

 

There are few surveillance data on physical activity patterns over many years in representative populations that use consistent methods of data collection. Data from consumer groups and national monitoring and surveillance systems among persons living in the United States generally show a stable pattern of both leisure time and sport activity (33)but a decrease in work-related activity starting in the 1950s (34). These types of data are useful at the ecologic level in order to describe lifestyle trends among the population and to provide background data for community-based interventions that eventually affect public policy. Environmental interventions that promote change in risk conditions at the community level have a greater public health impact than attempting to change risk factors at the individual level. Environmental strategies more directly related to promoting an active lifestyle involve altering the built environment in which people spend much of their time—the community, the workplace, and the school.

 

A report from the Transportation Research Board (TRB) and the Institute of Medicien (IOM) outlines a number of recommendations pertaining to physical activity and the built environment (35). These recommendations state the primary need for multidisciplinary and inter-agency research (particularly longitudinal research and "natural experiments") linking specific aspects of the built environment with different types of physical activity. Ecological studies that can geocode physical activity and health data from surveillance systems such as the Behavioral Risk Factor Surveillance System (BRFSS) or from the National Health and Nutrition Examination Survey (NHANES) could provide useful information on the environment and the specific locations where low activity and/or high prevalence of overweight is occurring. Similarly, statistical tools such as Geographical Information Systems (GIS) can provide more detailed information on the built environment (land use, sidewalks, green space) to link with surveillance data on physical activity patterns and various health indicators like obesity within a community. These data are also quite useful in tracking how changes to the environment affect changes in behavior and in subsequent health outcomes.

 

The Health Impact Statement historically has been used in environmental risk assessment to inform the public of the health consequences of various actions (e.g., the building of a new manufacturing plant in the community) and generally, they are effective at involving inter-agency action and public consensus. Since available evidence suggests that the built environment plays a major facilitating role in promoting an active lifestyle, urban planners, local zoning officials, those responsible for the construction of residences, developments, and supporting transportation systems, and members of the community must work together in the design of more activity-friendly environments.

 

SUMMARY

 

Most research to date suggests that exercise is more effective in the prevention of overweight and obesity than it is in its reversal. Weight loss programs that combine exercise with caloric restriction can maximize the net caloric deficit while reducing the loss of fat-free mass. Adding resistance training to aerobic exercise will enhance muscle quantity and quality, thereby providing health benefits independent of weight loss. Accumulating the necessary exercise and lifestyle physical activity in intermittent bouts, rather than one long continuous bout, can improve adherence and the success of weight loss and maintenance regimens.

 

uture research should focus on the prevention of excess weight gain over the life course. In addition to the behavioral and intervention studies of the past several decades, an understanding of the regulatory processes governing energy intake, energy storage, and energy expenditure and how the reinstatement of exercise can correct the disruption of neural pathways is vital to the future of obesity research. Molecular and clinical studies that can identify candidate genes and other biomarkers of energy regulation responding to exercise should link with large epidemiologic studies to determine the relations among these biological markers, physical activity patterns and long-term weight gain among various populations. Controlled intervention trials should continue to test the dose-response relation between physical activity duration (min/week), volume (kcal/week), and/or intensity and various functional endpoints as rigorously as do pharmacological trials. Finally, public health science needs to link with public health practice to better enable the translation of this knowledge into policies that can alter the environment in a way that promotes an active lifestyle for all.

 

ACKNOWLEDGMENTS

 

This work was supported in part by grants from the National Institutes of Health, National Heart Lung and Blood Institute (HL135089 to and TS and NSS).

 

REFERENCES

 

1)      Inoue, Y., Qin, B., Poti, J., Sokol, R., Gordon-Larsen, P. Epidemiology of obesity in adults: Latest trends. Curr Obes Rep. 2018,7:276-88.

2)      Ogden, C. L., Fryar, C. D., Martin, C. B., Freedman, D. S., Carroll, M. D., Gu, Q., et al. Trends in Obesity Prevalence by Race and Hispanic Origin-1999-2000 to 2017-2018. JAMA. 2020,324:1208-10.

3)      Hales, C. M., Carroll, M. D., Fryar, C. D., Ogden, C. L. Prevalence of Obesity and Severe Obesity Among Adults: United States, 2017-2018. NCHS Data Brief. 2020:1-8.

4)      DiPietro, L., Dziura, J., Blair, S. N. Estimated change in physical activity level (PAL) and prediction of 5-year weight change in men: The Aerobic Center Longitudinal Study. Int J Obesity. 2004,28:1541-7.

5)      Jakicic, J. M., Powell, K. E., Campbell, W. W., Dipietro, L., Pate, R. R., Pescatello, L. S., et al. Physical Activity and the Prevention of Weight Gain in Adults: A Systematic Review. Med. Sci. Sports Exerc. 2019,51:1262-9.

6)      Physical Activity Guidelines Advisory Committee. Physical Activity Guidelines Advisory Committee Report, 2018. Part F. Chapter 5. https://health.gov/paguidelines/guidelines/report.aspx. Accessed January 31, 2021. Washington, DC: US Department of Health and Human Services2018.

7)      Chakravarthy, M. V., Booth, F. W. Eating, exercise and "thrifty" genotypes:  connecting the dots toward an evolutionary understanding of modern chronic diseases. J Appl Physiol. 2004,96:3-10.

8)      Pontzer, H. Constrained Total Energy Expenditure and the Evolutionary Biology of Energy Balance. Exerc Sport Sci Rev. 2015,43:110-6.

9)      Bull, F., Al-Ansari, S. S., Biddle, S., Borodulin, K., Buman, M. P., Cardon, G., et al. World Health Organization 2020 guidelines on physical activity and sedentary behaviour. Br. J. Sports Med. 2020,54:1451-62.

10)    American College of Sports Medicine, Riebe, D., Ehrman, J. K., Liguori, G., Magal, M. Benefits and Risks Associated with Physical Activity.  ACSM's Guidelines for Exercise Testing and Prescription. Tenth ed. Philadelphia, PA: Wolters Kluwer; Lippincott Williams & Wilkins; 2018. p. 1-21.

11)    Brooks, G. A., Butte, N. F., Rand, W. M., Flatt, J. P., Caballero, B. Chronicle of the Institute of Medicine physical activity recommendation: how a physical activity recommendation came to be among dietary recommendations. Am. J. Clin. Nutr. 2004,79:921S-30S.

12)    Hill, J. O., Wyatt, H. R., Peters, J. C. Energy balance and obesity. Circulation. 2012,126:126-32.

13)    Hill, J. O., Wyatt, H. R. Role of physical activity in preventing and treating obesity. J Appl Physiol (1985). 2005,99:765-70.

14)    Saris, W. H. M., Blair, S. N., van Baak, M. A., Eaton, S. B., Davies, P. S. W., Di Pietro L, et al. How much physical activity is enough to prevent unhealthy weight gain? Outcome of the IASO 1st Stock Conference and consensus statement. Obesity Rev. 2003,4:101-14.

15)    Poirier, P., Després, J. P. Exercise in weight management of obesity. Cardiol. Clin. 2001,19:459-70.

16)    Ross, R., Janssen, I. Physical activity, total and regional obesity: dose-response considerations. Med Sci Sports Exerc. 2001,33:S521-7; discussion S8-9.

17)    Schoeller, D. A., Shay, K., Kushner, R. F. How much physical activity is needed to minimize weight gain in previously obese women? Am. J. Clin. Nutr. 1997,66:551-6.

18)    Jakicic, J. M., Clark, K., Colmena, E., Donnelly, J. E., Foreyt, J., Melanson, E., et al. Appropriate intervention strategies for weight loss and prevention of weight regain for adults. Med. Sci. Sports Exerc. 2001,33:2145-56.

19)    Ravussin, E., Lillioja, S., Anderson, T. E., Christin, L., Freymond, D., Abbott, W. G. H., et al.  Reduced rate of energy expenditure as a risk factor for body weight gain. N. Engl. J. Med. 1988,318:467-72.

20)    Ainsworth, B. E., Haskell, W. L., Whitt, M. C., Irwin, M. L., Swartz, A. M., Strath, S. J., et al. Compendium of physical activities: an update of activity codes and MET intensities. Med Sci Sports Exerc. 2000,32:S498-504.

21)    Blundell, J. E., King, N. A. Exercise, appetite control, and energy balance. Nutrition. 2000,16:519-22.

22)    Stonehouse, W., Wycherley, T. P., Luscombe-Marsh, N., Taylor, P., Brinkworth, G., Riley, M. Dairy Intake Enhances Body Weight and Composition Changes during Energy Restriction in 18-50-Year-Old Adults-A Meta-Analysis of Randomized Controlled Trials. Nutrients. 2016,8:394.

23)    Normandin, E., Chmelo, E., Lyles, M. F., Marsh, A. P., Nicklas, B. J. Effect of Resistance Training and Caloric Restriction on the Metabolic Syndrome. Med Sci Sports Exerc. 2017,49:413-9.

24)    Villareal, D. T., Aguirre, L., Gurney, A. B., Waters, D. L., Sinacore, D. R., Colombo, E., et al. Aerobic or resistance exercise, or both, in dieting obese older adults. N. Engl. J. Med. 2017,376:1943-55.

(25)  Jacobsen, D. J., Bailey, B. W., LeCheminant, J. D., Hill, J. O., Mayo, M. S., Donnelly, J. E. A comparison of three methods of analyzing post-exercise oxygen consumption. Int. J. Sports Med. 2005,26:34-8.

26)    Jakicic, J. M., Winters, C., Lang, W., Wing, R. R. Effects of intermittent exercise and use of home exercise equipment on adherence, weight loss, and fitness in overweight women: a randomized trial. JAMA. 1999,282:1554-60.

27)    Jakicic, J. M., Marcus, B. H., Gallagher, K. I., Napolitano, M., Lang, W. Effect of exercise duration and intensity on weight loss in overweight, sedentary women: a randomized trial. JAMA. 2003,290:1323-30.

28)    Westerterp, K. R. Pattern and intensity of physical activity. Nature. 2001,410:539.

29)    Thompson, W. R. Worldwide Survey Of Fitness Trends For 2016: 10th Anniversary Edition. ACSM's Health & Fitness Journal. 2015,19:9-18.

30)    Thompson, W. R. Worldwide Survey Of Fitness Trends For 2020. ACSM's Health & Fitness Journal. 2019,23:10-8.

31)    Wen, D., Zhang, X., Liu, X., Lei, J. Evaluating the Consistency of Current Mainstream Wearable Devices in Health Monitoring: A Comparison Under Free-Living Conditions. J Med Internet Res. 2017,19:e68.

32)    Seshadri, D. R., Drummond, C., Craker, J., Rowbottom, J. R., Voos, J. E. Wearable Devices for Sports: New Integrated Technologies Allow Coaches, Physicians, and Trainers to Better Understand the Physical Demands of Athletes in Real time. IEEE Pulse. 2017,8:38-43.

33)    Keadle, S. K., McKinnon, R., Graubard, B. I., Troiano, R. P. Prevalence and trends in physical activity among older adults in the United States: A comparison across three national surveys. Prev. Med. 2016,89:37-43.

34)    Church, T. S., Thomas, D. M., Tudor-Locke, C., Katzmarzyk, P. T., Earnest, C. P., Rodarte, R. Q., et al. Trends over 5 Decades in U.S. Occupation-Related Physical Activity and Their Associations with Obesity. PLoS One. 2011,6:e19657.

35)    Transportation Research Board Special Report 282 Does the built environment influence physical activity. Examining the evidence. . In: Medicine. TRBaIo, editor. Washington, DC2005. p. 1-15.

Adrenal Disorders in the Tropics

ABSTRACT

 

The adrenal gland in conjunction with the pituitary gland is one of the major components of the endocrine system and regulates blood volume, blood pressure, serum electrolytes, and stress responses. Dysfunction of the adrenal glands may be related to diseases of the adrenal glands or pituitary gland. Adrenal disorders may present either due to structural or functional abnormalities. In the tropical countries, adrenal insufficiency is primarily due to adrenal infection by tuberculosis, adrenal mycosis infections, and adrenal hemorrhages. HIV (Human immunodeficiency virus) related adrenal problems are also common. Adrenal dysfunction due to pituitary disorders still occur frequently in tropical region and include Sheehan’s syndrome, vasculotoxic snake bite, and thalassemia. Adrenal hormone excess typically occurs secondary to exogenous glucocorticoid use. Adrenal disorders that occur in the developed world occur with similar frequencies in tropical regions.

INTRODUCTION  

Adrenal glands are one of the major peripheral organs necessary for homeostasis including maintenance of blood volume, blood pressure, and serum electrolytes. Disorders of adrenal glands are common in clinical practice. Adrenal dysfunction in tropical countries often occurs due to specific etiologies that differ from the typical causes of adrenal dysfunctions that commonly occur in other parts of the world (Table 1). 

Table 1. Classification of Adrenal Disease in the Tropics

Adrenal insufficiency:

Primary: 

1)     Adrenal Tuberculosis

2)     Adrenal Mycosis

3)     Adrenal Haemorrhage

Secondary:

1)     Sheehan’s Syndrome

2)     Vasculotoxic Snake Bite

3)     Thalassemia’s

Both Primary and Secondary:

1)    HIV

Adrenal Hormone excess syndromes:

1.    Exogenous Glucocorticoid hormone excess syndromes

2.  Licorice induced syndrome of apparent mineralocorticoid excess

PRIMARY ADRENAL INSUFFICIENCY  

The causes of primary adrenal insufficiency that are more frequent in tropical regions include infection of the adrenal glands by tuberculosis or mycotic infections. In addition, autoimmune Addison’s disease or adrenal failure as a component of polyglandular syndromes are equally prevalent in tropical regions as is in other parts of the world. 

Adrenal Gland Tuberculosis

Adrenal gland tuberculosis or Tuberculous adrenalitis is the result of infection of adrenal gland by mycobacterium tuberculosis. The infection causes a destructive lesion of the adrenal cortex with uncertain chances of recovery and remains one of the most important causes of Addison’s disease in the tropical countries (1). In fact, the adrenal glands are the most common endocrine organs to be involved in tuberculosis (2). Adrenal gland tuberculosis occurs almost always secondarily due to the hematogenous spread of the bacilli to the gland with the primary focus in lung. Adrenal failure or Addison’s disease clinically manifest when at least 90% of the gland has been destroyed (1,2,3). Though classically the adrenal cortex is involved, the medulla also may be involved in many cases of adrenal tuberculosis (3,4).

PATHOPHYSIOLOGY 

It is interesting to know why the adrenal glands are susceptible to infections. In fact, adrenal gland infections are common in response to a distant infection elsewhere in the body and in disseminated infection. Autopsy examination revealed that the prevalence of adrenal tuberculosis is about 6% in patients with active tuberculosis (4). However, subclinical adrenal dysfunction may be present in about 60-70% of patients with active tuberculosis (5). In any of these situations, there is an exaggerated response of the hypothalamo-pituitary-adrenal axis to produce excess cortisol in response to the stress of infection. This stress induced hypercortisolemia shifts the balance in the Th1/Th2 cell ratio towards a Th2 response (6). This T cell dysfunction (which is primarily responsible for cell mediated immunity) and low DHEA levels increases the host susceptibility to infection to mycobacterium tuberculosis and other organisms (6). Low DHEAS levels have been documented in tuberculosis (1,6). In addition, endotoxin released in response to the hyperactive HPA axis can cause pathological changes in the adrenal glands to increase the susceptibility to infection (7). The intrinsically rich vascularity of the adrenal glands promotes all of these pathophysiological events.

Histopathologically, four classic patterns have been described in adrenal tuberculosis (3). These are:  granuloma (caseating or non-caseating), enlargement of the gland with destruction by inflammatory granuloma, mass lesions due to cold abscesses, and adrenal atrophy due to fibrosis related to chronic infection. Caseating granuloma is the commonest one and this is identified in about 70% of cases (4). However, granuloma with typical presence of Langhan’s giant cell are less common and identified in less than 50% of cases (4), probably due to anti-inflammatory effects of local glucocorticoids. Calcification of the gland is a common but it is present in other chronic infections of the adrenal glands (3). In about 25 % cases the infection may be unilateral (1).

PRESENTATION

Typical symptoms of adrenal gland tuberculosis in a patient with diagnosed tuberculosis (whether or not on anti-tubercular chemotherapy) are mucocutaneous pigmentation in association with chronic ill health, vomiting, postural hypotension, and anorexia (3). The features are similar to Addison’s disease due to other conditions. As the features of progressively evolving adrenal hypofunction are mostly nonspecific, a high index of suspicion is necessary in subjects with diagnosed active tuberculosis especially when pigmentation is absent. However clinical manifestations may take months to years to become apparent.

The patient may also present rarely with frank adrenal crisis with hypotension, hyponatremia, hyperkalemia, and low serum cortisol levels. The crisis may even be precipitated after administration of rifampicin which increases the hepatic metabolism of cortisol in the background of subclinical adrenal dysfunction (8). 

Adrenal tuberculosis may also present as an adrenal incidentaloma. Nonspecific abdominal pain, weight loss, dizziness, and vomiting may lead to imaging of the abdomen which may reveal an incidental adrenal mass often with calcification. The differential diagnosis of Addison’s disease with adrenal enlargement includes (apart from tuberculosis) malignancy, fungal infections, hemorrhage, amyloidosis, sarcoidosis, etc. (3).

Subclinical adrenal dysfunction is also very common and should be actively sought in all cases of active tuberculosis (5).

INVESTIGATIONS

Laboratory Studies

Common laboratory findings include anemia, hyponatremia, and hyperkalemia. In the presence of a positive Mantoux test in association with typical clinical manifestations of adrenal hypofunction, adrenal tuberculosis must be ruled out. Adrenal insufficiency should be ruled out by using a standard protocol. Serum cortisol levels <5 µg/dL and a plasma ACTH more than 2-fold the upper limit of the reference range is suggestive of primary adrenal insufficiency (9). The serum cortisol may remain in the low-normal to mid-normal range in many cases.  However, a standard dose (250 µg) intravenous cosyntropin (Synacthen) stimulation test establishes the diagnosis of adrenal insufficiency when the peak level of cortisol remains below 18 µg/d (9). Random cortisol levels, though useful during an acute crisis, is not usually sufficient to rule out adrenal insufficiency (9). Documentation of subclinical adrenal dysfunction may reveal mineralocorticoid deficiency alone (as demonstrated by raised plasma rennin activity) when stimulated cortisol is within the normal range (8).

Imaging of Adrenal Glands

CT scan of the abdomen is the most important non-invasive investigation with a very good spatial resolution to diagnose adrenal tuberculosis. The findings are usually bilateral and vary with the duration of the disease before diagnosis (1, 3). The most common early findings during the initial 2 years include a mass lesion with smooth adrenal contour preserved. The glands may show central or patchy hypodensity corresponding to areas of caseous necrosis (3). On contrast administration there is peripheral rim enhancement. Calcification is not a common feature in early tuberculosis (3).

With chronic infection, the adrenal glands become small and shrunken, often with associated calcifications and the margins become irregular (3). Though prevalence and intensity of calcification increases with the duration of tuberculosis, this is not a specific finding and may be associated with other conditions.

Though MRI is also done in many cases, this imaging modality has limitations to assess calcification. However, T1 weighted image shows hypointense or isointense areas and T2 weighted image shows hyperintense areas because of necrosis (3).

Percutaneous FNA/ TB PCR 

 For confirmation of adrenal tuberculosis tissue diagnosis is required. CT scan guided fine needle aspiration from the adrenal gland is necessary to obtain adequate tissue specimens (3, 10). Pathological and microbiological confirmation is necessary, especially where there is isolated adrenal involvement. However, it should be remembered that PCR and culture of these specimens for tuberculosis bacilli are not consistently positive (3). Hence a combination of histopathology, PCR, and culture may be necessary to confirm the diagnosis (3). However, routine search for pulmonary tuberculosis with necessary investigations is mandatory.

TREATMENT

Treatment of adrenal insufficiency in tuberculosis requires administration of both glucocorticoids and mineralocorticoids. As the medulla is frequently involved, patients may require higher doses for maintenance of blood pressure. At the same time, rifampicin used in the anti-tubercular regimen is a potent hepatic enzyme inducer and accelerates cortisol metabolism. This also may necessitate a higher dose of glucocorticoids for adequate treatment. However, aldosterone is less likely to be involved. Adrenal crisis is also reported to occur following the administration of rifampicin (11).

Therapy is monitored with blood pressure, body weight, well-being, serum electrolytes and blood glucose. Patients should be also be monitored for over treatment with glucocorticoids with weight gain, blood pressure, decreasing bone mineral density, and other manifestations of Cushing’s syndrome. All subjects should carry a ‘steroid card’ and should be advised strictly on how to increase the dose of glucocorticoid in stressful situations such as fever, infection, vomiting, trauma, etc.

PROGNOSIS FOR ADRENAL FUNCTION RECOVERY

Chances of adrenal recovery with anti-tuberculosis therapy are uncertain and unpredictable. When the disease is diagnosed late, the glandular destruction is usually significant and the gland becomes atrophic, and anti-tuberculosis therapy does not lead to a recovery of adrenal function (12, 13). If therapy is started early before the gland is destroyed recovery may occur (14, 15). It is also suggested that if the gland size remains the same on subsequent follow up CT scans, it is prudent to follow up the patient for adrenal function recovery.

Adrenal Mycosis

HISTOPLASMOSIS 

Adrenal Histoplasmosis caused by the dimorphic fungus Histoplasma capsulatum, is a recognized cause of adrenal insufficiency. Though this opportunistic pathogen is known to affect immunocompromised individuals predominantly (16), it can rarely infect immunocompetent individuals (16, 17).  This is the most fungal infection of the adrenal glands (16, 18).

Involvement of the adrenals can occur during disseminated infection or many years after disease resolution (18). Adrenal involvement can vary from an asymptomatic milder form to a very severe form that presents with extensive bilateral granulomatous involvement of the entire adrenal gland with calcified lesions culminating in acute adrenal insufficiency (18, 19). Rarely the involvement can be unilateral (17). The common differential diagnosis includes tuberculosis, other fungal infections, adrenal metastasis, primary adrenal malignancy, and primary adrenal lymphoma (16). In immunocompetent individuals it commonly presents with a unilateral or bilateral adrenal mass with constitutional symptoms.

The hypothesis for why histoplasmosis involves the adrenal glands with increased frequency includes the local high levels of glucocorticoids in association with a relative paucity of reticulo-endothelial cells within the adrenal gland (6). The gland is destroyed by direct infection that leads to local ischemia and infarction due to perivasculitis, and caseation (6).

Diagnosis depends on imaging studies with pathological confirmation. CT scan of the adrenal glands typically reveals symmetric enlargement with central hypodensity and characteristic peripheral rim like enhancement (20). Frequently calcification is also present, particularly during the healing phase (20). Percutaneous ultrasound or CT guided fine-needle aspiration or biopsy is necessary for tissue diagnosis (18). The characteristic cytopathological findings are the presence of numerous small oval yeast like structures inside the cytoplasm of macrophages (16). On a necrotic background, this yeast like structures inside the macrophages is surrounded by a clear ring of space resembling a capsule. However, the gold standard for diagnosis is documentation of the organism in the culture of pathological specimen (16). Bhansali et al reported a high uptake in adrenal glands in FDG-PET scan in patients with adrenal histoplasmosis (17). 

Treatment for adrenal histoplasmosis depends on the severity of the infection and the condition of the patient. For severe infection in critically ill patient’s amphotericin B is used initially followed by long-term therapy with oral itraconazole (16). Parenteral liposomal amphotericin B is given 3mg/kg body weight for 2 weeks (17). The duration of therapy with itraconazole varies from six months to two years depending on the patient’s condition. For mild to-moderate histoplasmosis, the recommended treatment is itraconazole. The recommended dose is 200 mg twice daily given for 12 months (16). When itraconazole is used, liver enzymes should be monitored on a regular basis (18).  Treatment for adrenal insufficiency follows the same principles as described earlier.

Though the remission rate from adrenal histoplasmosis is high with long-term oral itraconazole, adrenal insufficiency rarely resolves and reversal of adrenal dysfunction can be seen only in some patients after prolonged antifungal therapy (21).  However, histoplasma in adrenals is reported to persist even 7 years after antifungal therapy (22). 

OTHER FUNGAL INFECTIONS

Paracoccidioidomycosis Brasiliensis

Paracoccidioidomycosis brasiliensis is a dimorphic fungus and can cause chronic, progressive, suppurative and granulomatous disease which can lead to adrenal insufficiency (3). The disease is endemic in Latin America. Humans are the accidental host for the organism and females are rarely affected (23). Smoking and alcohol increase the risk. The lungs are the usual portals of entry. Juvenile forms of the disease are also known (23). Apart from frank adrenal crisis, it can present as progressive constitutional symptoms, hyperpigmentation, and low blood pressure with postural drop and bilateral adrenal enlargement in imaging studies with frank adrenal calcification detected by CT scans (24, 25). Histopathology with GMS stain shows multiple budding yeast with steering wheels appearance which is consistent with Paracoccidioides brasiliensis (24). However, confirmation of the organism by culture material is the gold standard for diagnosis. Serology for antibody detection is also useful in the diagnosis. Diagnosis and treatment of adrenal insufficiency is not different than described above for histoplasmosis. P. brasiliensis primarily causes adrenal destruction by embolic infection of small vessels by large fungal cells and granuloma formation (3). Subjects who receive early antifungals with itraconazole over a 1–2-year period may have a full recovery of adrenal function by preventing fungal embolism in adrenal gland vasculature and reducing ischemic necrotic destruction of the gland (3). Hence an early diagnosis is crucial for preventing the progression of adrenal dysfunction. However, persistence of high antibody titer against paracoccidioidomycosis at the end of treatment or during follow-up is a frequent finding in subjects with paracoccidioidomycosis.

Blastomyces Dermatitidis

Blastomyces dermatitidis is also a dimorphic fungus, which has a strong affinity for the adrenal gland for reasons described earlier. Overt adrenal insufficiency is less common and adrenal Blastomyces dermatitidis typically presents as bilateral adrenal incidentaloma during radiological investigations for other reasons (3). The portal of entry is through the lungs and when there is lymphohematogenous dissemination the disease spreads to other organs (26). In situations when it presents as adrenal insufficiency, the presentation, investigations, and management are similar to those described above. Diagnosis is by fine-needle aspiration guided by ultrasound or CT scan followed by cytologic and histologic examinations. However, the gold standard is fungal culture showing thick-walled, broad-based budding yeast cells (27). Treatment is with long term oral itraconazole. In patients with severe manifestations initial treatment with liposomal amphotericin B for 2 weeks could be used.

Cryptocoocus Neoformans

Cryptocoocus neoformans is an encapsulated yeast-like fungus which infects primarily immunodeficient hosts, particularly subjects infected with HIV or lymphohematogenous malignancies (28). In immunocompromised hosts it usually affects the central nervous system and lungs.  However immune-competent individuals may also suffer adrenal cryptococcosis (29). Adrenal dysfunction is uncommon until almost the whole of adrenal gland is infiltrated with C. neoformans and caseating granulomas. Cryptococcosis is diagnosed by fine-needle aspiration biopsy of the adrenal mass. The serum cryptococcal antigen titer is highly elevated. Treatment is with antifungal therapy with fluconazole and amphotericin B. Adrenal enlargement by Cryptococcus may be completely reversible without any abnormality after antifungal treatment (30). Cases not responsive to anti-fungal therapy have been reported to improve after unilateral or bilateral adrenalectomy (28, 29).

Miscellaneous

Pneumocystis jirovecii (previously known P. carinii) occurs in individuals with advanced HIV due to defects in cell mediated immunity. Spread to other organs including the adrenal glands is also possible (3). Adrenal failure associated with coccidioidomycosis and rarely candidiasis has also been reported.

Adrenal Hemorrhage; the Waterhouse Friderichsen Syndrome

This is a condition in which patient presents with acute hypotension and shock due to adrenal insufficiency arising from acute adrenal hemorrhage. The syndrome is typically related to infection with Neisseria meningitides infection (3). However, this is also known to occur in septicemia due to infections with Staphylococcus aureus, Streptococcus spp, Haemophilus influenzae, Corynebacterium diphtheria, etc. (3). Hence this is more common in the tropical region.  The condition is hypothesized to be due to interplay between endotoxemia and elevated ACTH. The adrenal gland is anatomically prone to hemorrhage as it has three separate arterial supplies and does not have proportional venous drainage (3). In endotoxemia, elevated ACTH increases the blood supply several fold in this compromised anatomical setting. At the same time increased adrenaline secretion in relation to stress leads to constriction of adrenal veins, which further increases this imbalance between arterial supply and venous drainage. Management includes immediate fluid replacement and parenteral glucocorticoids apart from the management of the underlying infection.

SECONDARY ADRENAL INSUFFICIENCY 

Adrenal insufficiency secondary to disorders of pituitary gland is also very common in developing countries in tropical regions.  Secondary adrenal insufficiency caused by pituitary tumors and apoplexy, pituitary surgery, radiation therapy, hypophysitis, various genetic disorders, and withdrawal of exogenous steroids are equally common in tropical regions but certain other disorders like Sheehan’s syndrome, thalassemia, and vasculotoxic snake bite induced pituitary failure are more common in tropical regions.

Sheehan’s Syndrome 

Sheehan’s syndrome consists of various degrees of pituitary insufficiency, which develops as a result of ischemic pituitary necrosis due to severe postpartum hemorrhage. The important pathogenetic/predisposing factors include a small sella, increased pituitary volume, vasospasm induced by postpartum hemorrhage, thrombosis, and probable pituitary autoimmunity (31). In developed countries there has been a drastic reduction in the incidence of Sheehan’s syndrome. This is primarily due to the remarkable improvement in obstetric care and availability of rapid blood transfusion. However, this remains as a major cause of hypopituitarism in the other parts of the world.

CLINICAL FEATURES

Most commonly the disorder presents as a lactation failure in the post-partum state and non-resumption of menses following child birth, which was complicated by massive post-partum hemorrhage leading to hypotension and shock. However, it may very rarely occur without massive bleeding or after normal delivery. Patients may present in the emergency with altered sensorium, loss of consciousness, seizure, shock, intractable vomiting, or more commonly with chronic complaints like asthenia and weakness, dizziness, anorexia, weight loss, nausea, and vomiting with a typical history of failure to resume menses and lactation failure following child birth (31). Apart from anterior pituitary hormone deficiency, symptoms like anemia, pancytopenia, osteoporosis, cognitive impairment, and poor quality of life are also present in these patients (31,32).  Very rarely diabetes insipidus may occur. However, the mean age of the participants may be as late as 40 years or more and the mean interval between inciting event to diagnosis may be as high as 10 years or more (33).

Adrenal insufficiency due to ACTH deficiency is reported to occur in up to 100% of cases (in fact deficiency of all anterior pituitary hormones occur in a variable percentage of patients and may be up to 100%) (32). Weakness, fatigue, and postural drop are common manifestations. Hyponatremia is particularly common in Sheehan’s syndrome, which may be due to glucocorticoids deficiency coupled with increased AVP release as a consequence of reduced blood pressure and cardiac output resulting from glucocorticoid deficiency (32).

DIAGNOSIS

The basal pituitary hormonal levels and those after dynamic tests are beyond the purview of this chapter. However adrenal insufficiency is diagnosed with a morning cortisol level of 3 mcg/dl with low or inappropriately normal ACTH or a cosyntropin stimulated cortisol level <18 mcg/dl. Documentation of growth hormone deficiency does not require a dynamic test in presence of other pituitary hormone deficiencies. Only low age specific and assay specific IGF-1 assay may be sufficient to document adult growth hormone deficiency (AGHD) (34).

The preferred radiological imag­ing is an MRI of hypothalamic pituitary area.  CT scan may also be helpful. MRI findings in Sheehan’s syndrome usually vary with the stages of the disease. In earlier stages of the disease there may be an enlarged pituitary gland with central hypodensity (suggestive of infarction). However, an empty sella (complete or partial) is considered to be a characteristic of Sheehan’s syndrome in established cases (32).

TREATMENT

The acute adrenal crisis in Sheehan’s syndrome is treated with intravenous glucocorticoids. In other patients’ glucocorticoids should be started orally with hydrocortisone 15-25 mg daily in 2-3 divided doses with the higher dose in the morning and a lower dose in the evening (35). Mineralocorticoids are not necessary in general (35). Once daily prednisolone may also be used at a dose of 2.5-5 mg once daily in the early morning. As GH deficiency decreases cortisol clearance, it may necessary to increase the dose of glucocorticoid for those who receive GH treatment (35). Therapy is monitored with blood pressure, body weight, well-being, serum electrolytes, and blood glucose. Patients should be also be monitored for an overdose of glucocorticoids with weight gain, blood pressure, decreased bone mineral density, and other symptoms and signs of Cushing’s syndrome. All subjects with Sheehan’s syndrome should carry a ‘steroid card’ and should be advised strictly on how to increase the dose of glucocorticoid in stressful situation such as fever, infection, vomiting, trauma, etc.

Subjects with Sheehan’s syndrome should also be treated with levothyroxine, combined oral contraceptives according to guideline, calcium and vitamin D supplements, and growth hormone therapy (if possible) according to the protocol of adult growth hormone deficiency.

Viscerotropic Snake Bite

Snakebite is a major public health problem in tropical regions and is considered as one of the most neglected tropical diseases. The development of a Sheehan-like syndrome with chronic hypopituitarism following Russell viper envenomation is fairly common. Hypoadrenalism due to ACTH deficiency is the commonest abnormality (36). However acute hypopituitarism with predominant glucocorticoids deficiency has also been reported (37).

The venom of vipers is vasculotoxic in nature and the clinical features of viper venomation include local cellulitis and tissue necrosis, bleeding manifestations, disseminated intravascular coagulation, shock, and acute kidney injury (AKI) (38). Hypopituitarism is particularly common following vasculotoxic snake bite in subjects who develop AKI requiring hemodialysis. Hypopituitarism can develop as early as 7 days following snake bites and should be evaluated for particularly in younger subjects, especially those requiring increasing number of sessions of hemodialysis and in subjects with abnormal 20 min WBCT (whole blood clotting test) at presentation (36,39). On the other hand, the time of onset/presentation of hypopituitarism following snake bite may be as long as up to 24 years (40). Acute hypopituitarism is thought to occur due to acute damage to the pituitary gland at the time of the precipitating event, but a gradual/slower progression of pituitary damage may occur over years due to other unknown mechanisms (36).

Those who survive acute snake bite may later present with altered sensorium, loss of consciousness, seizure, shock, intractable vomiting, or more commonly with chronic complaints like asthenia and weakness, dizziness, anorexia, weight loss, nausea, vomiting and amenorrhea in females (36).

Variable degrees of hypopituitarism may be present. Cortisol deficiency is reported to be the commonest abnormality. Secondary adrenal insufficiency is diagnosed with a morning cortisol level of 3 mcg/dl with low or inappropriately normal ACTH or a co-syntropin stimulated cortisol level <18 mcg/dl (36). Documentation of growth hormone deficiency is done as mentioned in section of Sheehan’s Syndrome (34).

The preferred radiological imag­ing is the MRI of hypothalamic pituitary area which may show partial or complete empty sella or evidences of old hemorrhage. However, these changes are not present in all cases (41).

Treatment of secondary adrenal insufficiency and other hormone deficiencies are similar to described above. All subjects with hypopituitarism on glucocorticoids supplements should carry a ‘steroid card’ and should be advised on how to increase the dose of glucocorticoid in stressful situation such as fever, infection, vomiting, trauma, etc.

Thalassemia Major

Thalassemia’s are inherited autosomal recessive disorders of hemoglobin synthesis. Thalassemia major is the most severe form of beta thalassemia which involves the beta chain of hemoglobin. Organ dysfunction in thalassemia is principally attributed to excessive iron overload and suboptimal chelation. The precise underlying mechanism of iron overload induced organ dysfunction is not very unclear. The current management of thalassemia includes regular transfusion programs and chelation therapy. Pre-marital counselling and assessment with HPLC to assess the asymptomatic carrier has reduced its prevalence significantly in the developed world. However, this is still a major problem in many parts of the world.  Prevalence of adrenal insufficiency is variable and depends on the severity of iron overload. This secondary hemochromatosis can disrupt adrenal function by affecting the hypothalamic-pituitary-adrenal axis at the hypothalamic or pituitary level (42). In more severe cases primary adrenal failure may supervene due to iron deposition in the adrenal glands (42). Additionally, an extramedullary hematopoietic tumor has been reported in HbE thalassemia and beta thalassemia as non-hormone secretory unilateral or bilateral adrenal enlargement resembling adrenal myelolipoma (43). 

Biochemical adrenal insufficiency is reported to occur from   0% to 45% of subjects with thalassemia major (42), but adrenal crisis or clinical adrenal insufficiency is extremely uncommon and mostly they are asymptomatic. However, subclinical cortisol deficiency is not uncommon. In this context it should be remembered that mild symptoms of adrenal insufficiency like asthenia, weight loss, or postural drops are frequently overlooked as these features are common in thalassemia subjects with low levels of hemoglobin (42).                           

The unique finding in subjects with thalassemia is the dissociation between adrenal androgen levels with cortisol and aldosterone levels. This paradox is reflected by frequent documentation of low serum DHEA, DHEA-sulfate, androstenedione, and testosterone levels in the presence of normal serum cortisol and aldosterone levels (44). Absence of adrenarche occurring in most adolescents with thalassemia major is probably explained by this phenomenon (45). 

Diagnosis of adrenal dysfunction in thalassemia is similar to other causes of secondary adrenal insufficiency. If the morning cortisol is not unequivocally low, synacthen stimulation test should be done with either the low dose (1 µg) or the standard high dose (250 µg). A peak cortisol level of >18 µg/dL after 30-60 min of intravenous synacthen excludes adrenal insufficiency. Alternately an insulin tolerance test with a similar cut-off may also be done.

Treatment of clinical adrenal insufficiency is similar to that described above. Subjects with subclinical adrenal insufficiency require only steroid coverage during periods of stress.

HIV AND ADRENAL DYSFUNCTION

Endocrine manifestations of HIV infection may include adrenal dysfunction, hypothyroidism, hypogonadism, insulin resistance and diabetes etc. Changes in the HPA (hypothalamic-pituitary-adrenal) axis are the most frequent abnormality (46). Adrenal dysfunction in HIV infection may be a consequence of concomitant systemic illness, opportunistic infections, and neoplasm (47).

Probably the most frequent adrenal abnormality is a stress induced elevation in serum cortisol and ACTH (46). This may be due to activation of the HPA axis due to HIV infection itself or pro-inflammatory cytokines (e.g., IL-1β, IL-6 and TNF-α) (46). Alternately a peripheral increase in the conversion of cortisone to cortisol due to activation of 11-β HSD type 1 in adipose tissue or decrease in cortisol metabolism may be responsible for increased cortisol with subnormal ACTH (46). Tissue hypersensitivity to glucocorticoids is also reported in subjects with HIV-1 infection, which may result in hippocampal atrophy, altered secretion of cytokine/interleukins, etc. (48).

On the other hand, subclinical or clinical adrenal dysfunction can happen in about 10-20% of subjects with advanced disease and multiple co-morbidities when about 80-90% of the gland is destroyed (46). The involvement and destruction by HIV, opportunistic infections, or malignancies in the adrenal glands or the hypothalamus and/or pituitary area can result in either primary or secondary adrenal sufficiency (47).

The opportunistic infections include cytomegalovirus (CMV), Mycobacterium avium-intracellular and M. tuberculosis, fungal infections (such as Histoplasma, Cryptococcus, and Pneumocystis jirovecii), and Toxoplasma gondii (47). Of these opportunistic infections, CMV infection is known to be the commonest etiology with earlier literature reporting Cytomegalovirus adrenalitis in nearly 80 % of cases of HIV infection (46). However, due to improvements in active management of HIV by HAART (highly active anti- retroviral therapy), the prevalence of adrenal insufficiency has decreased over the last two decades.

Medications used for the treatment of HIV infection and its complication may also result in adrenal dysfunction. For example:  Rifampicin used for mycobacterial infection is a known hepatic Cytochrome P 450 (CYP) enzyme inducer and can lower serum cortisol levels by enhanced cortisol metabolism. Ketoconazole used to treat severe mycotic infections inhibits adrenal steroid synthesis and can lead to glucocorticoid deficiency or even adrenal crisis in patients with impaired adrenal reserve (49). Interestingly, ART-related lipodystrophy (dorsocervical fat pad enlargement and visceral adiposity) may mimic Cushing’s syndrome but it is typically not associated with hypercortisolism (49). On the contrary, some protease inhibitors (e.g., ritonavir) used in ART are reported to decrease metabolism of endogenous and exogenously co-administered glucocorticoids, resulting in an iatrogenic Cushing's syndrome.

Tumors of the adrenal gland in HIV infected patients include Kaposi’s sarcoma and high-grade non-Hodgkin’s lymphoma. Kaposi’s sarcoma is secondary to co-infection with the oncogenic human herpes virus type 8 (HHV8) and non-Hodgkin’s lymphoma could be secondary to Epstein-Barr virus (EBV).

Assessment for symptoms of adrenal involvement requires a high degree of suspicion as constitutional symptoms of HIV may mask the features of adrenal insufficiency.  Morning serum cortisol should be done in all cases suspected for adrenal dysfunction. Stress induced hypercortisolemia does not require any further testing and low serum cortisol <5 μg/dl with an elevated ACTH level requires treatment with glucocorticoids and mineralocorticoids. In other cases, synthacthen stimulated cortisol is used to determine the course of treatment. Stimulated cortisol <18 μg/dl, especially if associated with elevated plasma ACTH, should be treated as adrenal insufficiency. Asymptomatic subjects with stimulated serum cortisol <18 μg/dl should be advised to take stress doses of glucocorticoids only as mentioned before.

Diagnosis and management of adrenal disorders in a patient with HIV infection does not differ from that in immunocompetent persons in general.

ADRENAL HORMONE EXCESS SYNDROMES

Glucocorticoid Excess Syndromes 

The primary cause of Cushing’s syndrome, more common in tropical regions, is exogenous glucocorticoids. The background etiology for exogenous steroid usage includes: nephrotic syndrome, rheumatoid arthritis and other collagen vascular disease, bronchial asthma, Graves’ orbitopathy, etc.  Glucocorticoids used as inhalational agent for bronchial asthma, in creams and ointments for eczematous skin lesions may also be responsible. Endogenous steroid excess (Cushing’s disease, ectopic ACTH syndromes, adrenal tumors) are equally common in tropical regions as in other areas of the world.

Often it is a challenge to suspect exogenous glucocorticoid use based on the patient’s history, especially in situations when glucocorticoids were not being used for a therapeutic purpose. Subjects presenting with features suggestive of Cushing’s syndrome should therefore mandatorily undergo testing for basal morning cortisol (with paired ACTH if possible) to rule out exogenous glucocorticoid use. A suppressed morning cortisol and plasma ACTH strongly suggests the diagnosis (50). One important caveat is that prednisolone may cross react with some cortisol assays giving false positive results in some chemiluminescent assay (51). Additionally, if the patient is receiving hydrocortisone, the result will also be fallacious to interpret. It is not uncommon in tropical regions that some form of glucocorticoids is being used in disguise as an alternative medicine for joint pain, respiratory problems, fever, or even as a weight gain therapy for young lean subjects. Hence a more detailed evaluation of the history with leading questions and scrutiny of all past records of medicine, including that of the alternative medicines, may sometimes reveal the offending agent. 

The clinical features that suggest exogenous Cushing’s syndrome are lack of pigmentation and the absence of hypertension and hirsutism (as exogenous Cushing’s syndrome does not contain mineralocorticoids and androgens as opposed to endogenous Cushing’s syndrome). Patients with exogenous Cushing’s syndrome are prone to develop glaucoma, osteoporosis, psychiatric disturbances, etc. (50).

Once diagnosed, these subjects should be advised to withdraw the offending agents and should be given hydrocortisone in the lowest possible dose for preventing adrenal crisis. The withdrawal of hydrocortisone subsequently after 3 months depends on the morning cortisol, after stopping the previous evening dose and subjecting the patient to short synacthen test to assess the recovery of HPA axis. Those with morning cortisol between 5 -18 µ/dl should be advised stress coverage only. For bone protection, all subjects with exogenous Cushing’s syndrome should receive bisphosphonate therapy unless contraindicated (52). Adequate calcium supplements with cholecalciferol should also be used.

For subjects receiving glucocorticoids for therapeutic purpose, it is essential to maintain bone protection, check for secondary diabetes and hypertension, and prevent gastric ulceration. Withdrawal (if at all possible) should be performed very slowly. When the therapeutic steroid reaches the lowest possible dose to prevent crisis, it is converted to equivalent dose of hydrocortisone and the same principle is used as described before.

Licorice Induced Syndrome Of Apparent Mineralocorticoid Excess 

Licorice root extracts are used as a herbal medicine for several conditions like cough, peptic ulceration, etc. Licorice is also used as a sweetener and mouth freshener particularly in tropical regions (53). Licorice possesses some glucocorticoid activity, antiandrogen effect, estrogenic activity, and mineralocorticoid like activity. Subjects consuming excessive licorice may develop hypertension and hypokalemia (53). Sometimes this is severe enough to cause a cardiac arrhythmia. While screening for primary aldosteronism for subjects presenting with hypertension and hypokalemia, plasma aldosterone and plasma rennin activity are found to be suppressed in patients using licorice (53). 

                                                                                                                                                                           The active ingredient of liquorice is glycyrrhizic acid, which is hydrolyzed into glycyrrhetinic acid in vivo. Glycyrrhetinic acid has a very low affinity for the mineralocorticoid receptor but is a potent competitive inhibitor of the enzyme 11β-HSD type 2 which is preferentially expressed in kidney (54). Hence it may cause acquired 11β-HSD type 2 deficiency. The physiological role of the enzyme 11β-HSD type 2 is to inactivate cortisol to cortisone and thereby preventing access of cortisol to mineralocorticoid receptor. Cortisol and aldosterone have equipotent stimulating activity on mineralocorticoid receptor (54). Hence any situation associated with suppressed 11β-HSD type 2 activities may lead to overstimulation of mineralocorticoid receptors by cortisol, leading to hypertension with hypokalemia and metabolic alkalosis. After correction of hypokalemia, the screening test reveals suppressed aldosterone and plasma rennin activity (54). The hypertension is primarily due to sodium and water retention. A careful history for licorice ingestion clinches the diagnosis.

Treatment consists of avoidance of licorice products. In the interim period patients should be treated with oral potassium and spironolactone after the completion of screening of aldosterone rennin ratio (ARR). Withdrawal of licorice, even after prolonged use or ingestion of large amounts, leads to a complete resolution of the symptoms of acquired apparent mineralocorticoid excess (55).

REFERENCES

  1. Upadhyay J, Sudhindra P, Abraham G, Trivedi N. Tuberculosis of the adrenal gland: a case report and review of the literature of infections of the adrenal gland. Int J Endocrinol. 2014;2014:876037. doi: 10.1155/2014/876037. Epub 2014 Aug 6. PMID: 25165474; PMCID: PMC4138934.
  2. Kelestimur F. The endocrinology of adrenal tuberculosis: the effects of tuberculosis on the hypothalamo-pituitary-adrenal axis and adrenocortical function. J Endocrinol Invest. 2004 Apr;27(4):380-6. doi: 10.1007/BF03351067. PMID: 15233561.
  3. Vinnard C, Blumberg EA. Endocrine and Metabolic Aspects of Tuberculosis. Microbiol Spectr. 2017 Jan;5(1):10.1128/microbiolspec.TNMI7-0035-2016. doi: 10.1128/microbiolspec.TNMI7-0035-2016. PMID: 28233510; PMCID: PMC5785104.
  4. Lam KY, Lo CY. A critical examination of adrenal tuberculosis and a 28-year autopsy experience of active tuberculosis. Clin Endocrinol (Oxf). 2001 May;54(5):633-9. doi: 10.1046/j.1365-2265.2001.01266.x. PMID: 11380494.
  5. Neogi S, Mukhopadhyay P, Sarkar N, Datta PK, Basu M, Ghosh S. Overt and Subclinical Adrenal Insufficiency in Pulmonary Tuberculosis. Endocr Pract. 2020 Dec 14:S1530-891X(20)48391-0. doi: 10.1016/j.eprac.2020.11.012. Epub ahead of print. PMID: 33645514.
  6. Paolo WF Jr, Nosanchuk JD. Adrenal infections. Int J Infect Dis. 2006 Sep;10(5):343-53. doi: 10.1016/j.ijid.2005.08.001. Epub 2006 Feb 17. PMID: 16483815; PMCID: PMC7110804.
  7. Beishuizen A, Thijs LG. Endotoxin and the hypothalamo-pituitary-adrenal (HPA) axis. J Endotoxin Res. 2003;9(1):3-24. doi: 10.1179/096805103125001298. PMID: 12691614.
  8. Denny N, Raghunath S, Bhatia P, Abdelaziz M. Rifampicin-induced adrenal crisis in a patient with tuberculosis: a therapeutic challenge. BMJ Case Rep. 2016 Nov 29;2016:bcr2016216302. doi: 10.1136/bcr-2016-216302. PMID: 27899384; PMCID: PMC5175016.
  9. Stefan R. Bornstein, Bruno Allolio, Wiebke Arlt, Andreas Barthel, Andrew Don-Wauchope, Gary D. Hammer, Eystein S. Husebye, Deborah P. Merke, M. Hassan Murad, Constantine A. Stratakis, David J. Torpy, Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline, The Journal of Clinical Endocrinology & Metabolism, Volume 101, Issue 2, 1 February 2016, Pages 364–389, https://doi.org/10.1210/jc.2015-1710
  10. Liatsikos EN, Kalogeropoulou CP, Papathanassiou Z, Tsota I, Athanasopoulos A, Perimenis P, Barbalias GA, Petsas T. Primary adrenal tuberculosis: role of computed tomography and CT-guided biopsy in diagnosis. Urol Int. 2006;76(3):285-7. doi: 10.1159/000091637. PMID: 16601397.
  11. Kyriazopoulou V, Parparousi O, Vagenakis AG. Rifampicin-induced adrenal crisis in addisonian patients receiving corticosteroid replacement therapy. J Clin Endocrinol Metab. 1984 Dec; 59(6):1204-6. doi: 10.1210/jcem-59-6-1204. PMID: 6490796.
  12. Bhatia E, Jain SK, Gupta RK, Pandey R. Tuberculous Addison's disease: lack of normalization of adrenocortical function after anti-tuberculous chemotherapy. Clin Endocrinol (Oxf). 1998 Mar;48(3):355-9. doi: 10.1046/j.1365-2265.1998.00409.x. PMID: 9578827.
  13. Laway, B.A., Mir, S.A., Ganie, M.A. et al. Nonreversal of adrenal hypofunction after treatment of adrenal tuberculosis. Egypt J Intern Med 27, 42–44 (2015). https://doi.org/10.4103/1110-7782.155860
  14. Penrice J, Nussey SS. Recovery of adrenocortical function following treatment of tuberculous Addison's disease. Postgrad Med J. 1992 Mar; 68(797):204-5. doi: 10.1136/pgmj.68.797.204. PMID: 1589379; PMCID: PMC2399240.
  15. Kelestimur F. Recovery of adrenocortical function following treatment of tuberculous Addison's disease. Postgrad Med J (1993) 69, 832-34
  16. Roxas MCA, Sandoval MAS, Salamat MS, Matias PJ, Cabal NP, Bartolo SS. Bilateral adrenal histoplasmosis presenting as adrenal insufficiency in an immunocompetent host in the Philippines. BMJ Case Rep. 2020 May 12;13(5):e234935. doi: 10.1136/bcr-2020-234935. PMID: 32404324; PMCID: PMC7228487.
  17. Bhansali A, Das S, Dutta P, Walia R, Nahar U, Singh SK, Vellayutham P, Gopal S. Adrenal histoplasmosis: unusual presentations. J Assoc Physicians India. 2012 Oct;60:54-8. PMID: 23777028.
  18. Jayathilake WAPP, Kumarihamy KWMPP, Ralapanawa DMPUK, Jayalath WATA, "A Rare Presentation of Possible Disseminated Histoplasmosis with Adrenal Insufficiency Leading to Adrenal Crisis in an Immunocompetent Adult: A Case Report", Case Reports in Medicine, vol. 2020, Article ID 8506746, 5 pages, 2020. https://doi.org/10.1155/2020/8506746
  19. Vyas S, Kalra N, Das PJ, Lal A, Radhika S, Bhansali A, Khandelwal N. Adrenal histoplasmosis: An unusual cause of adrenomegaly. Indian J Nephrol. 2011 Oct;21(4):283-5. doi: 10.4103/0971-4065.78071. PMID: 22022092; PMCID: PMC3193675.
  20. Mukherjee JJ, Villa ML, Tan L, Lee KO. Bilateral adrenal masses due to histoplasmosis. J Clin Endocrinol Metab. 2005 Dec; 90(12):6725-6. doi: 10.1210/jc.2005-1868. PMID: 16330806.
  21. Robinson LJ, Lu M, Elsayed S, Joy TR. Bilateral adrenal histoplasmosis manifesting as primary adrenal insufficiency. CMAJ. 2019 Nov 4;191(44):E1217-E1221. doi: 10.1503/cmaj.190710. PMID: 31685665; PMCID: PMC6834444.
  22. Kothari D, Chopra S, Bhardwaj M, Ajmani AK, Kulshreshtha B. Persistence of histoplasma in adrenals 7 years after antifungal therapy. Indian J Endocrinol Metab. 2013 May;17(3):529-31. doi: 10.4103/2230-8210.111679. PMID: 23869317; PMCID: PMC3712391.
  23. de Oliveira FM, Fragoso MCBV, Meneses AF, Vilela LAP, Almeida MQ, Palhares RB, de Arruda Mattos TV, Scalissi NM, Viana Lima J. Adrenal insufficiency caused by Paracoccidioidomycosis: three case reports and review. AACE Clin Case Rep. 2019 Mar 13;5(4):e238-e243. doi: 10.4158/ACCR-2018-0632. PMID: 31967043; PMCID: PMC6873835.
  24. Cataño J, Porras J. Adrenal Paracoccidioidomycosis. Am J Trop Med Hyg. 2020 Sep;103(3):935-936. doi: 10.4269/ajtmh.20-0083. PMID: 32896237; PMCID: PMC7470546.
  25. Tobón AM, Agudelo CA, Restrepo CA, Villa CA, Quiceno W, Estrada S, Restrepo A. Adrenal function status in patients with paracoccidioidomycosis after prolonged post-therapy follow-up. Am J Trop Med Hyg. 2010 Jul; 83(1):111-4. doi: 10.4269/ajtmh.2010.09-0634. PMID: 20595488; PMCID: PMC2912586.
  26. Kumar A, Sreehari S, Velayudhan K, Biswas L, Babu R, Ahmed S, Sharma N, Kurupath VP, Jojo A, Dinesh KR, Karim S, Biswas R. Autochthonous blastomycosis of the adrenal: first case report from Asia. Am J Trop Med Hyg. 2014 Apr;90(4):735-9. doi: 10.4269/ajtmh.13-0444. Epub 2014 Feb 3. PMID: 24493676; PMCID: PMC3973522.
  27. Rimondi AP, Bianchini E, Barucchello G, Panzavolta R. Addison's disease caused by adrenal blastomycosis: a case report with fine needle aspiration (FNA) cytology. Cytopathology. 1995 Aug;6(4):277-9. doi: 10.1111/j.1365-2303.1995.tb00480.x. PMID: 8520008.
  28. Matsuda Y, Kawate H, Okishige Y, Abe I, Adachi M, Ohnaka K, Satoh N, Inokuchi J, Tatsugami K, Naito S, Nomura M, Takayanagi R. Successful management of cryptococcosis of the bilateral adrenal glands and liver by unilateral adrenalectomy with antifungal agents: a case report. BMC Infect Dis. 2011 Dec 14; 11:340. doi: 10.1186/1471-2334-11-340. PMID: 22166121; PMCID: PMC3254187.
  29. Ito M, Hinata T, Tamura K, Koga A, Ito T, Fujii H, Hirata F, Sakuta H. Disseminated Cryptococcosis with Adrenal Insufficiency and Meningitis in an Immunocompetent Individual. Intern Med. 2017; 56(10):1259-1264. doi: 10.2169/internalmedicine.56.7356. Epub 2017 May 15. PMID: 28502948; PMCID: PMC5491828.
  30. Muraoka Y, Iwama S, Arima H. Normalization of Bilateral Adrenal Gland Enlargement after Treatment for Cryptococcosis. Case Rep Endocrinol. 2017; 2017:1543149. doi: 10.1155/2017/1543149. Epub 2017 Mar 26. PMID: 28458934; PMCID: PMC5385225.
  31. Keleştimur F. Sheehan's syndrome. Pituitary. 2003;6(4):181-8. doi: 10.1023/b:pitu.0000023425.20854.8e. PMID: 15237929.
  32. Karaca Z, Laway BA, Dokmetas HS, Atmaca H, Kelestimur F. Sheehan syndrome. Nat Rev Dis Primers. 2016 Dec 22; 2:16092. doi: 10.1038/nrdp.2016.92. PMID: 28004764.
  33. Mandal S, Mukhopadhyay P, Banerjee M, Ghosh S. Clinical, endocrine, metabolic profile, and bone health in Sheehan’s syndrome. Indian J Endocr Metab 2020; 24:338-42.
  34. Hartman ML, Crowe BJ, Biller BM, Ho KK, Clemmons DR, Chipman JJ; HyposCCS Advisory Board; U.S. HypoCCS Study Group. Which patients do not require a GH stimulation test for the diagnosis of adult GH deficiency? J Clin Endocrinol Metab. 2002 Feb; 87(2):477-85. doi: 10.1210/jcem.87.2.8216. PMID: 11836272.
  35. Kilicli F, Dokmetas HS, Acibucu F. Sheehan's syndrome. Gynecol Endocrinol. 2013 Apr;29(4):292-5. doi: 10.3109/09513590.2012.752454. Epub 2012 Dec 18. PMID: 23245206.
  36. Bhat S, Mukhopadhyay P, Raychaudhury A, Chowdhury S, Ghosh S. Predictors of hypopituitarism due to vasculotoxic snake bite with acute kidney injury. Pituitary. 2019 Dec; 22(6):594-600. doi: 10.1007/s11102-019-00990-8. PMID: 31556012
  37. Rajagopala S, Thabah MM, Ariga KK, Gopalakrishnan M. Acute hypopituitarism complicating Russell's viper envenomation: case series and systematic review. QJM. 2015 Sep;108(9):719-28. doi: 10.1093/qjmed/hcv011. Epub 2015 Jan 27. PMID: 25630907.
  38. Shivaprasad C, Aiswarya Y, Sridevi A, Anupam B, Amit G, Rakesh B, Annie PA, Anish K. Delayed hypopituitarism following Russell's viper envenomation: a case series and literature review. Pituitary. 2019 Feb; 22(1):4-12. doi: 10.1007/s11102-018-0915-1. PMID: 30317419.
  39. Benjamin JM, Chippaux JP, Sambo BT, Massougbodji A. Delayed double reading of whole blood clotting test (WBCT) results at 20 and 30 minutes enhances diagnosis and treatment of viper envenomation. J Venom Anim Toxins Incl Trop Dis. 2018 May 16; 24:14. doi: 10.1186/s40409-018-0151-1. PMID: 29796013; PMCID: PMC5956810
  40. Tun-Pe, Phillips RE, Warrell DA, Moore RA, Tin-Nu-Swe, Myint-Lwin, Burke CW. Acute and chronic pituitary failure resembling Sheehan's syndrome following bites by Russell's viper in Burma. Lancet. 1987 Oct 3;2(8562):763-7. doi: 10.1016/s0140-6736(87)92500-1. PMID: 2888987.
  41. Naik BN, Bhalla A, Sharma N, Mokta J, Singh S, Gupta P, Rai A, Subbiah S, Bhansali A, Dutta P (2018) Pituitary dysfunction in survivors of Russell’s viper snake bite envenomation: A prospective study. Neurol India 66(5):1351
  42. De Sanctis V, Soliman AT, Elsedfy H, Skordis N, Kattamis C, Angastiniotis M, Karimi M, Yassin MA, El Awwa A, Stoeva I, Raiola G, Galati MC, Bedair EM, Fiscina B, El Kholy M. Growth and endocrine disorders in thalassemia: The international network on endocrine complications in thalassemia (I-CET) position statement and guidelines. Indian J Endocrinol Metab. 2013 Jan;17(1):8-18. doi: 10.4103/2230-8210.107808. PMID: 23776848; PMCID: PMC3659911.
  43. Saraogi RK, Chowdhury S, Mukherjee S, Roy S, Chatterjee P. Adrenal extramedullary haematopoietic tumor in HbE Thalassemia. BMJ 2002; 18(7). South Asia Edition.
  44. Tiosano D, Hochberg Z. Endocrine complications of thalassemia. J Endocrinol Invest. 2001 Oct; 24(9):716-23. doi: 10.1007/BF03343916. PMID: 11716158.
  45. De P, Mistry R, Wright C, Pancham S, Burbridge W, Gangopadhayay K, Pang T, Das D. A Review of Endocrine Disorders in Thalassaemia. OJEMD 2014; 4(2).
  46. Bhatia E. Adrenal disorders in people with HIV: The highs and lows. Indian J Med Res. 2018 Feb; 147(2):125-127. doi: 10.4103/ijmr.IJMR_1087_17. PMID: 29806599; PMCID: PMC5991116.
  47. Eledrisi MS, Verghese AC. Adrenal insufficiency in HIV infection: a review and recommendations. Am J Med Sci. 2001 Feb; 321(2):137-44. doi: 10.1097/00000441-200102000-00005. PMID: 11217816.
  48. Bakari Adamu Girei, Sani-Bello Fatima. Endocrine Manifestations of HIV Infection.2013. http://dx.doi.org/10.5772/52684
  49. Unachukwu CN, Uchenna DI, Young EE. Endocrine and metabolic disorders associated with human immune deficiency virus infection. West Afr J Med. 2009 Jan;28(1):3-9. doi: 10.4314/wajm.v28i1.48415. PMID: 19662737.
  50. Hopkins RL, Leinung MC. Exogenous Cushing's syndrome and glucocorticoid withdrawal. Endocrinol Metab Clin North Am. 2005 Jun;34(2):371-84, ix. doi: 10.1016/j.ecl.2005.01.013. PMID: 15850848.
  51. http://www.meditecno.pt/Upload/Product/Archive/lkco1.pdf . Last accessed on 14.4.2021
  52. Patt H, Bandgar T, Lila A, Shah N. Management issues with exogenous steroid therapy. Indian J Endocrinol Metab. 2013 Dec; 17(Suppl 3):S612-7. doi: 10.4103/2230-8210.123548. PMID: 24910822; PMCID: PMC4046616.
  53. Palermo M, Quinkler M, Stewart PM. Apparent mineralocorticoid excess syndrome: an overview. Arq Bras Endocrinol Metabol. 2004 Oct; 48(5):687-96. doi: 10.1590/s0004-27302004000500015. Epub 2005 Mar 7. PMID: 15761540.
  54. Farese RV Jr, Biglieri EG, Shackleton CH, Irony I, Gomez-Fontes R. Licorice-induced hypermineralocorticoidism. N Engl J Med. 1991 Oct 24;325(17):1223-7. doi: 10.1056/NEJM199110243251706. PMID: 1922210.
  55. Gallacher SD, Tsokolas G, Dimitropoulos I. Liquorice-induced apparent mineralocorticoid excess presenting in the emergency department. Clin Med (Lond). 2017 Feb; 17(1):43-45. doi: 10.7861/clinmedicine.17-1-43. PMID: 28148579; PMCID: PMC6297599.

 

Hyperglycemic Crises: Diabetic Ketoacidosis And Hyperglycemic Hyperosmolar State

ABSTRACT

Diabetic ketoacidosis (DKA) and hyperglycemic hyperosmolar state (HHS) are acute metabolic complications of diabetes mellitus that can occur in patients with both type 1 and 2 diabetes mellitus. Timely diagnosis, comprehensive clinical and biochemical evaluation, and effective management is key to the successful resolution of DKA and HHS. Critical components of the hyperglycemic crises’ management include coordinating fluid resuscitation, insulin therapy, and electrolyte replacement along with the continuous patient monitoring using available laboratory tools to predict the resolution of the hyperglycemic crisis. Understanding and prompt awareness of potential special situations such as DKA or HHS presentation in the comatose state, possibility of mixed acid-base disorders obscuring the diagnosis of DKA, and risk of brain edema during therapy are important to reduce the risks of complications without affecting recovery from hyperglycemic crisis. Identification of factors that precipitated DKA or HHS during the index hospitalization should help prevent subsequent episode of hyperglycemic crisis.

INTRODUCTION

Diabetic ketoacidosis (DKA) and hyperosmolar hyperglycemic state (HHS) represent two extremes in the spectrum of decompensated diabetes. DKA and HHS remain important causes of morbidity and mortality among diabetic patients despite well-developed diagnostic criteria and treatment protocols (1). The annual incidence of DKA from population-based studies in 1980s was estimated to range from 4 to 8 episodes per 1,000 patient admissions with diabetes (2); the annualized incidence remains stable based on the 2017 national inpatient sample analysis (3). Overall, the incidence of DKA admissions in the US continues to increase, accounting  for about 140,000 hospitalizations in 2009 (Figure 1 a), 168,000 hospitalizations in 2014 (4,5), and most recently 220,340 admissions in 2017 (3) with similar trends observed in England (6) and Finland (7). The 2014 DKA hospitalization rates were the highest in persons aged <45 years (44.3 per 1,000) and lowest in persons aged ≥65 years (<2.0 per 1,000) (5); the age-related admission patterns remained the same in the 2017 analyses (3). The rate of hospital admissions for HHS is lower than of DKA and is less than 1% of all diabetic-related admissions (8,9). About 2/3 of adults presenting to the emergency department or admitted with DKA have a past history of type 1 diabetes (T1D), while almost 90% of the HHS patients have a known diagnosis of type 2 diabetes (T2D) (5). In 2014, there were reported 207,000 emergency department visits with a diagnosis of hyperglycemic crisis (10). Decompensated diabetes imposes a heavy burden in terms of economics and patient outcomes. DKA is responsible for more than 500,000 hospital days per year at an estimated annual direct medical expense and indirect cost of 2.4 billion USD in 1997 (CDC) (11). The cost of inpatient DKA care in the US has increased to 5.1 billion USD in 2014, corresponding to approximate charges related to DKA care varying between 20-26 thousand USD per admission (12,13) and continued to increase in 2017 when DKA admissions costed healthcare about 6.76 billion USD, corresponding to about 31 thousand USD per each admission (3). The mortality rate for DKA and hyperglycemic crises has been falling over the years (Figure 1b) (4) with estimates of fatality remaining under 1% for DKA (3); mortality can reach up to 20% in HHS (14). In 2010, among adults aged 20 years or older, hyperglycemic crisis caused 2,361 deaths (15). There was a decline in mortality from 2000 to 2014 across all age groups and both sexes with largest absolute decrease among persons aged ≥75 years (5). The mortality rate of HHS is higher, reaching 10-20% depending on associated comorbidities and severity of the initial presentation compared with DKA (14,16,17) and is highest in those with DKA+HHS (18). Severe dehydration, older age, and the presence of comorbid conditions in patients with HHS account for the higher mortality in these patients (17). Recent analyses suggested that patients who are black, female, and/or having Medicaid insurance had the highest risk of being admitted with DKA (3). 

Figure 1. Hyperglycemic Crises. A) Incidence of DKA 1980-2009 B) Crude and Age-Adjusted Death Rates for Hyperglycemic Crises as Underlying Cause per 100,000 Diabetic Population, United States, 1980–2009 C) Age-Adjusted DKA hospitalization rate per 1,000 persons with diabetes and in-hospital case-fatality rate, United States, 2000–2014 (5).

DEFINITIONS

DKA consists of the biochemical triad of hyperglycemia, ketonemia, and high anion gap metabolic acidosis (19) (Figure 2). The terms “hyperglycemic hyperosmolar nonketotic state” and “hyperglycemic hyperosmolar nonketotic coma” have been replaced with the term “hyperglycemic hyperosmolar state” (HHS) to highlight that 1) the hyperglycemic hyperosmolar state may consist of moderate to variable degrees of clinical ketosis detected by nitroprusside method, and 2) alterations in consciousness may often be present without coma.

 Figure 2. The triad of DKA (hyperglycemia, acidemia, and ketonemia) and other conditions with which the individual components are associated. From Kitabchi and Wall (19).

Both DKA and HHS are characterized by hyperglycemia and absolute or relative insulinopenia. Clinically, they differ by the severity of dehydration, ketosis, and metabolic acidosis (17).

DKA most often occurs in patients with T1D. It also occurs in T2D under conditions of extreme stress, such as serious infection, trauma, cardiovascular or other emergencies, and, less often, as a presenting manifestation of T2D, a disorder called ketosis-prone T2D (16). Similarly, whereas HHS occurs most commonly in T2D, it can be seen in T1D in conjunction with DKA. Presentations with overlapping DKA and HHS accounted for 27% of admissions for hyperglycemic crises based on one report (18).

PATHOGENESIS

The underlying defects in DKA and HHS are 1) reduced net effective action of circulating insulin as a result of decreased insulin secretion (DKA) or ineffective action of insulin in HHS (20-22), 2) elevated levels of counter regulatory hormones: glucagon (23,24), catecholamines (23,25), cortisol (23), and growth hormone (26,27), resulting in increased hepatic glucose production and impaired glucose utilization in peripheral tissues, and 3) dehydration and electrolyte abnormalities, mainly due to osmotic diuresis caused by glycosuria (28) (Figure 3). Diabetic ketoacidosis is also characterized by increased gluconeogenesis, lipolysis, ketogenesis, and decreased glycolysis (16).

Diabetic Ketoacidosis

In DKA, there is a severe alteration of carbohydrate, protein, and lipid metabolism (8). In general, the body is shifted into a major catabolic state with breakdown of glycogen stores, hydrolysis of triglycerides from adipose tissues, and mobilization of amino acids from muscle (16). The released triglycerides and amino acids from the peripheral tissues become substrates for the production of glucose and ketone bodies by the liver (29). Hyperglycemia and ketone bodies production play central roles in developing this metabolic decompensation (30). 

HYPERGLYCEMIA

The hyperglycemia in DKA is the result of three events: (a) increased gluconeogenesis; (b) increased glycogenolysis, and (c) decreased glucose utilization by liver, muscle, and fat. Insulinopenia and elevated cortisol levels also lead to a shift from protein synthesis to proteolysis with resultant increase in production of amino acids (alanine and glutamine), which further serve as substrates for gluconeogenesis (8,31). Furthermore, muscle glycogen is catabolized to lactic acid via glycogenolysis. The lactic acid is transported to the liver in the Cori cycle where it serves as a carbon skeleton for gluconeogenesis (32). Increased levels of glucagon, catecholamines, and cortisol with concurrent insulinopenia stimulate gluconeogenic enzymes, especially phosphoenol pyruvate carboxykinase (PEPCK) (26,33). Decreased glucose utilization is further exaggerated by increased levels of circulating catecholamines and FFA (34).

KETOGENESIS

Excess catecholamines coupled with insulinopenia promote triglyceride breakdown (lipolysis) to free fatty acids (FFA) and glycerol in adipose tissue. The latter provides a carbon skeleton for gluconeogenesis, while the former serves as a substrate for the formation of ketone bodies (35,36). The key regulatory site for fatty acid oxidation is known to be carnitine palmitoyl transferase 1(CPT1) which is inhibited by malonyl CoA in the normal non-fasted state but the increased ratio of glucagon and other counter regulatory hormones to insulin disinhibit fatty acid oxidation and incoming fatty acids from fat tissue can be converted to ketone bodies (37,38). Increased production of ketone bodies (β-hydroxybutyrate and acetoacetate) leads to ketonemia (39). Ketonemia is further maintained by the reduced liver clearance of ketone bodies in DKA. Extracellular and intracellular buffers neutralize hydrogen ions produced during hydrolysis of ketoacids. When overwhelming ketoacid production exceeds buffering capacity, a high anion gap metabolic acidosis develops. Studies in diabetic and pancreatectomized patients have demonstrated the cardinal role of hyperglucagonemia and insulinopenia in the genesis of DKA (40). In the absence of stressful situations, such as intravascular volume depletion or intercurrent illness, ketosis is usually mild (16,41).

Elevated levels of pro-inflammatory cytokines and lipid peroxidation markers, as well as procoagulant factors such as plasminogen activator inhibitor-1 (PAI-1) and C-reactive protein (CRP) have been demonstrated in DKA. The levels of these factors return to normal after insulin therapy and correction of hyperglycemia (42). This inflammatory and procoagulant state may explain the well-known association between hyperglycemic crisis and thrombotic state (43,44).

Hyperglycemic Hyperosmolar State

While DKA is a state of near absolute insulinopenia, there is sufficient amount of insulin present in HHS to prevent lipolysis and ketogenesis but not adequate to cause glucose utilization (as it takes 1/10 as much insulin to suppress lipolysis as it does to stimulate glucose utilization) (33,34). In addition, in HHS there is a smaller increase in counter regulatory hormones (20,45).

Figure 3. Pathogenesis of DKA and HHS: stress, infection, or insufficient insulin. FFA, free fatty acid. Adapted from Kitabchi et al. (1).

PRECEPITATING FACTORS

The two most common precipitating factors in the development of DKA or HHS are inadequate insulin therapy (whether omitted or insufficient insulin regimen) or the presence of infection (46,47). Other provoking factors include myocardial infarction, cerebrovascular accidents, pulmonary embolism, pancreatitis, alcohol and illicit drug use (Figure 4). In addition, numerous underlying medical illness and medications that cause the release of counter regulatory hormones and/or compromise the access to water can result in severe volume depletion and HHS (46). Drugs such as corticosteroids, thiazide diuretics, sympathomimetic agents (e.g., dobutamine and terbutaline), and second generation antipsychotic agents may precipitate DKA or HHS (17). Most recently, two new classes of medications have emerged as triggers for DKA. Sodium-glucose cotransporter 2 (SGLT-2) inhibitors (canagliflozin, dapagliflozin, and empagliflozin) that are used for diabetes treatment have been implicated in the development of DKA in patients with both T1D and T2D (48). Though the absolute risk of DKA in patients treated with SGLT-2 inhibitors is small, this class of medications raises DKA risk by 2-4-fold in patients withT2D and its incidence can be up to 5% in patients with T1D (49,50). Also, anti-cancer medications that belong to classes of immune checkpoint inhibitors such as Ipilimumab, Nivolumab, Pembrolizumab, can cause new-onset diabetes mellitus in up to 1% of the patients receiving immune checkpoint inhibitors with about half of these patients presenting with DKA as the initial presentation of diabetes, particularly in those individuals who may have underlying beta-cell autoimmunity  (51,52) (53-56). In young patients with T1D, insulin omission due to fear of hypoglycemia or weight gain, the stress of chronic disease, and eating disorders, may contribute in 20% of recurrent DKA (57). Cocaine use also is associated with recurrent DKA (58,59). Mechanical problems with continuous subcutaneous insulin infusion (CSII) devices can precipitate DKA (60); however, with an improvement in technology and better education of patients, the incidence of DKA have been declining in insulin pump users (61). There are also case reports of patients with DKA as the primary manifestation of acromegaly (62-64).

Increasing numbers of DKA cases have been reported in patients with Type 2 DM. Available evidence shows that almost 50 % of newly diagnosed adult African American and Hispanic patients with DKA have T2D (65). These ketosis-prone type 2 diabetic patients develop sudden-onset impairment in insulin secretion and action, resulting in profound insulinopenia (66). Clinical and metabolic features of these patients include high rates of obesity, a strong family history of diabetes, a measurable pancreatic insulin reserve, and a low prevalence of autoimmune markers of β-cell destruction (67-69). Aggressive management with insulin improves β-cell function, leading to discontinuance of insulin therapy within a few months of follow-up and 40 % of these patients remain non-insulin dependent for 10 years after the initial episode of DKA (68). The etiology of acute transient failure of β-cells leading to DKA in these patients is not known, however, the suggested mechanisms include glucotoxicity, lipotoxicity, and genetic predisposition (70,71).  A genetic disease, glucose-6-phosphate dehydrogenase deficiency, has been also linked with ketosis-prone diabetes (72). In a most recent review of factors that can precipitate DKA, the authors emphasized that clinicians should consider factors such as socioeconomic disadvantage, adolescent age, female sex, prior DKA, and psychiatric comorbidities as potential DKA triggers in patients with T1D (50). Further, in US adults with T1D, HbA1c ³ 9% was associated with 12-fold higher incidence of DKA (73). Finally, with recent accumulation of knowledge of health hazards related to the COVID-19 pandemic, there is early evidence that COVID-19 infection can trigger DKA in patients with diabetes who otherwise may not have risk factors to develop ketoacidosis (74). Particular attention should be provided to those DKA patients who are COVID-19-positive on admission as early evidence demonstrated a 6-fold increase in mortality in this group of patients compared with those admitted with DKA without COVID-19 (75).

With growing use of SGLT-2 inhibitors, it is worth elucidating potential risk factors that can mediate heightened DKA risk in patients with diabetes. It is now clear that T1D is an independent DKA risk factor regardless of whether other clinical circumstances known to trigger ketoacidosis are present or not. In people with T2D, low-carbohydrate diet, excessive ETOH intake, presence of autoimmunity, and/or exposure to stress situations such as infection, surgery, trauma, dehydration are now identified as DKA risk factors in those treated with SGLT-2 inhibitors (50,76). 

 

Figure 4. Common precipitating factors in DKA. Data are % of all cases except Nyenwe et al where new onset disease was not included in the percentage and complete data on these items were not given; therefore, the total is less than 100%. Adapted with modification from reference 1.

CLINICAL FEATURES 

Symptoms and Signs

DKA usually evolves rapidly within a few hours of the precipitating event(s). On the other hand, development of HHS is insidious and may occur over days to weeks (16). The common clinical presentation of DKA and HHS is due to hyperglycemia and include polyuria, polyphagia, polydipsia, weight loss, weakness, and physical signs of intravascular volume depletion, such as dry buccal mucosa, sunken eye balls, poor skin turgor, tachycardia, hypotension and shock in severe cases. Of note, patients with euglycemic DKA including those treated with SGLT-2 inhibitors, may have less polydipsia and polyuria and may rather initially present with non-specific symptoms such as fatigue and malaise (77,78). Kussmaul respiration, acetone breath, nausea, vomiting, and abdominal pain may also occur primarily in DKA and are due to ketosis and acidosis. Abdominal pain, which correlates with the severity of acidosis (79), may be severe enough to be confused with acute abdomen in 50-75% of cases (80). Therefore, in the presence of acidosis, DKA as an etiology of abdominal pain should be considered. Patients usually have normal body temperature or mild hypothermia regardless of presence of infection (81). Therefore, a careful search for a source of infection should be performed even in the absence of fever. Neurological status in patients with DKA may vary from full alertness to a profound lethargy and coma, However, mental status changes in DKA are less frequent than HHS. The relationship of depressed consciousness and severity of hyperosmolality or DKA causes has been controversial (82,83). Some studies suggested that pH is the cause of mental status changes (84); while, others concluded that osmolality (85) is responsible for the comatose state. More recently, it has been proposed that consciousness level in adolescents with DKA was related to the severity of acidosis (pH) and not to a blood glucose levels (86).  In our earlier studies of patients with DKA using low dose versus high dose insulin therapy, we evaluated the initial biochemical values of 48 patients with stupor/coma versus non comatose patients (87). Our study showed that glucose, bicarbonate, BUN and osmolality, and not pH were significantly different between non-comatose and comatose patients. Furthermore, in 3 separate studies in which 123 cases of DKA were evaluated, serum osmolality was also the most important determinant of mental status changes (19). However, in our recent retrospective study, it was shown that acidosis was independently associated with altered sensorium, but hyperosmolarity and serum “ketone” levels were not (88) (Figure 5). In that study, a combination of acidosis and hyperosmolarity at presentation may identify a subset of patients with severe DKA (7% in this study) who may benefit from more aggressive treatment and monitoring. Identifying this group of patients, who are at a higher risk for poorer prognosis, may be helpful in triaging them, thus further improving the outcome (88). Furthermore, according to one study, ICU-admitted patients with DKA are less ill, and have lower disease severity scores, mortality, and shorter length of ICU and hospital stay, than non-DKA patients. Disease severity scores are not, but precipitating cause is, predictive of prolonged hospital stays in patients with DKA (89).

Figure 5. Admission clinical and biochemical profile in comatose vs non-comatose patients with DKA (88).

In patients with HHS, neurological symptoms include clouding of sensorium which can progress to mental obtundation and coma (90). Occasionally, patients with HHS may present with focal neurological deficits and seizures (91,92). Most of the patients with HHS and an effective serum osmolality of >320 mOsm/kg are obtunded or comatose; on the other hand, the altered mental status rarely exists in patients with serum osmolality of <320 mOsm/kg (8). Therefore, severe alteration in the level of consciousness in patients with serum osmolality of <320 mOsm/kg requires evaluation for other causes including CVA and other catastrophic events like myocardial and bowel infarctions.

LABORATORY ABNORMALITIES AND DIAGNOSIS OF HYPERGLYCEMIC CRISES

The initial laboratory evaluation of patients with suspected DKA or HHS should include determination of plasma glucose, blood urea nitrogen, serum creatinine, serum ketones, electrolytes (with calculated anion gap), osmolality, urinalysis, urine ketones by dipstick, arterial blood gases, and complete blood count with differential. An electrocardiogram, blood, urine or sputum cultures and chest X-ray should also be performed, if indicated. HbA1c may be useful in differentiating chronic hyperglycemia of uncontrolled diabetes from acute metabolic decompensation in a previously well-controlled diabetic patient (17). Figure 6 summarizes the biochemical criteria for DKA and HHS and electrolyte deficits in these two conditions. It also provides a simple method for calculating anion gap and serum osmolality.

Figure 6. Diagnostic Criteria and Typical Total Body Deficits of Water and Electrolytes in Diabetic Ketoacidosis (DKA) and Hyperglycemic Hyperosmolar Syndrome (HHS)

DKA can be classified as mild, moderate, or severe based on the severity of metabolic acidosis and the presence of altered mental status (17). Over 30% of patients have features of both DKA and HHS (16) with most recent evidence confirming that about 1 out of 4 patients will have both conditions at the time of presentation with hyperglycemic crisis (18). Patients with HHS typically have pH >7.30, bicarbonate level >20 mEq/L, and negative ketone bodies in plasma and urine. However, some of them may have ketonemia. Several studies on serum osmolarity and mental alteration have established a positive linear relationship between osmolarity, pH, and mental obtundation (87).  Therefore, the occurrence of coma in the absence of definitive elevation of serum osmolality requires immediate consideration of other causes of mental status change. The levels of β-hydroxybutyrate (β-OHB) of ≥3.8mmol/L measured by a specific assay were shown to be highly sensitive and specific for DKA diagnosis (93). In patients with chronic kidney disease stage 4-5, the diagnosis of DKA could be challenging due to the presence of concomitant underlying chronic metabolic acidosis or mixed acid-base disorders. An anion gap of >20 mEq/L usually supports the diagnosis of DKA in these patients (94). Based on the 2009 American Diabetes Association publication, “euglycemic DKA” is characterized by metabolic acidosis, increased total body ketone concentration and blood glucose levels ≤250 mg/dL and is thought to occur in up to approximately 10% of patients with DKA and mostly associated with conditions associated with low glycogen reserves and/or increased rates of glucosuria such as pregnancy, liver disorders, and alcohol consumption (1). Since approval in 2013 of SGLT-2 inhibitors for therapy of T2D, multiple reports emerged demonstrating that the use of these medications can result in “euglycemic” DKA (48,78,95). Therefore, DKA must be excluded if high anion gap metabolic acidosis is present in a diabetic patient treated with SGLT-2 inhibitors irrespective if hyperglycemia is present or not. On the other hand, an SGLT-2 inhibitor can be also associated with hyperglycemic DKA in individuals who have sufficient glycogen storage to maintain hyperglycemia even in the setting of enhanced glucosuria (49,96).

The major cause of water deficit in DKA and HHS is glucose-mediated osmotic diuresis, which leads to loss of water in excess of electrolytes (97). Despite the excessive water loss, the admission serum sodium tends to be low. Because serum glucose in the presence of insulinopenia of DKA and HHS cannot penetrate to cells, in hyperglycemic crises, glucose becomes osmotically effective and causes water shifts from intracellular space to the extra cellular space resulting in dilution of sodium concentration – dilutional or hyperosmolar hyponatremia. Initially it has been thought that true sodium concentration (millimolar) can be obtained by multiplying excess glucose above 100 mg/dL by 1.6 /100 (98).  It is, however, accepted now that true or corrected serum sodium concentration in patients experiencing hyperglycemic crisis should be calculated by adding 2.4 mmol/L to the measured serum sodium concentration for every 100 mg/dL incremental rise in serum glucose concentration above serum glucose concentration of 100 mg/dL (99). If the corrected sodium level remains low, hypertriglyceridemia (secondary to uncontrolled diabetes) should be also suspected. In this condition the plasma becomes milky and lipemia retinalis may be visible in physical examination (100). Osmotic diuresis and ketonuria also promote a total body sodium deficit via urinary losses, although concurrent conditions, such as diarrhea and vomiting, can further contribute to sodium losses. Total body sodium loss can result in contraction of extracellular fluid volume and signs of intravascular volume depletion. Serum potassium may be elevated on arrival due to insulin deficiency, volume depletion and a shift of potassium from intracellular to extra cellular compartments in response to acidosis (101). However, total body potassium deficit is usually present from urinary potassium losses due to osmotic diuresis and ketone excretion. More frequently, the initial serum potassium level is normal or low which is a danger sign. Initiation of insulin therapy, which leads to the transfer of potassium into cells, may cause fatal hypokalemia if potassium is not replaced early.  Phosphate depletion in DKA is universal but on admission, like the potassium, it may be low, normal or high (102).

The differences and similarities in the admission biochemical data in patients with DKA or HHS are shown in Figure 7.

Figure 7. Biochemical data in patients with HHS and DKA (1).

Leukocytosis is a common finding in patients with DKA or HHS, but leukocytosis greater than 25,000 /μL suggests ongoing infection requiring further work up (103). The exact etiology of this non-specific leukocytosis is not known. One study also showed nonspecific leukocytosis in subjects with hypoglycemia induced by insulin injection and suggested that this phenomenon may be due to the increased levels of catecholamines, cortisol, and proinflammatory cytokines such as TNF-α during acute stress (104). Hypertriglyceridemia may be present in HHS  (105) and is almost always seen in DKA (79).  Hyperamylasemia, which correlates with pH and serum osmolality and elevated level of lipase, may occur in 16 - 25% of patients with DKA (106). The origin of amylase in DKA is usually non-pancreatic tissue such as the parotid gland (107).

Pitfalls of Laboratory Tests and Diagnostic Considerations for Interpreting Acid Based Status in DKA

False positive values for lipase may be seen if plasma glycerol levels are very high due to rapid breakdown of adipose tissue triglycerides (glycerol is the product measured in most assays for plasma lipase). Therefore, elevated pancreatic enzymes may not be reliable for the diagnosis of pancreatitis in the DKA setting. Other pitfalls include artificial elevation of serum creatinine due to interference from ketone bodies when a colorimetric method is used (108). Most of the laboratory tests for ketone bodies use the nitroprusside method, which detects acetoacetate, but not β-hydroxybutyrate (β-OHB). Additionally, since β-OHB is converted to acetoacetate during treatment (109), the serum ketone test may remain positive for a prolonged period suggesting erroneously that ketonemia is deteriorating; therefore, the follow up measurement of ketones during the treatment by nitroprusside method is not recommended (16). Newer glucose meters have the capability to measure β-OHB, which overcomes this problem (110,111). Furthermore drugs that have sulfhydryl groups can interact with the reagent in the nitroprusside reaction, giving a false positive result (112). Particularly important in this regard is captopril, an angiotensin converting enzyme inhibitor prescribed for the treatment of hypertension and diabetic nephropathy. Therefore, for the diagnosis of DKA, clinical judgment and consideration of other biochemical data are required to interpret the value of positive nitroprusside reactions in patients on captopril. Most laboratories can now measure β-OHB levels.

The classical presentation of acid-base disorders in DKA consists of increased anion gap metabolic acidosis where the relation of plasma anion gap change and bicarbonate change (Δ-Δ, ratio of AG change over change in bicarbonate) equals to 1 due to parallel reduction in plasma bicarbonate with the addition of ketoacids into the extravascular fluid space. With frequent additional bicarbonate losses in urine in the form of ketoanions during DKA, the initiation of intravenous volume resuscitation with chloride-containing solutions can further lower plasma bicarbonate and unmask non-anion gap metabolic acidosis when Δ-Δ becomes less than 1 due to changes in plasma bicarbonate that exceed the expected changes in AG. Respiratory compensation will accompany metabolic acidosis with reduction in PCO2 in arterial blood gas. The expected changes in PCO2 can be calculated using Winter’s formula: PCO2 (mmHg) = 1.5 (Bicarbonate) + 8 ± 2 (113). Therefore, inappropriately high or low levels of PCO2, determined by ABG will suggest the presence of a mixed acid-based disorder. For example, DKA patients with concomitant fever or sepsis may have additional respiratory alkalosis manifesting by lower-than-expected PCO2. In contrast, a higher than calculated PCO2 level signifies additional respiratory acidosis and can be seen in patients with underlying chronic lung disease.  Vomiting is a common clinical manifestation in DKA and leads to a loss of hydrogen ions in gastric content and the development of metabolic alkalosis. Patients with DKA and vomiting may have relatively normal plasma bicarbonate levels and close to normal pH. However, AG will remain elevated and be an important clue for DKA. In addition, Δ-Δ ratio will be over 2 suggesting that there is less than expected reduction in bicarbonate as compared with increase in AG and confirm the presence of a mixed acid-base disorder (combination of metabolic acidosis and metabolic alkalosis). We recommend measurement of β-OHB in instances when a mixed acid-base disorder is present in patients with hyperglycemic crisis and DKA is suspected. 

DIFFERENTIAL DIAGNOSIS

Patients may present with metabolic conditions resembling DKA or HHS. For example, in alcoholic ketoacidosis (AKA), total ketone bodies are much greater than in DKA with a higher β-OHB to acetoacetate ratio of 7:1 versus a ratio of 3:1 in DKA (8). The AKA patients seldom present with hyperglycemia (114). It is also possible that patients with a low food intake may present with mild ketoacidosis (starvation ketosis); however, serum bicarbonate concentration of less than 18 or hyperglycemia will be rarely present. Additionally, DKA has to be distinguished from other causes of high anion gap metabolic acidosis including lactic acidosis, advanced chronic renal failure, as well as ingestion of drugs such as salicylate, methanol, and ethylene glycol. Isopropyl alcohol, which is commonly available as rubbing alcohol, can cause considerable ketosis and high serum osmolar gap without metabolic acidosis. Moreover, there is a tendency to hypoglycemia rather than hyperglycemia with isopropyl alcohol injection (115,116). Finally, patients with diabetes insipidus presenting with severe polyuria and dehydration, who are subsequently treated with free water in a form of intravenous dextrose water, can have hyperglycemia- a clinical picture that can be confused with HHS (117) (Figure 8).

Figure 8. Laboratory evaluation of metabolic causes of acidosis and coma (16).

TREATMENT OF DKA

The goals of therapy in patients with hyperglycemic crises include: 1) improvement of circulatory volume and tissue perfusion, 2) gradual reduction of serum glucose and osmolality, 3) correction of electrolyte imbalance, and 4) identification and prompt treatment of co-morbid precipitating causes (8). It must be emphasized that successful treatment of DKA and HHS requires frequent monitoring of patients regarding the above goals by clinical and laboratory parameters. Suggested approaches for the management of patients with DKA and HHS are illustrated in Figures 9 and 10.

Fluid Therapy

DKA and HHS are volume-depleted states with total body water deficit of approximately 6 L in DKA and 9 L in HHS (16,118,119). Therefore, the initial fluid therapy is directed toward expansion of intravascular volume and securing adequate urine flow. The initial fluid of choice is isotonic saline at the rate of 15–20 ml /kg body weight per hour or 1–1.5 L during the first hour. The choice of fluid for further repletion depends on the hydration status, serum electrolyte levels, and urinary output. In patients who are hypernatremic or eunatremic, 0.45% NaCl infused at 4–14 ml/kg/hour is appropriate, and 0.9% NaCl at a similar rate is preferred in patients with hyponatremia. The goal is to replace half of the estimated water and sodium deficit over a period of 12-24 hours [161]. In patients with hypotension, aggressive fluid therapy with isotonic saline should continue until blood pressure is stabilized. The administration of insulin without fluid replacement in such patients may further aggravate hypotension (16).  Furthermore, the use of hydrating fluid in the first hour of therapy before insulin administration provides time to obtain serum potassium value before insulin administration, prevents possible deterioration of hypotensive patients with the use of insulin without adequate hydration, and decreases serum osmolality (17). Hydration alone may also reduce the level of counter-regulatory hormones and hyperglycemia (28). Intravascular volume expansion reduces serum blood glucose, BUN, and potassium levels without significant changes in pH or HCO3.The mechanism for lowering glucose is believed to be due to osmotic diuresis and modulation of counter-regulatory hormone release (23,120). We recommend avoiding too rapid correction of hyperglycemia (which may be associated with cerebral edema especially in children) and also inhibiting hypoglycemia (23,120). In HHS, the reduction in insulin infusion rate and/or use of D5 ½ NS should be started when blood glucose reaches 300 mg/dL, because overzealous use of hypotonic fluids has been associated with the development of cerebral edema (121). In one recent review, authors suggested gradual reduction in osmolality not exceeding 3 mOsm/kg H2O per hour and a fall of serum sodium at a rate of less than 0.5 mmol/L per hour in order to prevent significant osmotic shifts of water to intracellular compartment during the management of hyperglycemic crises (122). It should be emphasized that urinary losses of water and electrolytes are also need to be considered.

Insulin Therapy

The cornerstone of DKA and HHS therapy is insulin in physiologic doses. Insulin should only be started after serum potassium value is > 3.3 mmol/L (8). In DKA, we recommend using intravenous (IV) bolus of regular insulin (0.1 u/kg body weight) followed by a continuous infusion of regular insulin at the dose of 0.1u/kg/hr. The insulin infusion rate in HHS should be lower as major pathophysiological process in these patients is severe dehydration. The optimal rate of glucose reduction is between 50-70 mg/hr. If desirable glucose reduction is not achieved in the first hour, an additional insulin bolus at 0.1 u/kg can be given. As mentioned earlier, when plasma glucose reaches 200-250 mg/dL in DKA or 300 in HHS, insulin rate should be decreased to 0.05 U/kg/hr, followed, as indicated, by the change in hydration fluid to D5 ½ NS. The rate of insulin infusion should be adjusted to maintain blood glucose between 150-200 mg/dL in DKA until it is resolved, and 250-300 mg/dL in HHS until mental obtundation and hyperosmolar state are corrected. 

A study that investigated the optimum route of insulin therapy in DKA demonstrated that the time for resolution of DKA was identical in patients who received regular insulin via intravenous, intramuscular, or subcutaneous routes (123). However, patients who received intravenous insulin showed a more rapid decline in blood glucose and ketone bodies in the first 2 hours of treatment. Patients who received intravenous insulin attained an immediate pharmacologic level of insulin concentration. Thus, it was established that an intravenous loading dose of insulin would be beneficial regardless of the subsequent route of insulin administration during treatment. A follow up study demonstrated that a priming or loading dose given as one half by IV route and another half by intramuscular route was as effective as one dose given intravenously in lowering the level of ketone bodies in the first hour (124). A bolus or priming dose of insulin has been used in a number of studies. The need of such a method, when using intravenous infusion of insulin, is not clear, as there is no prospective randomized study to establish efficacy of bolus or priming dose before infusion of insulin. However, our study in children demonstrated the effectiveness of intravenous injection of insulin without a bolus dose (125). Therefore, it would appear that if intravenous insulin is used, priming or bolus dose insulin might not be necessary. 

Several clinical studies have shown the potency and cost effectiveness of subcutaneous rapid-acting insulin analogs (lispro or aspart) in the management of patients with uncomplicated mild to moderate DKA (126,127). The patients received subcutaneous rapid-acting insulin doses of 0.2 U/kg initially, followed by 0.1 U/kg every 1 hour or an initial dose of 0.3 U/kg followed by 0.2 U/kg every 2 hours until blood glucose was < 250 mg/dL. Then the insulin dose was decreased by half to 0.05, or 0.1 U/kg respectively, and administered every 1 or 2 hours until resolution of DKA. There were no differences in length of hospital stay, total amount of insulin needed for resolution of hyperglycemia or ketoacidosis, or in the incidence of hypoglycemia among treatment groups.  The use of insulin analogs allowed treatment of DKA in general wards or the emergency department and so reduced cost of hospitalization by 30% without any significant changes in hypoglycemic events (126). Similar results have been reported recently in pediatric patients with DKA (128). The administration of continuous IV infusion of regular insulin is the preferred route because of its short half-life and easy titration and the delayed onset of action and prolonged half-life of subcutaneous regular insulin. It is important to point out that the IV use of fast-acting insulin analogs is not recommended for patients with severe DKA or HHS, as there are no studies to support their use. Again, these agents may not be effective in patients with severe fluid depletion since they are given subcutaneously.

Potassium Therapy

Although total-body potassium is depleted (129,130), mild to moderate hyperkalemia frequently seen in patients with DKA is due to acidosis and insulinopenia. Insulin therapy, correction of acidosis, and volume expansion decrease serum potassium concentrations. To prevent hypokalemia, potassium replacement is initiated after serum levels fall below 5.3 mmol/L in patients with adequate urine output (50 ml/h). Adding 20–30 mmol potassium to each liter of infused fluid is sufficient to maintain a serum potassium concentration within the normal range of 4–5 mmol/L (8). Patients with DKA who had severe vomiting or had been on diuretics may present with significant hypokalemia. In such cases, potassium replacement should begin with fluid therapy, and insulin treatment should be postponed until potassium concentration becomes > 3.3 mmol/L in order to prevent arrhythmias and respiratory muscle weakness (131).

Figure 9. Protocol for the management of adult patients with DKA. Adapted from (94).

Bicarbonate Therapy

The use of bicarbonate in treatment of DKA remains controversial. In patients with pH >7.0, insulin therapy inhibits lipolysis and also corrects ketoacidosis without use of bicarbonate. Bicarbonate therapy has been associated with some adverse effects, such as hypokalemia (132), decreased tissue oxygen uptake and cerebral edema (133,134) and delay in the resolution of ketosis (135).  However, patients with severe DKA (low bicarbonate <10 mEq/L, or Pco2 < 12) may experience deterioration of pH if not treated with bicarbonate. A prospective randomized study in patients with pH between 6.9 and 7.1 showed that bicarbonate therapy had no risk or benefit in DKA (136). Therefore, in patients with pH between 6.9 and 7.0, it may be beneficial to give 50 mmol of bicarbonate in 200 ml of sterile water with 10 mmol KCL over two hours to maintain the pH at > 7.0 (8,137,138). Considering the adverse effects of severe acidosis such as impaired myocardial contractility, adult patients with pH < 6.9 should be given 100 mmol sodium bicarbonate in 400 ml sterile water (an isotonic solution) with 20 mmol KCl administered at a rate of 200 ml/h for two hours until the venous pH becomes greater than 7.0. Venous pH should be assessed every 2 hours until the pH rises to 7.0; treatment can be repeated every 2 hours if necessary.

Phosphate Therapy

There is no evidence that phosphate therapy is necessary in treatment for better outcome of DKA (139-142).  However, in patients with potential complications of hypophosphatemia, including cardiac and skeletal muscle weakness, the use of phosphate may be considered (143). Phosphate administration may result in hypocalcemia when used in high dose (139,142).

TREATMENT OF HHS

A similar therapeutic approach can be also recommended for treatment of HHS, but no bicarbonate therapy is needed for HHS, and changing to glucose-containing fluid is done when blood glucose reaches 300 mg/dL.

Figure 10. Protocol for the management of adult patients with HHS.

Severe hyperosmolarity and dehydration associated with insulin resistance and presence of detectable plasma insulin level are the hallmarks of HHS pathophysiology. The main emphasis in the management of HHS is effective volume repletion and normalization of serum osmolality (14). There are no randomized controlled studies that evaluated safe and effective strategies in the treatment of HHS (121). It is important to start HHS therapy with the infusion of normal saline and monitor corrected serum sodium in order to determine appropriate timing of the change to hypotonic fluids. Insulin substitution approach should be very conservative as it is expected that insulin resistance will improve with rehydration. We recommend against rapid decreases in serum glucose and correction of serum sodium in order to avoid untoward effects of shifts in osmolarity on brain volume. This notion should particularly apply in the management of HHS in elderly and patients with multiple medical problems in whom it may not be clear how long these subjects experienced severe hyperglycemia prior to the admission to the hospital.

RESOLUTION OF DKA AND HHS

During follow up, blood should be drawn every 2-4 h for determination of serum electrolytes, glucose, blood urea nitrogen, creatinine, osmolality, and venous pH. After the initial arterial pH is drawn, venous pH can be used to assess the acid/base status. An equivalent arterial pH value is calculated by adding 0.03 to the venous pH value (144). The resolution of DKA is reached when the blood glucose is < 200 mg/dl, serum bicarbonate is ³15 mEq/L, pH is >7.30 and anion gap is ≤12 mEq/L (17). HHS is resolved when serum osmolality is < 320 mOsm/kg with a gradual recovery to mental alertness. The latter may take twice as long as to achieve blood glucose control. Ketonemia typically takes longer to clear than hyperglycemia.

The proposed ADA criteria for DKA resolution include serum glucose level <200 mg/dL and two of the following: serum bicarbonate level ³15 mEq/L, pH >7.3, and anion gap ≤12 mEq/L (1). Therefore, the treatment goal of DKA is to improve hyperglycemia and to stop ketosis with subsequent resolution of acidosis. In this regard, it is important to distinguish ketosis and acidosis, as the two terms are not always synonymous in DKA. Ketoacid production in DKA results in reduction in plasma bicarbonate (HCO3-) levels due to neutralization of hydrogen ion produced during dissociation of ketoacids in the extravascular fluid space. Concomitantly, ketoacid anion is added into extravascular space resulting in anion gap (AG) increase. The change in HCO3- concentration (Δ HCO3-/normal serum HCO3- – observed serum HCO3-) usually corresponds to equal changes in serum anion gap (Δ AG/observed AG – normal AG, both corrected for decreases and increases in plasma albumin concentration). Therefore, the ratio of AG excess to HCO3- deficit (delta-delta, or Δ-Δ) is close to 1 (143,145,146). In most patients with DKA bicarbonate deficit exceeds the addition of ketoanions, even though Δ-Δ ratio remains close to 1 (147). This is observed due to several reasons. First, hyperglycemia-induced osmotic diuresis leads to excretion of large amounts of sodium and potassium ions that is accompanied by the excretion of ketoanions. Ultimately, the amount of excreted ketoanions depends on degree of kidney function preservation with the largest amount of ketoanion loss in patients with relatively preserved glomerular filtration rate (145). Each ketoanion can be converted back to HCO3- during resolution of DKA and, therefore, ketoanion loss results in the loss of HCO3-. Additionally, extravascular fluid space contraction during DKA, leads to elevation of plasma HCO3-. Therefore, intravenous administration of sodium and chloride-containing fluids leads to further HCO3- reduction and hyperchloremic metabolic acidosis (143,145). This is an important point as persistent decrease in plasma HCO3- concentration should not be interpreted as a sign of continuous DKA if ketosis and hyperglycemia are resolving. Although not evaluated in prospective studies, measurement of serial levels of blood beta-hydroxybutyrate (β-OHB) can be useful adjunct to monitor the resolution of DKA (148). The expected fall in β-OHB with the adequate insulin dosing is 1mmol/L/hr; a lower decrease in blood β-OHB may suggest inadequate insulin provision.

Once DKA has resolved, patients who are able to eat can be started on a multiple dose insulin regimen with long-acting insulin and short/rapid acting insulin given before meals as needed to control plasma glucose. Intravenous insulin infusion should be continued for 2 hours after giving the subcutaneous insulin to maintain adequate plasma insulin levels. Immediate discontinuation of intravenous insulin may lead to hyperglycemia or recurrence of ketoacidosis. If the patient is unable to eat, it is preferable to continue the intravenous insulin infusion and fluid replacement. Patients with known diabetes may be given insulin at the dose they were receiving before the onset of hyperglycemic crises. In patients with new onset diabetes, a multi-dose insulin regimen should be started at a dose of 0.5-0.8 U/kg per day, including regular or rapid-acting and basal insulin until an optimal dose is established (17).

COMPLICATIONS

The most common complications of DKA and HHS include hypoglycemia and hypokalemia due to overzealous treatment with insulin and bicarbonate (hypokalemia), but these complications occur infrequently with current low dose insulin regimens. Nevertheless, in a recent retrospective study, both severe hypokalemia defined as K £ 2.5 mEq/L and severe hypoglycemia < 40 mg/dL were significantly and independently associated with increased risk of mortality in patients admitted to the tertiary care center for treatment of hypoglycemic crisis (18). During the recovery phase of DKA, patients commonly develop a short-lived hyperchloremic non-anion gap acidosis, which usually has few clinical consequences (149). Hyperchloremic acidosis is caused by the loss of large amounts of ketoanions, which are usually metabolized to bicarbonate during the evolution of DKA, and excess infusion of chloride containing fluids during treatment (150).

Cerebral edema, a frequently fatal complication of DKA, occurs in 0.7–1.0% of children, particularly those with newly diagnosed diabetes (120). It may also occur in patients with known diabetes and in very young adults usually under 20 years of age (151,152). Cerebral edema has also been reported in patients with HHS, with some cases of mortality (90). Clinically, cerebral edema is characterized by deterioration in the level of consciousness, lethargy, decreased arousal, and headache. Headache is the earliest clinical manifestation of cerebral edema. This is followed by altered level of consciousness and lethargy. Neurological deterioration may lead to seizures, incontinence, pupillary changes, bradycardia, and respiratory arrest. It may be so rapid in onset due to brain stem herniation that no papilledema is found. If deteriorating clinical symptoms occur, the mortality rate may become higher than 70%, with only 7–14% of patients recovering without permanent neurological deficit. Mannitol infusion and mechanical ventilation are used to combat cerebral edema. The cause of cerebral edema is not known with certainty. It may result from osmotically driven movement of water into the central nervous system when plasma osmolality declines too rapidly during treatment of DKA or HHS. As glucose concentration improves following insulin infusion and administration of the intravenous fluids, serum osmotic gradient previously contributed by hyperglycemia reduces which limits water shifts from the intracellular compartment. However, hyperglycemia treatment is associated with “recovery” in serum sodium that restores water transfer between extracellular and intracellular compartments and prevents water accumulation in cells (99). In cases when the serum glucose concentration improves to a greater extent than the serum sodium concentration rises, serum effective osmolality will decrease and may precipitate brain edema (153,154). Although the osmotically mediated mechanism seems most plausible, one study using magnetic resonance imaging (MRI) showed that cerebral edema was due to increased cerebral perfusion (135). Another postulated mechanism for cerebral edema in patients with DKA involves the cell membrane Na+/H+ exchangers, which are activated in DKA. The high H+ level allows more influx of Na+ thus increasing more influx of water to the cell with consequent edema (155). β-hydroxybutyrate and acetoacetate may also play a role in the pathogenesis of cerebral edema. These ketone bodies have been shown to affect vascular integrity and permeability, leading to edema formation (156). In summary, reasonable precautionary measures to decrease the risk of cerebral edema in high-risk patients include 1) avoidance of overenthusiastic hydration and rapid reduction of plasma osmolality and 2) close hemodynamic monitoring (157). Based on the recent reports, particular care should be offered to patients with end stage renal disease as these individuals are more likely to die, to have higher rates of hypoglycemia, or to be volume overloaded when admitted to the hospital with DKA (158).  

Hypoxemia and rarely non-cardiogenic pulmonary edema may complicate the treatment of DKA [242]. Hypoxemia may be related to the reduction in colloid osmotic pressure that leads to accumulation of water in lungs and decreased lung compliance. The pathogenesis of pulmonary edema may be similar to that of cerebral edema suggesting that the sequestration of fluid in the tissues may be more widespread than is thought. Thrombotic conditions and disseminated intravascular coagulation may contribute to the morbidity and mortality of hyperglycemic emergencies (159-161). Prophylactic use of heparin, if there is no gastrointestinal hemorrhage, should be considered.

PREVENTION

About one in five patients with T1D admitted for DKA will be readmitted for DKA within 30 days (162). Several studies suggested that the omission of insulin is one of the most common precipitating factors of DKA, sometimes because patients are socio-economically underprivileged, and may not have access to or afford medical care (163-165). In addition, they may have a propensity to use illicit drugs such as cocaine, which has been associated with recurrent DKA (58), or live in areas with higher food deprivation risk (166). Therefore, it is important to continuously re-assess socio-economic status of patients who had at least one episode of DKA. The most recent data demonstrating a significant increase in DKA hospitalization rates in diabetic persons aged 45 years and younger (10) suggests that this group of patients may require particular attention to understand why they are more vulnerable than others to develop hyperglycemic crisis. Education of the patient about sick day management is very vital to prevent DKA, and should include information on when to contact the health care provider, blood glucose goals, use of insulin, and initiation of appropriate nutrition during illness and should be reviewed with patients periodically. Patients must be advised to continue insulin and to seek professional advice early in the course of the illness. COVID-19-positive patients with diabetes outside of the hospital environment should be particularly vigilant in point-of-care monitoring of home blood glucose and/or β-OHB until the resolution of infection. Close follow up is very important, as it has been shown that three-monthly visits to the endocrine clinic will reduce the number of ER admission for DKA (167). Close observation, early detection of symptoms and appropriate medical care would be helpful in preventing HHS in the elderly.

A study in adolescents with T1D suggests that some of the risk factors for DKA include higher HbA1c, uninsured children, and psychological problems (168). In other studies, education of primary care providers and school personnel in identifying the signs and symptoms of DKA has been shown to be effective in decreasing the incidence of DKA at the onset of diabetes (169). In another study outcome data of 556 patients with diabetes under continuing care over a 7-year period were examined. The hospitalization rates for DKA and amputation were decreased by 69 % due to continuing care and education (170). There is early evidence that use of continuous glucose monitoring (CGM) can decrease DKA incidence (171,172). Contrary to the initial observations connecting DKA episodes with insulin pump malfunction, the newer pumps are associated with reduced DKA risk without or with concomitant CGM application in T1D youth (173). Given the increased DKA risks associated with HbA1c ³ 9% in patients with T1D, all efforts should be applied to understand and potentially address reasons for poor chronic glycemic control as this may prevent DKA admission. Considering DKA and HHS as potentially fatal and economically burdensome complications of diabetes, every effort for diminishing the possible risk factors is worthwhile.  

SGLT-2 inhibitor-induced DKA in patients with T2D is a potentially avoidable condition in light of accumulating knowledge of potential triggers prompting the development of this hyperglycemic emergency (174). A recent international consensus statement on the DKA risk management in patients with T1D treated with SGLT-2 inhibitors (76) can be effectively applied to the care of patients with T2D as well. Avoidance or temporary discontinuation of SGLT-2 inhibitors in clinical situations that independently increase risk of intravascular volume depletion and/or development of ketosis-prone conditions listed in the Figure 11 can mitigate the DKA risk. The DEEARAILS pneumonic can help recalling these clinical situations.  

Figure 11. Precipitating factors for DKA in patients taking SGLT2 inhibitors. LADA= latent autoimmune diabetes in adults

 

REFERENCES

 

  1. Kitabchi AE, Umpierrez GE, Miles JM, Fisher JN. Hyperglycemic crises in adult patients with diabetes. Diabetes care. 2009;32(7):1335-1343.
  2. Johnson DD, Palumbo PJ, Chu CP. Diabetic ketoacidosis in a community-based population. Mayo Clin Proc. 1980;55(2):83-88.
  3. Ramphul K, Joynauth J. An Update on the Incidence and Burden of Diabetic Ketoacidosis in the U.S. Diabetes care. 2020;43(12):e196-e197.
  4. CDC. 2014 national diabetes fact sheet (http://www.cdc.gov/diabetes/statistics/dkafirst/index.htm). accessed 04.07.15.
  5. Benoit SR, Zhang Y, Geiss LS, Gregg EW, Albright A. Trends in Diabetic Ketoacidosis Hospitalizations and In-Hospital Mortality - United States, 2000-2014. MMWR Morb Mortal Wkly Rep. 2018;67(12):362-365.
  6. Zhong VW, Juhaeri J, Mayer-Davis EJ. Trends in Hospital Admission for Diabetic Ketoacidosis in Adults With Type 1 and Type 2 Diabetes in England, 1998-2013: A Retrospective Cohort Study. Diabetes care. 2018;41(9):1870-1877.
  7. Thomas M, Harjutsalo V, Feodoroff M, Forsblom C, Gordin D, Groop PH. The Long-Term Incidence of Hospitalization for Ketoacidosis in Adults with Established T1D-A Prospective Cohort Study. J Clin Endocrinol Metab. 2020;105(1).
  8. Kitabchi AE, Umpierrez GE, Murphy MB, Barrett EJ, Kreisberg RA, Malone JI, Wall BM. Management of hyperglycemic crises in patients with diabetes. Diabetes care. 2001;24(1):131-153.
  9. Matz R. Management of the hyperosmolar hyperglycemic syndrome. Am Fam Physician. 1999;60(5):1468-1476.
  10. CDC. 2017 national diabetes statistics (https://www.cdc.gov/diabetes/pdfs/data/statistics/national-diabetes-statistics-report.pdf) accessed 05.05.18.
  11. Javor KA, Kotsanos JG, McDonald RC, Baron AD, Kesterson JG, Tierney WM. Diabetic ketoacidosis charges relative to medical charges of adult patients with type I diabetes. Diabetes care. 1997;20(3):349-354.
  12. Desai D, Mehta D, Mathias P, Menon G, Schubart UK. Health Care Utilization and Burden of Diabetic Ketoacidosis in the U.S. Over the Past Decade: A Nationwide Analysis. Diabetes care. 2018;41(8):1631-1638.
  13. Fernando SM, Bagshaw SM, Rochwerg B, McIsaac DI, Thavorn K, Forster AJ, Tran A, Reardon PM, Rosenberg E, Tanuseputro P, Kyeremanteng K. Comparison of outcomes and costs between adult diabetic ketoacidosis patients admitted to the ICU and step-down unit. Journal of critical care. 2019;50:257-261.
  14. Karslioglu French E, Donihi AC, Korytkowski MT. Diabetic ketoacidosis and hyperosmolar hyperglycemic syndrome: review of acute decompensated diabetes in adult patients. BMJ. 2019;365:l1114.
  15. Prevention. CfDCa. National Diabetes Statistics Report: Estimates of Diabetes and Its Burden in the United States, 2014. Atlanta, GA: U.S. Department of Health and Human Services; 2014. 2014.
  16. Kitabchi AE, Nyenwe EA. Hyperglycemic crises in diabetes mellitus: diabetic ketoacidosis and hyperglycemic hyperosmolar state. Endocrinol Metab Clin North Am. 2006;35(4):725-751, viii.
  17. Kitabchi AE, Umpierrez GE, Murphy MB, Kreisberg RA. Hyperglycemic crises in adult patients with diabetes: a consensus statement from the American Diabetes Association. Diabetes care. 2006;29(12):2739-2748.
  18. Pasquel FJ, Tsegka K, Wang H, Cardona S, Galindo RJ, Fayfman M, Davis G, Vellanki P, Migdal A, Gujral U, Narayan KMV, Umpierrez GE. Clinical Outcomes in Patients With Isolated or Combined Diabetic Ketoacidosis and Hyperosmolar Hyperglycemic State: A Retrospective, Hospital-Based Cohort Study. Diabetes care. 2020;43(2):349-357.
  19. Kitabchi AE, Wall BM. Diabetic ketoacidosis. Med Clin North Am. 1995;79(1):9-37.
  20. Chupin M, Charbonnel B, Chupin F. C-peptide blood levels in keto-acidosis and in hyperosmolar non-ketotic diabetic coma. Acta Diabetol Lat. 1981;18(2):123-128.
  21. Kipnis DM. Insulin secretion in diabetes mellitus. Annals of internal medicine. 1968;69(5):891-901.
  22. Kitabchi AE, Ayyagari V, Guerra SM. The efficacy of low-dose versus conventional therapy of insulin for treatment of diabetic ketoacidosis. Annals of internal medicine. 1976;84(6):633-638.
  23. Kitabchi AE, Young R, Sacks H, Morris L. Diabetic ketoacidosis: reappraisal of therapeutic approach. Annu Rev Med. 1979;30:339-357.
  24. Muller WA, Faloona GR, Unger RH. Hyperglucagonemia in diabetic ketoacidosis. Its prevalence and significance. Am J Med. 1973;54(1):52-57.
  25. Christensen NJ. Plasma norepinephrine and epinephrine in untreated diabetics, during fasting and after insulin administration. Diabetes. 1974;23(1):1-8.
  26. Alberti KG. Role of glucagon and other hormones in development of diabetic ketoacidosis. Lancet. 1975;1(7920):1307-1311.
  27. Unger RH. High Growth-Hormone Levels in Diabetic Ketoacidosis: A Possible Cause of Insulin Resistance. JAMA. 1965;191:945-947.
  28. Waldhausl W, Kleinberger G, Korn A, Dudczak R, Bratusch-Marrain P, Nowotny P. Severe hyperglycemia: effects of rehydration on endocrine derangements and blood glucose concentration. Diabetes. 1979;28(6):577-584.
  29. Foster DW, McGarry JD. The metabolic derangements and treatment of diabetic ketoacidosis. N Engl J Med. 1983;309(3):159-169.
  30. Miles JM, Rizza RA, Haymond MW, Gerich JE. Effects of acute insulin deficiency on glucose and ketone body turnover in man: evidence for the primacy of overproduction of glucose and ketone bodies in the genesis of diabetic ketoacidosis. Diabetes. 1980;29(11):926-930.
  31. Felig P, Wahren J. Influence of endogenous insulin secretion on splanchnic glucose and amino acid metabolism in man. The Journal of clinical investigation. 1971;50(8):1702-1711.
  32. Hue L. Gluconeogenesis and its regulation. Diabetes Metab Rev. 1987;3(1):111-126.
  33. Schade DS, Eaton RP. The temporal relationship between endogenously secreted stress hormones and metabolic decompensation in diabetic man. J Clin Endocrinol Metab. 1980;50(1):131-136.
  34. McGarry JD, Dobbins RL. Fatty acids, lipotoxicity and insulin secretion. Diabetologia. 1999;42(2):128-138.
  35. McGarry JD. Lilly Lecture 1978. New perspectives in the regulation of ketogenesis. Diabetes. 1979;28(5):517-523.
  36. Nurjhan N, Consoli A, Gerich J. Increased lipolysis and its consequences on gluconeogenesis in non-insulin-dependent diabetes mellitus. The Journal of clinical investigation. 1992;89(1):169-175.
  37. Balasse EO, Fery F. Ketone body production and disposal: effects of fasting, diabetes, and exercise. Diabetes Metab Rev. 1989;5(3):247-270.
  38. McGarry JD, Woeltje KF, Kuwajima M, Foster DW. Regulation of ketogenesis and the renaissance of carnitine palmitoyltransferase. Diabetes Metab Rev. 1989;5(3):271-284.
  39. Reichard GA, Jr., Skutches CL, Hoeldtke RD, Owen OE. Acetone metabolism in humans during diabetic ketoacidosis. Diabetes. 1986;35(6):668-674.
  40. Barnes AJ, Bloom SR, Goerge K, Alberti GM, Smythe P, Alford FP, Chisholm DJ. Ketoacidosis in pancreatectomized man. N Engl J Med. 1977;296(22):1250-1253.
  41. Cahill GF, Jr. Starvation in man. N Engl J Med. 1970;282(12):668-675.
  42. 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(8):2079-2086.
  43. Cherian SV, Khara L, Das S, Hamarneh WA, Garcha AS, Frechette V. Diabetic ketoacidosis complicated by generalized venous thrombosis: a case report and review. Blood Coagul Fibrinolysis. 2012;23(3):238-240.
  44. Wakabayashi S, Tsujimoto T, Kishimoto M, Ikeda N, Inoue K, Ihana N, Hamasaki H, Noto H, Yamamoto-Honda R, Kajio H, Noda M. Acute Multiple Arteriovenous Thromboses in a Patient with Diabetic Ketoacidosis. Intern Med. 2015;54(16):2025-2028.
  45. Gerich JE, Martin MM, Recant L. Clinical and metabolic characteristics of hyperosmolar nonketotic coma. Diabetes. 1971;20(4):228-238.
  46. Wachtel TJ, Silliman RA, Lamberton P. Predisposing factors for the diabetic hyperosmolar state. Arch Intern Med. 1987;147(3):499-501.
  47. Wachtel TJ, Tetu-Mouradjian LM, Goldman DL, Ellis SE, O'Sullivan PS. Hyperosmolarity and acidosis in diabetes mellitus: a three-year experience in Rhode Island. J Gen Intern Med. 1991;6(6):495-502.
  48. Fadini GP, Bonora BM, Avogaro A. SGLT2 inhibitors and diabetic ketoacidosis: data from the FDA Adverse Event Reporting System. Diabetologia. 2017;60(8):1385-1389.
  49. Bonora BM, Avogaro A, Fadini GP. Euglycemic Ketoacidosis. Curr Diab Rep. 2020;20(7):25.
  50. Ehrmann D, Kulzer B, Roos T, Haak T, Al-Khatib M, Hermanns N. Risk factors and prevention strategies for diabetic ketoacidosis in people with established type 1 diabetes. Lancet Diabetes Endocrinol. 2020;8(5):436-446.
  51. Miyoshi Y, Ogawa O, Oyama Y. Nivolumab, an Anti-Programmed Cell Death-1 Antibody, Induces Fulminant Type 1 Diabetes. Tohoku J Exp Med. 2016;239(2):155-158.
  52. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, Larkin J, Lorigan P, Neyns B, Blank CU, Hamid O, Mateus C, Shapira-Frommer R, Kosh M, Zhou H, Ibrahim N, Ebbinghaus S, Ribas A, investigators K-. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015;372(26):2521-2532.
  53. Clotman K, Janssens K, Specenier P, Weets I, De Block CEM. Programmed Cell Death-1 Inhibitor-Induced Type 1 Diabetes Mellitus. J Clin Endocrinol Metab. 2018;103(9):3144-3154.
  54. Liu J, Zhou H, Zhang Y, Fang W, Yang Y, Huang Y, Zhang L. Reporting of Immune Checkpoint Inhibitor Therapy-Associated Diabetes, 2015-2019. Diabetes care. 2020;43(7):e79-e80.
  55. Stamatouli AM, Quandt Z, Perdigoto AL, Clark PL, Kluger H, Weiss SA, Gettinger S, Sznol M, Young A, Rushakoff R, Lee J, Bluestone JA, Anderson M, Herold KC. Collateral Damage: Insulin-Dependent Diabetes Induced With Checkpoint Inhibitors. Diabetes. 2018;67(8):1471-1480.
  56. Wright JJ, Salem JE, Johnson DB, Lebrun-Vignes B, Stamatouli A, Thomas JW, Herold KC, Moslehi J, Powers AC. Increased Reporting of Immune Checkpoint Inhibitor-Associated Diabetes. Diabetes care. 2018;41(12):e150-e151.
  57. Polonsky WH, Anderson BJ, Lohrer PA, Aponte JE, Jacobson AM, Cole CF. Insulin omission in women with IDDM. Diabetes care. 1994;17(10):1178-1185.
  58. Nyenwe EA, Loganathan RS, Blum S, Ezuteh DO, Erani DM, Wan JY, Palace MR, Kitabchi AE. Active use of cocaine: an independent risk factor for recurrent diabetic ketoacidosis in a city hospital. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2007;13(1):22-29.
  59. Warner EA, Greene GS, Buchsbaum MS, Cooper DS, Robinson BE. Diabetic ketoacidosis associated with cocaine use. Arch Intern Med. 1998;158(16):1799-1802.
  60. Peden NR, Braaten JT, McKendry JB. Diabetic ketoacidosis during long-term treatment with continuous subcutaneous insulin infusion. Diabetes care. 1984;7(1):1-5.
  61. Weissberg-Benchell J, Antisdel-Lomaglio J, Seshadri R. Insulin pump therapy: a meta-analysis. Diabetes care. 2003;26(4):1079-1087.
  62. Katz JR, Edwards R, Khan M, Conway GS. Acromegaly presenting with diabetic ketoacidosis. Postgrad Med J. 1996;72(853):682-683.
  63. Szeto CC, Li KY, Ko GT, Chow CC, Yeung VT, Chan JC, Cockram CS. Acromegaly in a woman presenting with diabetic ketoacidosis and insulin resistance. Int J Clin Pract. 1997;51(7):476-477.
  64. Vidal Cortada J, Conget Donlo JI, Navarro Tellez MP, Halperin Rabinovic I, Vilardell Latorre E. [Diabetic ketoacidosis as the first manifestation of acromegaly]. An Med Interna. 1995;12(2):76-8.
  65. Umpierrez GE, Casals MM, Gebhart SP, Mixon PS, Clark WS, Phillips LS. Diabetic ketoacidosis in obese African-Americans. Diabetes. 1995;44(7):790-795.
  66. Umpierrez GE, Smiley D, Kitabchi AE. Narrative review: ketosis-prone type 2 diabetes mellitus. Annals of internal medicine. 2006;144(5):350-357.
  67. Kitabchi AE. Ketosis-prone diabetes--a new subgroup of patients with atypical type 1 and type 2 diabetes? J Clin Endocrinol Metab. 2003;88(11):5087-5089.
  68. Maldonado M, Hampe CS, Gaur LK, D'Amico S, Iyer D, Hammerle LP, Bolgiano D, Rodriguez L, Rajan A, Lernmark A, Balasubramanyam A. Ketosis-prone diabetes: dissection of a heterogeneous syndrome using an immunogenetic and beta-cell functional classification, prospective analysis, and clinical outcomes. J Clin Endocrinol Metab. 2003;88(11):5090-5098.
  69. Mauvais-Jarvis F, Sobngwi E, Porcher R, Riveline JP, Kevorkian JP, Vaisse C, Charpentier G, Guillausseau PJ, Vexiau P, Gautier JF. Ketosis-prone type 2 diabetes in patients of sub-Saharan African origin: clinical pathophysiology and natural history of beta-cell dysfunction and insulin resistance. Diabetes. 2004;53(3):645-653.
  70. Banerji MA, Chaiken RL, Huey H, Tuomi T, Norin AJ, Mackay IR, Rowley MJ, Zimmet PZ, Lebovitz HE. GAD antibody negative NIDDM in adult black subjects with diabetic ketoacidosis and increased frequency of human leukocyte antigen DR3 and DR4. Flatbush diabetes. Diabetes. 1994;43(6):741-745.
  71. Umpierrez GE, Woo W, Hagopian WA, Isaacs SD, Palmer JP, Gaur LK, Nepom GT, Clark WS, Mixon PS, Kitabchi AE. Immunogenetic analysis suggests different pathogenesis for obese and lean African-Americans with diabetic ketoacidosis. Diabetes care. 1999;22(9):1517-1523.
  72. Sobngwi E, Gautier JF, Kevorkian JP, Villette JM, Riveline JP, Zhang S, Vexiau P, Leal SM, Vaisse C, Mauvais-Jarvis F. High prevalence of glucose-6-phosphate dehydrogenase deficiency without gene mutation suggests a novel genetic mechanism predisposing to ketosis-prone diabetes. J Clin Endocrinol Metab. 2005;90(8):4446-4451.
  73. Pettus JH, Zhou FL, Shepherd L, Preblick R, Hunt PR, Paranjape S, Miller KM, Edelman SV. Incidences of Severe Hypoglycemia and Diabetic Ketoacidosis and Prevalence of Microvascular Complications Stratified by Age and Glycemic Control in U.S. Adult Patients With Type 1 Diabetes: A Real-World Study. Diabetes care. 2019;42(12):2220-2227.
  74. Li J, Wang X, Chen J, Zuo X, Zhang H, Deng A. COVID-19 infection may cause ketosis and ketoacidosis. Diabetes Obes Metab. 2020;22(10):1935-1941.
  75. Pasquel FJ, Messler J, Booth R, Kubacka B, Mumpower A, Umpierrez G, Aloi J. Characteristics of and Mortality Associated With Diabetic Ketoacidosis Among US Patients Hospitalized With or Without COVID-19. JAMA Netw Open. 2021;4(3):e211091.
  76. Danne T, Garg S, Peters AL, Buse JB, Mathieu C, Pettus JH, Alexander CM, Battelino T, Ampudia-Blasco FJ, Bode BW, Cariou B, Close KL, Dandona P, Dutta S, Ferrannini E, Fourlanos S, Grunberger G, Heller SR, Henry RR, Kurian MJ, Kushner JA, Oron T, Parkin CG, Pieber TR, Rodbard HW, Schatz D, Skyler JS, Tamborlane WV, Yokote K, Phillip M. International Consensus on Risk Management of Diabetic Ketoacidosis in Patients With Type 1 Diabetes Treated With Sodium-Glucose Cotransporter (SGLT) Inhibitors. Diabetes care. 2019;42(6):1147-1154.
  77. Ahmed M, McKenna MJ, Crowley RK. Diabetic Ketoacidosis in Patients with Type 2 Diabetes Recently Commenced on Sglt-2 Inhibitors: An Ongoing Concern. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2017;23(4):506-508.
  78. Peters AL, Buschur EO, Buse JB, Cohan P, Diner JC, Hirsch IB. Euglycemic Diabetic Ketoacidosis: A Potential Complication of Treatment With Sodium-Glucose Cotransporter 2 Inhibition. Diabetes care. 2015;38(9):1687-1693.
  79. Umpierrez G, Freire AX. Abdominal pain in patients with hyperglycemic crises. Journal of critical care. 2002;17(1):63-67.
  80. Campbell IW, Duncan LJ, Innes JA, MacCuish AC, Munro JF. Abdominal pain in diabetic metabolic decompensation. Clinical significance. JAMA. 1975;233(2):166-168.
  81. Matz R. Hypothermia in diabetic acidosis. Hormones. 1972;3(1):36-41.
  82. Alberti KG, Nattrass M. Severe diabetic ketoacidosis. Med Clin North Am. 1978;62(4):799-814.
  83. Ruderman NB, Goodman MN. Brain metabolism in diabetes. Horm Metab Res Suppl. 1980;9:1-8.
  84. Rosival V. The influence of blood hydrogen ion concentration on the level of consciousness in diabetic ketoacidosis. Ann Clin Res. 1987;19(1):23-25.
  85. Fulop M, Rosenblatt A, Kreitzer SM, Gerstenhaber B. Hyperosmolar nature of diabetic coma. Diabetes. 1975;24(6):594-599.
  86. Edge JA, Roy Y, Bergomi A, Murphy NP, Ford-Adams ME, Ong KK, Dunger DB. Conscious level in children with diabetic ketoacidosis is related to severity of acidosis and not to blood glucose concentration. Pediatr Diabetes. 2006;7(1):11-15.
  87. Morris LR, Kitabchi AE. Efficacy of low-dose insulin therapy for severely obtunded patients in diabetic ketoacidosis. Diabetes care. 1980;3(1):53-56.
  88. Nyenwe EA, Razavi LN, Kitabchi AE, Khan AN, Wan JY. Acidosis: the prime determinant of depressed sensorium in diabetic ketoacidosis. Diabetes care. 2010;33(8):1837-1839.
  89. Freire AX, Umpierrez GE, Afessa B, Latif KA, Bridges L, Kitabchi AE. Predictors of intensive care unit and hospital length of stay in diabetic ketoacidosis. Journal of critical care. 2002;17(4):207-211.
  90. Arieff AI. Cerebral edema complicating nonketotic hyperosmolar coma. Miner Electrolyte Metab. 1986;12(5-6):383-389.
  91. Guisado R, Arieff AI. Neurologic manifestations of diabetic comas: correlation with biochemical alterations in the brain. Metabolism: clinical and experimental. 1975;24(5):665-679.
  92. Harden CL, Rosenbaum DH, Daras M. Hyperglycemia presenting with occipital seizures. Epilepsia. 1991;32(2):215-220.
  93. Sheikh-Ali M, Karon BS, Basu A, Kudva YC, Muller LA, Xu J, Schwenk WF, Miles JM. Can serum beta-hydroxybutyrate be used to diagnose diabetic ketoacidosis? Diabetes care. 2008;31(4):643-647.
  94. Gosmanov AR, Gosmanova EO, Dillard-Cannon E. Management of adult diabetic ketoacidosis. Diabetes Metab Syndr Obes. 2014;7:255-264.
  95. Handelsman Y, Henry RR, Bloomgarden ZT, Dagogo-Jack S, DeFronzo RA, Einhorn D, Ferrannini E, Fonseca VA, Garber AJ, Grunberger G, LeRoith D, Umpierrez GE, Weir MR. American Association of Clinical Endocrinologists and American College of Endocrinology Position Statement on the Association of Sglt-2 Inhibitors and Diabetic Ketoacidosis. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2016;22(6):753-762.
  96. Kum-Nji JS, Gosmanov AR, Steinberg H, Dagogo-Jack S. Hyperglycemic, high anion-gap metabolic acidosis in patients receiving SGLT-2 inhibitors for diabetes management. Journal of diabetes and its complications. 2017;31(3):611-614.
  97. Umpierrez GE, Khajavi M, Kitabchi AE. Review: diabetic ketoacidosis and hyperglycemic hyperosmolar nonketotic syndrome. Am J Med Sci. 1996;311(5):225-233.
  98. Robin AP, Ing TS, Lancaster GA, Soung LS, Sparagana M, Geis WP, Hano JE. Hyperglycemia-induced hyponatremia: a fresh look. Clin Chem. 1979;25(3):496-497.
  99. Spasovski G, Vanholder R, Allolio B, Annane D, Ball S, Bichet D, Decaux G, Fenske W, Hoorn EJ, Ichai C, Joannidis M, Soupart A, Zietse R, Haller M, van der Veer S, Van Biesen W, Nagler E, Hyponatraemia Guideline Development G. Clinical practice guideline on diagnosis and treatment of hyponatraemia. Nephrol Dial Transplant. 2014;29 Suppl 2:i1-i39.
  100. Kaminska ES, Pourmotabbed G. Spurious laboratory values in diabetic ketoacidosis and hyperlipidemia. The American journal of emergency medicine. 1993;11(1):77-80.
  101. Adrogue HJ, Lederer ED, Suki WN, Eknoyan G. Determinants of plasma potassium levels in diabetic ketoacidosis. Medicine (Baltimore). 1986;65(3):163-172.
  102. Wilson HK, Keuer SP, Lea AS, Boyd AE, 3rd, Eknoyan G. Phosphate therapy in diabetic ketoacidosis. Arch Intern Med. 1982;142(3):517-520.
  103. Slovis CM, Mork VG, Slovis RJ, Bain RP. Diabetic ketoacidosis and infection: leukocyte count and differential as early predictors of serious infection. The American journal of emergency medicine. 1987;5(1):1-5.
  104. 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(4):443-448.
  105. Bewsher PD, Petrie JC, Worth HG. Serum lipid levels in hyperosmolar non-ketotic diabetic coma. Br Med J. 1970;3(5714):82-84.
  106. Vinicor F, Lehrner LM, Karn RC, Merritt AD. Hyperamylasemia in diabetic ketoacidosis: sources and significance. Annals of internal medicine. 1979;91(2):200-204.
  107. Yadav D, Nair S, Norkus EP, Pitchumoni CS. Nonspecific hyperamylasemia and hyperlipasemia in diabetic ketoacidosis: incidence and correlation with biochemical abnormalities. Am J Gastroenterol. 2000;95(11):3123-3128.
  108. Gerard SK, Khayam-Bashi H. Characterization of creatinine error in ketotic patients. A prospective comparison of alkaline picrate methods with an enzymatic method. Am J Clin Pathol. 1985;84(5):659-664.
  109. Stephens JM, Sulway MJ, Watkins PJ. Relationship of blood acetoacetate and 3-hydroxybutyrate in diabetes. Diabetes. 1971;20(7):485-489.
  110. Fulop M, Murthy V, Michilli A, Nalamati J, Qian Q, Saitowitz A. Serum beta-hydroxybutyrate measurement in patients with uncontrolled diabetes mellitus. Arch Intern Med. 1999;159(4):381-384.
  111. Porter WH, Yao HH, Karounos DG. Laboratory and clinical evaluation of assays for beta-hydroxybutyrate. Am J Clin Pathol. 1997;107(3):353-358.
  112. Csako G, Elin RJ. Unrecognized false-positive ketones from drugs containing free-sulfhydryl group(s). JAMA. 1993;269(13):1634.
  113. Albert MS, Dell RB, Winters RW. Quantitative displacement of acid-base equilibrium in metabolic acidosis. Annals of internal medicine. 1967;66(2):312-322.
  114. Halperin ML, Hammeke M, Josse RG, Jungas RL. Metabolic acidosis in the alcoholic: a pathophysiologic approach. Metabolism: clinical and experimental. 1983;32(3):308-315.
  115. Bjellerup P, Kallner A, Kollind M. GLC determination of serum-ethylene glycol, interferences in ketotic patients. J Toxicol Clin Toxicol. 1994;32(1):85-87.
  116. Paulson WD, Gadallah MF. Diagnosis of mixed acid-base disorders in diabetic ketoacidosis. Am J Med Sci. 1993;306(5):295-300.
  117. Freidenberg GR, Kosnik EJ, Sotos JF. Hyperglycemic coma after suprasellar surgery. N Engl J Med. 1980;303(15):863-865.
  118. Hillman K. Fluid resuscitation in diabetic emergencies--a reappraisal. Intensive Care Med. 1987;13(1):4-8.
  119. Diabetes Canada Clinical Practice Guidelines Expert C, Goguen J, Gilbert J. Hyperglycemic Emergencies in Adults. Can J Diabetes. 2018;42 Suppl 1:S109-S114.
  120. Rosenbloom AL. Intracerebral crises during treatment of diabetic ketoacidosis. Diabetes care. 1990;13(1):22-33.
  121. Pasquel FJ, Umpierrez GE. Hyperosmolar hyperglycemic state: a historic review of the clinical presentation, diagnosis, and treatment. Diabetes care. 2014;37(11):3124-3131.
  122. Cardoso L, Vicente N, Rodrigues D, Gomes L, Carrilho F. Controversies in the management of hyperglycaemic emergencies in adults with diabetes. Metabolism: clinical and experimental. 2017;68:43-54.
  123. Fisher JN, Shahshahani MN, Kitabchi AE. Diabetic ketoacidosis: low-dose insulin therapy by various routes. N Engl J Med. 1977;297(5):238-241.
  124. Sacks HS, Shahshahani M, Kitabchi AE, Fisher JN, Young RT. Similar responsiveness of diabetic ketoacidosis to low-dose insulin by intramuscular injection and albumin-free infusion. Annals of internal medicine. 1979;90(1):36-42.
  125. Burghen GA, Etteldorf JN, Fisher JN, Kitabchi AQ. Comparison of high-dose and low-dose insulin by continuous intravenous infusion in the treatment of diabetic ketoacidosis in children. Diabetes care. 1980;3(1):15-20.
  126. Umpierrez GE, Cuervo R, Karabell A, Latif K, Freire AX, Kitabchi AE. Treatment of diabetic ketoacidosis with subcutaneous insulin aspart. Diabetes care. 2004;27(8):1873-1878.
  127. Umpierrez GE, Latif K, Stoever J, Cuervo R, Park L, Freire AX, A EK. Efficacy of subcutaneous insulin lispro versus continuous intravenous regular insulin for the treatment of patients with diabetic ketoacidosis. Am J Med. 2004;117(5):291-296.
  128. Della Manna T, Steinmetz L, Campos PR, Farhat SC, Schvartsman C, Kuperman H, Setian N, Damiani D. Subcutaneous use of a fast-acting insulin analog: an alternative treatment for pediatric patients with diabetic ketoacidosis. Diabetes care. 2005;28(8):1856-1861.
  129. Atchley DW, Loeb RF, Richards DW, Benedict EM, Driscoll ME. ON DIABETIC ACIDOSIS: A Detailed Study of Electrolyte Balances Following the Withdrawal and Reestablishment of Insulin Therapy. The Journal of clinical investigation. 1933;12(2):297-326.
  130. Beigelman PM. Potassium in severe diabetic ketoacidosis. Am J Med. 1973;54(4):419-420.
  131. Abramson E, Arky R. Diabetic acidosis with initial hypokalemia. Therapeutic implications. JAMA. 1966;196(5):401-403.
  132. Viallon A, Zeni F, Lafond P, Venet C, Tardy B, Page Y, Bertrand JC. Does bicarbonate therapy improve the management of severe diabetic ketoacidosis? Crit Care Med. 1999;27(12):2690-2693.
  133. Glaser NS, Wootton-Gorges SL, Marcin JP, Buonocore MH, Dicarlo J, Neely EK, Barnes P, Bottomly J, Kuppermann N. Mechanism of cerebral edema in children with diabetic ketoacidosis. J Pediatr. 2004;145(2):164-171.
  134. Krane EJ, Rockoff MA, Wallman JK, Wolfsdorf JI. Subclinical brain swelling in children during treatment of diabetic ketoacidosis. N Engl J Med. 1985;312(18):1147-1151.
  135. Okuda Y, Adrogue HJ, Field JB, Nohara H, Yamashita K. Counterproductive effects of sodium bicarbonate in diabetic ketoacidosis. J Clin Endocrinol Metab. 1996;81(1):314-320.
  136. Morris LR, Murphy MB, Kitabchi AE. Bicarbonate therapy in severe diabetic ketoacidosis. Annals of internal medicine. 1986;105(6):836-840.
  137. Green SM, Rothrock SG, Ho JD, Gallant RD, Borger R, Thomas TL, Zimmerman GJ. Failure of adjunctive bicarbonate to improve outcome in severe pediatric diabetic ketoacidosis. Ann Emerg Med. 1998;31(1):41-48.
  138. Lever E, Jaspan JB. Sodium bicarbonate therapy in severe diabetic ketoacidosis. Am J Med. 1983;75(2):263-268.
  139. Fisher JN, Kitabchi AE. A randomized study of phosphate therapy in the treatment of diabetic ketoacidosis. J Clin Endocrinol Metab. 1983;57(1):177-180.
  140. Keller U, Berger W. Prevention of hypophosphatemia by phosphate infusion during treatment of diabetic ketoacidosis and hyperosmolar coma. Diabetes. 1980;29(2):87-95.
  141. Kreisberg RA. Phosphorus deficiency and hypophosphatemia. Hosp Pract. 1977;12(3):121-128.
  142. Winter RJ, Harris CJ, Phillips LS, Green OC. Diabetic ketoacidosis. Induction of hypocalcemia and hypomagnesemia by phosphate therapy. Am J Med. 1979;67(5):897-900.
  143. Adrogue HJ, Wilson H, Boyd AE, 3rd, Suki WN, Eknoyan G. Plasma acid-base patterns in diabetic ketoacidosis. N Engl J Med. 1982;307(26):1603-1610.
  144. Kelly AM. The case for venous rather than arterial blood gases in diabetic ketoacidosis. Emerg Med Australas. 2006;18(1):64-67.
  145. Adrogue HJ, Eknoyan G, Suki WK. Diabetic ketoacidosis: role of the kidney in the acid-base homeostasis re-evaluated. Kidney Int. 1984;25(4):591-598.
  146. Oster JR, Epstein M. Acid-base aspects of ketoacidosis. Am J Nephrol. 1984;4(3):137-151.
  147. Kamel KS, Halperin ML. Acid-base problems in diabetic ketoacidosis. N Engl J Med. 2015;372(6):546-554.
  148. Wallace TM, Matthews DR. Recent advances in the monitoring and management of diabetic ketoacidosis. QJM. 2004;97(12):773-780.
  149. Fleckman AM. Diabetic ketoacidosis. Endocrinol Metab Clin North Am. 1993;22(2):181-207.
  150. Oh MS, Carroll HJ, Goldstein DA, Fein IA. Hyperchloremic acidosis during the recovery phase of diabetic ketosis. Annals of internal medicine. 1978;89(6):925-927.
  151. Duck SC, Wyatt DT. Factors associated with brain herniation in the treatment of diabetic ketoacidosis. J Pediatr. 1988;113(1 Pt 1):10-14.
  152. Silver SM, Clark EC, Schroeder BM, Sterns RH. Pathogenesis of cerebral edema after treatment of diabetic ketoacidosis. Kidney Int. 1997;51(4):1237-1244.
  153. Haringhuizen A, Tjan DH, Grool A, van Vugt R, van Zante AR. Fatal cerebral oedema in adult diabetic ketoacidosis. Neth J Med. 2010;68(1):35-37.
  154. Hoorn EJ, Carlotti AP, Costa LA, MacMahon B, Bohn G, Zietse R, Halperin ML, Bohn D. Preventing a drop in effective plasma osmolality to minimize the likelihood of cerebral edema during treatment of children with diabetic ketoacidosis. J Pediatr. 2007;150(5):467-473.
  155. Smedman L, Escobar R, Hesser U, Persson B. Sub-clinical cerebral oedema does not occur regularly during treatment for diabetic ketoacidosis. Acta Paediatr. 1997;86(11):1172-1176.
  156. Isales CM, Min L, Hoffman WH. Acetoacetate and beta-hydroxybutyrate differentially regulate endothelin-1 and vascular endothelial growth factor in mouse brain microvascular endothelial cells. Journal of diabetes and its complications. 1999;13(2):91-97.
  157. Edge JA. Cerebral oedema during treatment of diabetic ketoacidosis: are we any nearer finding a cause? Diabetes Metab Res Rev. 2000;16(5):316-324.
  158. Galindo RJ, Pasquel FJ, Fayfman M, Tsegka K, Dhruv N, Cardona S, Wang H, Vellanki P, Umpierrez GE. Clinical characteristics and outcomes of patients with end-stage renal disease hospitalized with diabetes ketoacidosis. BMJ Open Diabetes Res Care. 2020;8(1).
  159. Buyukasik Y, Ileri NS, Haznedaroglu IC, Karaahmetoglu S, Muftuoglu O, Kirazli S, Dundar S. Enhanced subclinical coagulation activation during diabetic ketoacidosis. Diabetes care. 1998;21(5):868-870.
  160. McLaren EH, Cullen DR, Brown MJ. Coagulation abnormalities in diabetic coma before and 24 hours after treatment. Diabetologia. 1979;17(6):345-349.
  161. Timperley WR, Preston FE, Ward JD. Cerebral intravascular coagulation in diabetic ketoacidosis. Lancet. 1974;1(7864):952-956.
  162. Everett E, Mathioudakis NN. Association of socioeconomic status and DKA readmission in adults with type 1 diabetes: analysis of the US National Readmission Database. BMJ Open Diabetes Res Care. 2019;7(1):e000621.
  163. Maldonado MR, Chong ER, Oehl MA, Balasubramanyam A. Economic impact of diabetic ketoacidosis in a multiethnic indigent population: analysis of costs based on the precipitating cause. Diabetes care. 2003;26(4):1265-1269.
  164. Musey VC, Lee JK, Crawford R, Klatka MA, McAdams D, Phillips LS. Diabetes in urban African-Americans. I. Cessation of insulin therapy is the major precipitating cause of diabetic ketoacidosis. Diabetes care. 1995;18(4):483-489.
  165. Randall L, Begovic J, Hudson M, Smiley D, Peng L, Pitre N, Umpierrez D, Umpierrez G. Recurrent diabetic ketoacidosis in inner-city minority patients: behavioral, socioeconomic, and psychosocial factors. Diabetes care. 2011;34(9):1891-1896.
  166. Lindner LME, Rathmann W, Rosenbauer J. Inequalities in glycaemic control, hypoglycaemia and diabetic ketoacidosis according to socio-economic status and area-level deprivation in Type 1 diabetes mellitus: a systematic review. Diabet Med. 2018;35(1):12-32.
  167. Laffel LM, Brackett J, Ho J, Anderson BJ. Changing the process of diabetes care improves metabolic outcomes and reduces hospitalizations. Qual Manag Health Care. 1998;6(4):53-62.
  168. Rewers A, Chase HP, Mackenzie T, Walravens P, Roback M, Rewers M, Hamman RF, Klingensmith G. Predictors of acute complications in children with type 1 diabetes. JAMA. 2002;287(19):2511-2518.
  169. Vanelli M, Chiari G, Ghizzoni L, Costi G, Giacalone T, Chiarelli F. Effectiveness of a prevention program for diabetic ketoacidosis in children. An 8-year study in schools and private practices. Diabetes care. 1999;22(1):7-9.
  170. Runyan JW, Jr., Zwaag RV, Joyner MB, Miller ST. The Memphis diabetes continuing care program. Diabetes care. 1980;3(2):382-386.
  171. Charleer S, Mathieu C, Nobels F, De Block C, Radermecker RP, Hermans MP, Taes Y, Vercammen C, T'Sjoen G, Crenier L, Fieuws S, Keymeulen B, Gillard P, Investigators RT. Effect of Continuous Glucose Monitoring on Glycemic Control, Acute Admissions, and Quality of Life: A Real-World Study. J Clin Endocrinol Metab. 2018;103(3):1224-1232.
  172. Parkin CG, Graham C, Smolskis J. Continuous Glucose Monitoring Use in Type 1 Diabetes: Longitudinal Analysis Demonstrates Meaningful Improvements in HbA1c and Reductions in Health Care Utilization. J Diabetes Sci Technol. 2017;11(3):522-528.
  173. Cardona-Hernandez R, Schwandt A, Alkandari H, Bratke H, Chobot A, Coles N, Corathers S, Goksen D, Goss P, Imane Z, Nagl K, O'Riordan SMP, Jefferies C, Group SS. Glycemic Outcome Associated With Insulin Pump and Glucose Sensor Use in Children and Adolescents With Type 1 Diabetes. Data From the International Pediatric Registry SWEET. Diabetes care. 2021.
  174. Fleming N, Hamblin PS, Story D, Ekinci EI. Evolving Evidence of Diabetic Ketoacidosis in Patients Taking Sodium-Glucose Cotransporter 2 Inhibitors. J Clin Endocrinol Metab. 2020;105(8).

 

Primary Hyperparathyroidism

ABSTRACT

Primary hyperparathyroidism (PHPT) is characterized by hypercalcemia and elevated or inappropriately normal parathyroid hormone (PTH) levels. PHPT results from excessive secretion of PTH from one or more of the parathyroid glands. The clinical presentation of PHPT has evolved since the 1970’s with the advent of the routine measurement of serum calcium at that time. Classical PHPT, with its associated severe hypercalcemia, osteitis fibrosa cystica, nephrolithiasis, and neuropsychological symptoms, once common is now infrequent. Today most patients are asymptomatic and have mild hypercalcemia, but may have evidence of subclinical skeletal and renal sequelae such as osteoporosis and hypercalciuria as well as vertebral fractures and nephrolithiasis both of which may be asymptomatic. Parathyroidectomy is the only curative treatment for PHPT and is recommended in patients with symptoms and those with asymptomatic disease who have evidence of end-organ sequelae. Parathyroidectomy results in an increase in BMD and a reduction in nephrolithiasis.

 

INTRODUCTION

Primary hyperparathyroidism (PHPT) is characterized by hypercalcemia and elevated or inappropriately normal parathyroid hormone (PTH) levels. The disorder today bears few similarities to the severe condition described by Fuller Albright and others as a “disease of stones, bones, and groans” in the 1930s (1-3).  The skeletal hallmark of PHPT was osteitis fibrosa cystica, radiographically characterized by brown tumors of the long bones, subperiosteal bone resorption, distal tapering of the clavicles and phalanges, and “salt-and-pepper” erosions of the skull (4). Nephrocalcinosis and nephrolithiasis were present in the majority of patients, and neuromuscular dysfunction with muscle weakness was also common. With the advent of the automated serum chemistry autoanalyzer in the 1970s, the diagnosis of PHPT was increasingly recognized, leading to a four- to five-fold increase in incidence (5-7). Classic symptomatology, concomitantly, became much less frequent. In the United States and elsewhere in the developed world, symptomatic PHPT is now the exception and more than three fourths of patients having no symptoms attributable to their disease, making PHPT a disease that has “evolved” from its classic presentation (Table 1) (8). Symptomatic nephrolithiasis is still observed, although much less frequently than in the past. Now, radiologically evident bone disease is rare, but subclinical skeletal involvement can be readily detected by bone densitometry (9). This chapter describes the modern presentation, diagnosis and management of PHPT.

 

Table 1. Changing Clinical Profile of Primary Hyperparathyroidism

 

Cope (1930-1965)

Heath et al (1965-1974)

Mallette et al (1965-1972)

Silverberg et al (1984-2009)

Nephrolithiasis (%)

57

51

37

17

Skeletal disease (%)

23

10

14

1.4

Hypercalciuria (%)

NR

36

40

39

Asymptomatic (%)

0.6

18

22

80

NR= not reported

 

RISK FACTORS, PATHOLOGY, AND ANATOMICAL LOCATION         

PHPT results from excessive secretion of PTH from one or more of the parathyroid glands. The underlying cause of sporadic PHPT is unknown in most cases. While external neck radiation and lithium therapy are risk factors for the development of sporadic PHPT, most patients do not report these exposures (10-12). Chronically low calcium intake and higher body weight have also been recently described to be risk factors (13,14). The genetic pathogenesis of sporadic PHPT is unclear in most patients but genes regulating the cell cycle are thought to be important given the clonal nature of sporadic parathyroid adenomas.

 

By far the most common pathological finding in patients with PHPT is a solitary parathyroid adenoma, occurring in 80% of patients (15). In 2-4% of patients, PHPT is due to multiple adenomas (16). In approximately 15% of patients, all four parathyroid glands are involved (15,17). Parathyroid carcinoma accounts for <1% of all cases of PHPT(18). The etiology of four-gland parathyroid hyperplasia is multifactorial. There are no clinical features that definitively differentiate single versus multiglandular disease, but risk factors include inherited genetic syndromes such as multiple endocrine neoplasia (MEN) type 1 or type 2a and lithium exposure (17).

 

Parathyroid adenomas can be found in many unexpected anatomic locations. Parathyroid tissue embryonal migration patterns account for a plethora of possible sites of ectopic parathyroid adenomas. The most common atypical locations are within the thyroid gland, the superior mediastinum, and within the thymus (19). Occasionally, adenomas are identified in the retroesophageal space, the pharynx, the lateral neck, and even in the alimentary submucosa of the esophagus (20-22). On histologic examination, most parathyroid adenomas are encapsulated and are composed of parathyroid chief cells. Adenomas containing mainly oxyphilic or oncocytic cells are rare, but can give rise to clinical PHPT (23). Very rarely, PHPT may be due to parathyromatosis. This refers to an uncommon condition in which benign hyperfunctioning parathyroid tissue is scattered throughout the neck and/or in the superior mediastinum (see Unusual Presentations) (24).

 

EPIDEMIOLOGY

The incidence of PHPT has changed dramatically over the last half century (5,6,25,26). Before the advent of the multichannel autoanalyzer in the early 1970s, Heath et al reported an incidence of 7.8 cases per 100,000 persons in Rochester, Minnesota (5). With the introduction of routine calcium measurements in the mid-1970s, this rate rose dramatically to 51.1 cases per 100,000 in the same community. After prevalent cases were diagnosed, the incidence declined to approximately 27 per 100,000 persons per year in the United States until 1998, at which time another sharp increase was noted (25,27,28). This second peak has been attributed to the introduction of osteoporosis screening guidelines and targeted testing in those with osteoporosis (28). Recent works shows the incidence of PHPT increases with age and is higher in women and African-Americans than in men and other racial groups, respectively (29).

 

Greater appreciation of the catabolic potential of PTH in postmenopausal women with osteoporosis has led to measurement of PTH even in subjects who do not have hypercalcemia. This trend has led to the emergence of a new entity, normocalcemic PHPT or NPHPT(30). This condition is characterized by normal serum calcium, elevated PTH, and exclusion of known causes of secondary hyperparathyroidism. The incidence of NPHPT is unknown, but recent studies suggest a prevalence ranging from 0.2-3.1% (31,32).

 

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS

The diagnosis of PHPT is made when hypercalcemia and elevated PTH levels are present.

PTH levels that are inappropriately normal are also consistent with the diagnosis. The other major cause of hypercalcemia, malignancy, is readily discriminated from PHPT by a suppressed PTH level. Further, both the clinical presentation and biochemical profile of PHPT and hypercalcemia of malignancy help distinguish them. Patients with hypercalcemia of malignancy typically have severe and symptomatic hypercalcemia and advanced cancers that are clinically obvious. On the other hand, in PHPT most patients are asymptomatic and the serum calcium level is typically mildly elevated (within 1 mg/dl of the upper limit of normal). Extremely rarely, a patient with malignancy will be shown to have elevated PTH levels resulting from ectopic secretion of native PTH from the tumor itself (33). Much more commonly, the malignancy is associated with the secretion of parathyroid hormone–related protein (PTHrP), a molecule that does not cross-react in intact PTH assay (discussed below). Finally, it is possible that a malignancy is present in association with PHPT. When the PTH level is elevated in someone with a malignancy, this is more likely to be the case than a true ectopic PTH syndrome.

 

While ninety percent of patients with hypercalcemia have either PHPT or malignancy, the differential diagnosis of hypercalcemia includes a number of other etiologies such as vitamin D intoxication, granulomatous disease, and others (33).  With the exception of lithium and thiazide use and familial hypocalciuric hypercalcemia (FHH), virtually all other causes of hypercalcemia are associated with suppressed levels of PTH. If lithium and/or thiazides can be safely withdrawn, serum calcium and PTH levels that continue to be elevated 3-6 months later, confirm the diagnosis of PHPT. FHH, on the other hand, is differentiated from PHPT by family history, typically (but not always) low urinary calcium excretion, and mutations in the calcium sensing receptor (CASR) or more recently associated GNA11 and AP2S1 genes (34-36). In addition, virtual complete genetic penetrance leads to its clinical appearance typically before the age of 30. It is extremely unusual for FHH to present without an antecedent history after the age of 50.

 

To distinguish PTH-mediated from non-PTH mediated causes of hypercalcemia, PTH should be measured with an intact immunoradiometric (IRMA) or immunochemiluminometric (ICMA) assay, which readily discriminates between PHPT and hypercalcemia of malignancy. In PHPT, PTH concentrations are usually frankly elevated, but most often within 2 times the upper limit of normal. A minority may have PTH levels in the normal range, typically in the upper range of normal. In PHPT, such values, although within the normal range, are clearly abnormal in a hypercalcemic setting. Several factors affect the PTH level in those with and without PHPT, including age, vitamin D levels, and renal function. Because PTH levels normally rise with age, the broad normal range (typically 10-65 pg/mL) reflects values for the entire population. In the younger individual (< 45 years), one expects a narrower and lower normal range (10-45 pg/mL). Occasionally, the PTH level as measured will be as low as 20-30 pg/mL. Such unusual examples require a more careful consideration of other causes of hypercalcemia, but such individuals will usually be shown to have PHPT because hypercalcemia that is not PTH-mediated suppresses the PTH concentration to levels that are either undetectable or at the lower limits of the reference range. Souberbielle et al (37) have illustrated that the normal range is dependent on whether or not the reference population is or is not vitamin D deficient. When vitamin D–deficient individuals were excluded, the upper limit of the PTH reference interval decreased. Patients with PHPT and vitamin D deficiency have a “heightened” PTH levels compared to those who are vitamin D sufficient (38).

 

On the other hand, renal dysfunction tends to elevate PTH levels via a number of mechanisms, including reduced clearance and degradation of PTH. Indeed, patients with PHPT and severe renal dysfunction (glomerular filtration rate < 30ml/min), may also have higher PTH levels compared to those with better renal function (39). In addition, the “intact” IRMA for PTH overestimates the concentration of biologically active PTH, particularly in renal failure. In 1998, Lepage et al (40) demonstrated a large non-(1–84) PTH fragment that comigrated with a large aminoterminally truncated fragment (PTH[7–84]) and showed substantial cross-reactivity in commercially available IRMAs. This large, inactive moiety constituted as much as 50% of immunoreactivity by IRMA for PTH in individuals with chronic renal failure (41). Recognition of this molecule led to the development of a new IRMA using affinity-purified polyclonal antibodies to PTH (39–84) and to the extreme N-terminal amino acid regions, PTH (1–4) (42,43). This “whole PTH” or third generation assay detects only the full-length PTH molecule, PTH (1–84). This assay has clear utility in uremic patients, but in PHPT, both assays are equally useful (40,44-46). Using the third-generation assay for PTH (1-84), a second molecular form of PTH(1-84) that is immunologically intact at both extremes has been identified. This molecule reacts only poorly in second-generation PTH assays. It represents less than 10% of the immunoreactivity in normal individuals and up to 15% in renal failure patients. In a limited number of patients with a severe form of PHPT or with parathyroid cancer, it may be over-expressed (47).

 

PHPT can be discriminated from secondary and tertiary hyperparathyroidism by its different biochemical profile. Secondary hyperparathyroidism is associated with an appropriate elevation in PTH in response to a hypocalcemic provocation and either a frankly low or normal serum calcium level. Secondary hyperparathyroidism is often due to vitamin D deficiency. Other causes include malabsorption, kidney disease, or hypercalciuria. Infrequently, patients with secondary hyperparathyroidism may become hypercalcemic, and will ultimately be found to have PHPT, when the underlying condition (for example, vitamin D deficiency) is corrected (48). In these cases, the hypercalcemia of PHPT was ‘masked’ by the co-existing condition. On the other hand, tertiary hyperparathyroidism describes a condition in which prolonged, severe secondary hyperparathyroidism (as in end-stage renal disease) evolves into a hypercalcemic state due to the development of autonomous functioning of one or more of the hyperplastic parathyroid glands. This can be observed in patients on dialysis or after renal transplant. Tertiary hyperparathyroidism is usually obvious from the history.

 

Normocalcemic primary hyperparathyroidism (NPHPT) describes a condition characterized by normal serum albumin-corrected calcium levels and ionized calcium values with an elevated PTH level. This condition can only be diagnosed when all known causes of secondary hyperparathyroidism have been excluded. Patients with NPHPT typically are diagnosed when PTH is measured in the course of an evaluation for low bone mass. NPHPT may represent the earliest manifestations of PHPT, a “forme fruste” of the disease. Several reports have appeared describing these individuals, with some patients progressing to overt hypercalcemia while under observation (30,32,49,50).

 

Although the term “normocalcemic PHPT” has been in use for decades, there has been considerable controversy concerning the accuracy of this designation. In many cases, the increases in PTH levels were attributable to the limitations of available assay technology. The older midmolecule radioimmunoassay for PTH, previously in common use, measured hormone fragments in addition to the intact molecule. Spuriously elevated PTH levels, particularly in those with renal insufficiency in whom clearance of hormone fragments is impaired, were seen. Alternative explanations for hyperparathyroidism with NPHPT have been discovered, including medications, hypercalciuria, renal insufficiency, and certain forms of liver and gastrointestinal disease. In recent years, it has become clear that many patients designated as having NPHPT were vitamin D deficient. Vitamin D deficiency with coexisting PHPT can give the semblance of normal calcium levels when in fact they would have been hypercalcemic if the vitamin D levels were normal. Since a possible view of NPHPT is a condition fostered by an element of vitamin D resistance, it is important to ensure vitamin D sufficiency. While the Institute of Medicine states that normal levels of vitamin D, as measured by 25-hydroxyvitamin D, are 20 ng/ml, it did not address conditions of abnormal mineral metabolism, such as PHPT. In particular, in NPHPT, we and others recommend that levels of 25-hydorxyvitamin D be raised, if necessary, to > 30 ng/mL for at least 3 months in order to rule out an element of vitamin D insufficiency in this population. Biochemical profiles for the various causes of hypercalcemia and hyperparathyroidism are shown in Table 2.

 

Table 2. Biochemical Profiles for Various Causes of Hypercalcemia and Hyperparathyroidism

Cause

Serum Calcium

        PTH

Urine Calcium

Primary Hyperparathyroidism

Elevated

Elevated or Inappropriately Normal

High or High Normal

Hypercalcemia of Malignancy and non-PTH Mediated Hypercalcemia

Elevated

Suppressed

Typically High*

Secondary Hyperparathyroidism

Normal or Low

Elevated

Low in vitamin D deficiency, malabsorption, chronic renal failure,

High in Idiopathic Hypercalciuria

Tertiary Hyperparathyroidism

Elevated

Elevated

Low before transplant

FHH

Elevated

Typically High Normal or Elevated

Typically Low

Normocalcemic Primary Hyperparathyroidism

Normal

High

<350 mg/24 hours (30)

*may vary by cause

 

OTHER BIOCHEMICAL FEATURES

In PHPT, serum phosphorus tends to be in the lower range of normal, but frank hypophosphatemia is present in less than one fourth of patients. Hypophosphatemia, when present, is due to the phosphaturic actions of PTH. Average total urinary calcium excretion is at the upper end of the normal range, with about 40% of all patients experiencing hypercalciuria. Serum 25-hydroxyvitamin D levels tend to be in the lower end of the normal range. Although mean values of 1,25-dihydroxyvitamin D are in the high-normal range, approximately one third of patients have frankly elevated levels of 1,25-dihydroxyvitamin D (51). This pattern is a result of the actions of PTH to increase expression of the 1-alpha hydroxylase that converts 25-hydroxyvitamin D to 1,25-dihydroxyvitamin D.  A typical biochemical profile is shown in Table 3.

 

Table 3. Biochemical Profile in Primary Hyperparathyroidism (n = 137)

 

Patients (Mean ± SEM)

Normal Range

Serum calcium

10.7 ± 0.1 mg/dL

8.2-10.2 mg/dL

Serum phosphorus

2.8 ± 0.1 mg/dL

2.5-1.5 mg/dL

Total alkaline phosphatase

114 ± 5 IU/L

<100 IU/L

Serum magnesium

2.0 ± 0.1 mg/dL

1.8-2.4 mg/dL

PTH (IRMA)

119 ± 7 pg/mL

10-65 pg/mL

25(OH)D

19 ± 1 ng/mL

30-80 ng/mL

1,25(OH)2D

54 ± 2 pg/mL

15-60 pg/mL

Urinary calcium

240 ± 11 mg/g creatinine

 

Urine DPD

17.6 ± 1.3 nmol/mmol creatinine

<14.6 nmol/mmol creatinine

Urine PYD

46.8 ± 2.7 nmol/mmol creatinine

<51.8 nmol/mmol creatinine

DPD, Deoxypyridinoline; PTH (IRMA), parathyroid hormone (immunoradiometric assay); PYD, pyridinoline.

 

CLINICAL PRESENTATION

PHPT typically occurs in individuals in their middle years, with a peak incidence between ages 50 and 60 years. However, the condition can occur at any age. Women are affected more frequently than men, in a ratio of approximately 3-4:1. Several different presentations of PHPT are possible and were originally described successively in time (2,30,52). The classical symptomatic presentation was described first; later, asymptomatic PHPT emerged due to biochemical screening, and most recently the normocalcemic variant was discovered as described above. However, these three forms of PHPT contemporaneously exist today. Which presentation predominates depends upon population- and geographic-specific screening practices. It is also postulated that vitamin D deficiency may affect clinical presentation. Vitamin D deficiency heightens PTH elevations and this can worsen the hyperparathyroid process (53). In regions of the world and populations where biochemical screening is not routine and incidentally where vitamin D deficiency is endemic, symptomatic PHPT is the most common form and PHPT will appear to be uncommon because it is only discovered when symptomatic (54-64).  In areas and populations where screening is routine, asymptomatic PHPT will predominate and the incidence of PHPT is higher (52,65).

 

This chapter focuses on asymptomatic PHPT, as it is the predominant form in the United States and in most of the developed world. At the time of diagnosis, most patients with PHPT do not exhibit classic symptoms or signs associated with disease. Clinically overt kidney stones and fractures are rare (66). Constitutional complaints such as weakness, easy fatigability, depression, and intellectual weariness are seen with some regularity (see later discussion) (67). The physical examination is generally unremarkable. Band keratopathy, a hallmark of classic PHPT, occurs because of deposition of calcium phosphate crystals in the cornea, but is virtually never seen grossly. Even by slit-lamp examination, this finding is rare. The neck shows no masses. The neuromuscular system is normal. The sections below provide a detailed description of the multi-systemic manifestations of PHPT.

 

Diseases associated epidemiologically with PHPT have included hypertension (68-70), peptic ulcer disease, gout, or pseudogout (71,72). More recently celiac disease has been associated with PHPT (73). Some concomitant disorders such as hypertension are commonly seen, but it is not established that any of these associated disorders are etiologically linked to the disease.

 

The Skeleton 

The classic radiologic bone disease of PHPT, osteitis fibrosa cystica, is rarely seen today in the United States. Most series place the incidence of osteitis fibrosa cystica at less than 2% of patients with PHPT. The absence of classic radiographic features (salt-and-pepper skull, tapering of the distal third of the clavicle, subperiosteal bone resorption of the phalanges, brown tumors) does not mean that the skeleton is not affected. With more sensitive techniques, it has become clear that skeletal involvement in the hyperparathyroid process is actually quite common. This section reviews the profile of the skeleton in PHPT as it is reflected in assays for bone markers, bone densitometry, bone histomorphometry, and new skeletal imaging techniques.

 

BONE TURNOVER MARKERS

PTH stimulates both bone resorption and bone formation. Markers of bone turnover, which reflect those dynamics, provide clues to the extent of skeletal involvement in PHPT (74).

 

Bone Formation Markers

Osteoblast products, including bone-specific alkaline phosphatase activity, osteocalcin, and serum amino-terminal propeptide of type I collagen (P1NP), reflect bone formation (74). In PHPT, alkaline phosphatase levels, the most widely clinically available marker, can be mildly elevated, but in many patients, total alkaline phosphatase values are within normal limits (75,76). In a small study from our group (77), bone-specific alkaline phosphatase activity correlated with PTH levels and BMD at the lumbar spine and femoral neck. Osteocalcin is also generally increased in patients with PHPT (77-79). Sclerostin is an important regulator of bone formation. Patients with PHPT  have low sclerostin levels, suggesting PTH down regulates sclerostin (80). As expected the bone formation marker, serum amino-terminal propeptide of type I collagen (P1NP), is negatively associated with sclerostin in PHPT (81). In a small series of 27 patients followed for up to a year post-PTX, circulating sclerostin increases shortly after post-surgery but return to the age reference range within 10 days (82).

 

Bone Resorption Markers

Markers of bone resorption include the osteoclast product, tartrate-resistant acid phosphatase (TRAP), and collagen breakdown products such as hydroxyproline, hydroxypyridinium cross-links of collagen, and N- and C-telopeptides of type 1 collagen (NTX and CTX) (74). Urinary hydroxyproline, once the only available marker of bone resorption, no longer offers sufficient sensitivity or specificity to make it useful. Although urinary hydroxyproline was frankly elevated in patients with osteitis fibrosa cystica, in mild asymptomatic PHPT it is generally normal. Hydroxypyridinium cross-links of collagen, pyridinoline (PYD), and deoxypyridinoline (DPD), on the other hand, are often elevated in PHPT. They return to normal after parathyroidectomy (83). DPD and PYD both correlate positively with PTH concentrations. Studies of NTX, CTX and TRAP are limited, although levels of the latter have been shown to be elevated (48). Thus, sensitive assays of bone formation and bone resorption are both elevated in mild PHPT.

 

Longitudinal Bone Turnover Marker Studies

Studies of bone turnover markers in the longitudinal follow-up of patients with PHPT indicate a reduction in these markers following parathyroidectomy. Information from our group (83,84), Guo et al (85), and Tanaka et al (86) all report declining levels of bone markers following surgery. The kinetics of change in bone resorption versus bone formation following parathyroidectomy provide insight into skeletal recovery. We have found that markers of bone resorption decline rapidly following successful parathyroidectomy, whereas indices of bone formation follow a more gradual decrease (83). Urinary PYD and DPD decreased significantly as early as 2 weeks following parathyroid surgery, preceding reductions in alkaline phosphatase. Similar data were reported from Tanaka et al (86), who demonstrated a difference in time course  between changes in NTX (reflecting bone resorption) and osteocalcin (reflecting bone formation) following parathyroidectomy, and Minisola et al (87), who reported a drop in bone resorptive markers and no significant change in alkaline phosphatase or osteocalcin. The persistence of elevated bone formation markers coupled with rapid declines in bone resorption markers indicate a shift in the coupling between bone formation and bone resorption toward an accrual of bone mineral postoperatively. More recent data indicate that levels of preoperative markers of bone turnover (formation and resorption) are positively associated with the extent of bone accrual after parathyroidectomy, though some of the patients included in this study had more severe PHPT than is typically seen in the United States today (88).

 

BONE DENSITOMETRY

The advent of bone mineral densitometry as a major diagnostic tool for osteoporosis occurred at a time when the clinical profile of PHPT was changing from a symptomatic to an asymptomatic disease. This fortuitous timing allowed questions about skeletal involvement in PHPT to be addressed when specific gross radiologic features of PHPT had all but disappeared. Observations of skeletal health in PHPT made by bone densitometry have established the importance of this technology in the evaluation of all patients with PHPT. The Consensus Development Conference on Asymptomatic Primary Hyperparathyroidism in 1990 implicitly acknowledged this point when bone mineral densitometry was included as a separate criterion for clinical decision making (89). Since that time, bone densitometry has become an indispensable component of both evaluating the patient and establishing clinical guidelines for management and monitoring.

 

The known physiologic proclivity of PTH to be catabolic at sites of cortical bone make a cortical site essential to any complete densitometric study of PHPT. By convention, the distal third of the radius is the site used. The early densitometric studies in PHPT also showed another physiologic property of PTH, namely, to preserve bone at cancellous sites. The lumbar spine is an important site to measure not only because it is predominantly cancellous bone, but also because postmenopausal women are at risk for cancellous bone loss. In PHPT, bone density at the distal third of the radius is diminished (90,91) while at the lumbar spine it is only minimally reduced (Figure 1). The hip region, containing relatively equal amounts of cortical and cancellous elements, shows bone density intermediate between the cortical and cancellous sites. The results support not only the notion that PTH is catabolic for cortical bone but also the view that PTH is generally protective against bone loss in cancellous bone (92-94). In postmenopausal women, the same pattern was observed (91). Postmenopausal women with PHPT, therefore, show a reversal of the pattern typically associated with postmenopausal bone loss. Rather than preferential loss of cancellous bone at the lumbar spine, the cortical site of the distal radius is more often affected in postmenopausal women with PHPT.

Figure 1. The pattern of bone loss in primary hyperparathyroidism. A typical pattern of bone loss is seen in asymptomatic patients with primary hyperparathyroidism. The lumbar spine is relatively well preserved while the distal radius (1/3 site) is preferentially affected. (Reprinted with permission from Silverberg SJ, Shane E, DeLaCruz L, et al. Skeletal disease in primary hyperparathyroidism. J Bone Mineral Res 1989;4:283-291).

The bone density profile in which there is relative preservation of skeletal mass at the vertebrae and reduction at the more cortical distal radius is not always seen in PHPT. Although this pattern is evident in the vast majority of patients, small groups of patients show evidence of vertebral osteopenia at the time of presentation. In our natural-history study, approximately 15% of patients had a lumbar spine Z score of less than –1.5 at the time of diagnosis (95). Only half of these patients were postmenopausal women, so not all vertebral bone loss could be attributed entirely to estrogen deficiency. These patients are of interest with regard to changes in bone density following parathyroidectomy and are discussed in further detail later. The extent of vertebral bone involvement will vary as a function of disease severity. In the typical mild form of the disease, the pattern described earlier is seen. When PHPT is more advanced, there will be more generalized involvement, with involvement of the lumbar bone. When PHPT is severe or more symptomatic, all bones can be extensively involved. Vitamin D deficiency in mild asymptomatic PHPT seems to have minimal effect on BMD with only slightly reduced BMD at the 1/3 radius in those with low vitamin D (96,97).

 

BONE HISTOMORPHOMETRY 

Analyses of percutaneous bone biopsies from patients with PHPT have provided direct information that could only be indirectly surmised by bone densitometry and by bone markers. Both static and dynamic parameters present a picture of cortical thinning, maintenance of cancellous bone volume, and a very dynamic process associated with high turnover and accelerated bone remodeling. Cortical thinning, inferred by bone mineral densitometry, is clearly documented in a quantitative manner by iliac crest bone biopsy (98,99). Van Doorn et al (100) demonstrated a positive correlation between PTH levels and cortical porosity. These findings are consistent with the known effect of PTH to be catabolic at endocortical surfaces of bone. Osteoclasts are thought to erode more deeply along the corticomedullary junction under the influence of PTH.

 

Histomorphometric studies have also contributed information about cancellous bone  in PHPT (100). Again, as suggested by bone densitometry, cancellous bone volume is well preserved in PHPT. This is seen as well among postmenopausal women with PHPT. Several studies have shown that cancellous bone is actually increased in PHPT as compared to normal subjects (101,102). When cancellous bone volume is compared among age- and sex-matched subjects with PHPT or postmenopausal osteoporosis, a dramatic difference is evident. Whereas postmenopausal women with osteoporosis have reduced cancellous bone volume, women with PHPT have higher cancellous bone volume (101). The region(s) of bone loss in PHPT is (are) directed toward the cortical bone compartment, with good maintenance of cancellous bone volume unless the PHPT is unusually active.

 

In PHPT, age-related bone loss appears to be mitigated. In a study of 27 patients with PHPT (10 men and 17 women), static parameters of bone turnover (osteoid surface, osteoid volume, and eroded surface) were increased, as expected, in patients relative to control subjects (103). However, in control subjects, trabecular number varied inversely with age, whereas trabecular separation increased with advancing age. These observations are expected concomitants of aging. In marked contrast, in the patients with PHPT, no such age dependency was seen. There was no relationship between trabecular number or separation and age in PHPT, suggesting that the actual plates and their connections were being maintained over time more effectively than one would have expected by aging per se. Thus, PHPT seems to retard the normal age-related processes associated with trabecular loss.

 

In PHPT, indices of trabecular connectivity are greater than expected, whereas indices of disconnectivity are decreased. When three matched groups of postmenopausal women were assessed (a normal group, a group with postmenopausal osteoporosis, and a group with PHPT), women with PHPT were shown to have trabeculae with less evidence of disconnectivity compared with normal, despite increased levels of bone turnover (102,103). Thus, cancellous bone is preserved in PHPT through the maintenance of well-connected trabecular plates. To determine the mechanism of cancellous bone preservation in PHPT, static and dynamic histomorphometric indices were compared between normal and hyperparathyroid postmenopausal women. In normal postmenopausal women, there is an imbalance in bone formation and resorption, which favors excess bone resorption. In postmenopausal women with PHPT, on the other hand, the adjusted apposition rate is increased. Bone formation, thus favored, may explain the efficacy of PTH at cancellous sites in patients with osteoporosis (92,104-106). Assessment of bone remodeling variables in patients with PHPT shows increases in the active bone-formation period (101) (Table 4). The increased bone formation rate and total formation period may explain the preservation of cancellous bone seen in this disease.

 

Table 4. Wall Width and Remodeling Variables in PHPT and Control Groups (Mean ± SEM)

Variable

PHPT (n = 19)

Control (n = 34)

P

Wall width (μm)

40.26 ± 0.36

34.58 ± 0.45

<.0001

Eroded perimeter (%)

9.00 ± 0.86

4.76 ± 0.39

<.0001

Osteoid perimeter (%)

26.84 ± 2.79

15.04 ± 1.09

<.0001

Osteoid width (μm)

13.39 ± 0.54

9.92 ± 0.36

<.0001

Single-labeled perimeter (%)

11.56 ± 1.63

4.47 ± 0.48

<.0001

Double-labeled perimeter (%)

10.41 ± 1.28

4.45 ± 0.65

<.0001

Mineralizing perimeter (%)

16.19 ± 1.75

6.68 ± 0.83

<.0001

Mineralizing perimeter/osteoid perimeter (%)

63.0 ± 5.0

44.04 ± 4.0

<.01

Mineral apposition rate (μm/day)

0.63 ± 0.03

0.63 ± 0.02

NS

Bone formation rate (μm 2/μm/day)

0.10 ± 0.01

0.042 ± 0.006

<.0001

Adjusted apposition rate (μm/day)

0.40 ± 0.04

0.29 ± 0.03

<.015

Activation frequency/yr

0.95 ± 0.12

0.45 ± 0.06

<.0002

Mineralization lag time (days)

44.0 ± 6.5

57.0 ± 8.9

NS

Osteoid maturation time (days)

22.5 ± 1.8

16.6 ± 0.9

<.003

Total formation period (days)

129.2 ± 21.0

208.8 ± 32.5

NS

Active formation period (days)

67.8 ± 5.1

57.3 ± 2.3

<.05

Resorption period (days)

48.4 ± 7.3

84.8 ± 25.0

NS

Remodeling period (days)

172.5 ± 25.2

299.9 ± 55.1

NS

NS, Not significant; PHPT, primary hyperparathyroidism. Modified from Dempster DW, Parisien M, Silverberg SJ, et al: On the mechanism of cancellous bone preservation in postmenopausal women with mild primary hyperparathyroidism, J Clin Endocrinol Metab. 1999; 84:1562-1566.

 

More recently, further analysis of trabecular microarchitecture has taken advantage of newer technologies that have largely been confirmatory. In a three-dimensional analysis of transiliac bone biopsies using microCT technology, a highly significant correlation was observed with the conventional histomorphometry described earlier (107). In comparison to age-matched control subjects without PHPT, postmenopausal women with PHPT had higher bone volume (BV/TV), higher bone surface area (BS/TV), higher connectivity density (Conn.D), and lower trabecular separation (Tb.Sp.). There were also less marked age-related declines in BV/TV and Conn.D as compared to controls, with no decline in BS/TV. Using the technique of backscattered electron imaging (qBEI) to evaluate trabecular BMD distribution (BMDD) in iliac crest bone biopsies, Roschger et al (108) showed reduced average mineralization density and an increase in the heterogeneity of the degree of mineralization, consistent with reduced mean age of bone tissue. Studies of collagen maturity using Fourier Transform Infrared Spectroscopy provide further support for these observations (109). Bone strength, therefore, in PHPT has to take into account a number of factors related to skeletal properties of bone besides BMD (110).

 

NEW IMAGING TECHNIQUES

Newer non-invasive skeletal imaging technologies offer new insight into the skeletal manifestations of PHPT beyond observations made by DXA and radiography. The trabecular bone score or TBS provides an indirect assessment of trabecular microstructure from the DXA image. In those without PHPT, it has been shown to predicts fracture independently of BMD (111). In PHPT, TBS reveals trabecular microstructural deterioration at the spine, despite preserved BMD by DXA at this site (112). High resolution peripheral quantitative CT (HRpQCT) is a technology that noninvasively and directly measures skeletal microstructure at the distal radius and tibia. Utilizing this technology, studies in patients with PHPT indicate not only cortical thinning, but additionally trabecular deficits at the radius and tibia (Figure 2) (113-115). At the radius, trabeculae were fewer, thinner, more widely and heterogeneously spaced.  At the tibia, trabeculae were more heterogeneously spaced (116). These deficits led to reduced stiffness when images were analyzed using microfinite element analysis, a technique that integrates structural and denisitometric information from the HRpQCT image into an estimated of mechanical competence. These recent findings, pointing to abnormalities in the trabecular compartment of bone, help to account for the increased risk of vertebral fractures (see below) observed in PHPT that had remained unexplained prior to the advent of such technologies (117-120). The difference in microskeletal abnormalities between the iliac crest bone biopsy data and HRpQCT may well reflect site-specific sampling differences.

Figure 2. High-resolution peripheral quantitative CT images of the radius in a patient with primary hyperparathyroidism (PHPT; left) and a normal control (right). Trabecular deterioration is evident in PHPT. Image from Stein EM, Silva BC, Boutroy S, et al. Primary hyperparathyroidism is associated with abnormal cortical and trabecular microstructure and reduced bone stiffness in postmenopausal women. J Bone Miner Res 2012

FRACTURES

Fractures were a common clinical event in classic PHPT. In modern PHPT, one would anticipate, based on the BMD patterns observed with DXA, an increased risk of peripheral fractures, but a reduction in vertebral fractures. While not all studies are consistent and much of the data is retrospective and/or cross-sectional, the majority of studies suggest an increased risk for vertebral fractures in patients with PHPT (117-123). Moreover, recent data indicates that many vertebral fractures are in fact clinically silent (118,124). The paradox of increased vertebral fracture risk despite preserved lumbar spine BMD in PHPT had remained unclear until the advent of TBS and HRpQCT, which clearly document trabecular deficits in addition to previously recognized cortical skeletal deterioration. Using the Danish National Patient registry and a nested case-control design, Ejlsmark-Svensson et al. recently showed that vertebral fractures in patients with PHPT occur at a higher BMD than in patients without PHPT, again pointing to the importance of other elements of bone quality in PHPT (125). In one recent study, among asymptomatic PHPT patients, only those who met surgical guidelines showed a higher incidence of vertebral fractures compared with controls (118).

 

The risk for hip fracture is not clearly increased in PHPT. In a study that focused on hip fracture, a population-based prospective analysis (mean of 17 years’ duration; 23,341 person years) showed women with PHPT in Sweden not to be at increased risk (126). The Mayo Clinic experience with PHPT and risk of fracture reviewed 407 cases of PHPT recognized during the 28-year period between 1965 and 1992 (117). Fracture risk was assessed by comparing fractures at a number of sites with numbers of fractures expected based on gender and age from the general population. The clinical presentation of these patients with PHPT was typical of the mild form of the disease, with the serum calcium being only modestly elevated at 10.9 ± 0.6 mg/dL. The data from this retrospective epidemiologic study indicate that overall fracture risk was significantly increased at many sites such as the vertebral spine, the distal forearm, the ribs, and the pelvis. There was no increase in hip fractures. One might expect to see an increased incidence of distal forearm fractures as seen in the May study, because the hyperparathyroid process tends to lead to a reduction of cortical bone (distal forearm) in preference to cancellous bone (vertebral spine). Unfortunately, there were no densitometric data provided in this study, so one could not relate bone density to fracture incidence.

 

The impact of PHPT on fracture incidence appears complex and may be site-specific. This relationship is likely influenced by site-specific changes in areal bone density, bone size, and microstructure. Excess PTH would induce cortical thinning due to endosteal bone resorption but would also increase periosteal apposition, thus increasing bone diameter. Decreased areal bone density would increase fracture risk, while increased bone diameter and preserved microstructure at certain sites, might protect against fractures. Prospective studies are needed to elucidate the site-specific risk of fracture in PHPT.

 

Nephrolithiasis and Renal Function

In the past, classic clinical descriptions of PHPT emphasized kidney stones as a principal complication of the disease (127). The cause of nephrolithiasis in PHPT is probably multifactorial. An increase in the amount of calcium filtered at the glomerulus resulting from hypercalcemia may lead to hypercalciuria despite the physiologic actions of PTH to facilitate calcium reabsorption. A component of absorptive hypercalciuria exists in this disorder. The enhanced intestinal calcium absorption is likely a result of increased production of 1,25-dihydroxyvitamin d, a consequence of PTH’s action to increase the synthesis of this active metabolite (128) (129). Indeed, urinary calcium excretion is correlated with 1,25-dihydroxyvitamin d levels (129,130). The skeleton provides yet another possible source for the increased levels of calcium in the glomerular filtrate. Hyperparathyroid bone resorption might contribute to hypercalciuria, and subsequently to nephrolithiasis, even though there is no convincing evidence to support this hypothesis (131). Finally, alteration in local urinary factors, such as a reduction in inhibitor activity or an increase in stone-promoting factors, may predispose some patients with PHPT to nephrolithiasis (131,132). It remains unclear whether the urine of patients with hyperparathyroid stone disease is different in this regard from that of other stone formers.

 

Studies in the 1970s and 1980s documented a higher incidence of renal stone disease than do reports of more recent experience.  Although the incidence of symptomatic nephrolithiasis today is much less common than it was in classical PHPT, kidney stones remain the most common manifestation of symptomatic PHPT (see Table 1). Estimates suggest symptomatic kidney stones in 15% to 20% of all patients (133). Screening for asymptomatic nephrolithiasis, indicates that the prevalence is actually much higher and this is now recommended in the most recent set of guidelines on the management of asymptomatic primary hyperparathyroidism (134-138).

 

The etiology of why stones develop in some but not others with PHPT has been postulated since the 1930s, but is still not well understood.  In the 1930s, it was generally accepted that bone and stone disease did not coexist in the same patient with classic PHPT (2,139). Albright and Reifenstein (139) theorized that a low dietary calcium intake led to bone disease, whereas adequate or high dietary calcium levels caused stone disease. Dent et al (140), who provided convincing evidence against this construct, proposed the existence of two forms of circulating PTH, one causing renal stones and the other causing bone disease. A host of mechanisms, including differences in dietary calcium, calcium absorption, forms of circulating PTH, and levels of 1,25-dihydroxyvitamin d, were proposed to account for the clinical distinction between bone and stone disease in PHPT (131,140). Today, there is no clear evidence for two distinct subtypes of PHPT or that the process affecting the skeleton and kidneys occur in different subsets of patients (127). Cortical bone demineralization is as common and as extensive in those with and without nephrolithiasis (127,131).

 

Although more recent work has suggested that 1,25-dihydroxyvitamin d plays an etiologic role in the development of nephrolithiasis in PHPT, not all studies are consistent with this premise (127,131,132,138,141). Other investigations have shown risk factors for nephrolithiasis include younger age and male sex, whereas degree of hypercalcemia and hypercalciuria, PTH levels and other urinary factors have less consistently been associated (38,135,136,138,141-143). Hypomagnesuria has recently been associated with silent kidney stones in PHPT (144).

 

Other renal manifestations of PHPT include hypercalciuria, which is seen in approximately 40% of patients, and nephrocalcinosis. The frequency of nephrocalcinosis is unknown, but it appears to be relatively uncommon today (135). Though there were clear reports of renal impairment in early descriptions of PHPT, the prevalence of renal dysfunction (estimated glomerular filtration rate (eGFR) <60 ml/min) today in patients with mild PHPT is low with recent studies suggesting rates of 15–17% (39,145,146). Neither the severity of PHPT nor having a history of nephrolithiasis were risk factors for reduced eGFR in a 2014 study in those with mild PHPT; instead, traditional risk factors, such as age, hypertension, use of antihypertensive medication, and fasting glucose levels were associated with poorer kidney function (145). Longitudinal data is reassuring in this regard, as renal function remains stable in PHPT over long periods of follow-up (52,147).  

 

Other Organ Involvement

CARDIOVASCULAR SYSTEM

Interest in the effect of PHPT on cardiovascular function is rooted in pathophysiologic observations of the hypercalcemic state. Hypercalcemia has been associated with increases in blood pressure, left ventricular hypertrophy, heart muscle hypercontractility, and arrhythmias (148,149). Furthermore, evidence of calcium deposition has been documented in the form of calcifications in the myocardium, heart valves, and coronary arteries. The association of overt cardiovascular symptomatology with modern-day PHPT is unclear because of inconsistencies between studies. An explanation for the inconsistent results reported in the literature on the cardiovascular manifestations of PHPT relates to the fact that the clinical profile of the disease has changed. As a result, the cohorts that have been studied have varied greatly in the severity of their underlying disease. This is particularly true in terms of the serum calcium and parathyroid hormone concentrations, with data from cohorts with marked hypercalcemia and hyperparathyroidism showing the most cardiovascular involvement. Because it is thought that both calcium and PTH can independently affect the cardiovascular system, such variability among cohorts can give rise to inconsistent results. Recent studies have focused on not only cardiovascular mortality, but also hypertension, coronary artery disease (CAD), valve calcification, left ventricular hypertrophy (LVH), carotid disease, and vascular stiffness.

 

Cardiovascular Mortality

There is little doubt that in very active PHPT, cardiovascular mortality is increased (150-153). Of some interest are the postoperative observations in which the higher cardiovascular mortality rate persists for years after cure (154). These observations differ markedly from those in which asymptomatic PHPT has been studied. Although limited, the studies have not shown any increase in mortality (155,156). The Mayo Clinic studies help to bring these observations together. In the mildly hypercalcemic individuals, overall and cardiovascular mortality was reduced, but in those whose serum calcium was in the highest quartile, cardiovascular mortality was increased (156). The idea that the more common asymptomatic form of PHPT is not associated with increased mortality is supported by data from Nilsson et al (157) and by other studies (158,159) in which more recently enrolled subjects had better survival than those who entered earlier and presumably had more active disease.

 

Hypertension

Hypertension, a common feature of PHPT when it is part of a MEN syndrome with pheochromocytoma or hyperaldosteronism, has also been reported as more prevalent in sporadic, asymptomatic PHPT than in appropriately matched control groups. The mechanism of this association is unknown, and the condition does not clearly remit following cure of the hyperparathyroid state (68,70,160-163).

 

Coronary Artery Disease (CAD) and Valve Calcification

Both calcium and PTH have independently been shown to be associated with coronary heart disease (164,165). Aside from autopsy studies such as those of Roberts and Waller (166), in which coronary atherosclerosis was seen in patients with marked hypercalcemia (16.8 to 27.4 mg/dL), the more recent literature has been controversial. When CAD is present in PHPT, it is most likely due to traditional risk factors rather than the disease itself (159,167,168). Some studies have actually shown that in mild PHPT, there is better exercise tolerance as determined by the electrocardiogram (169). Valve calcification, which is present in severe PHPT, has been shown to be more extensive (greater valve area) when present in those with mild PHPT versus controls (149,170,171), and is associated with increased PTH levels but it is not reversible with parathyroidectomy (171).

 

Left Ventricular Hypertrophy

Left ventricular hypertrophy (LVH) is a strong predictor of cardiovascular disease and mortality (172,173). LVH has been associated with PHPT in many, but not all, studies (174). A 2015 meta-analysis indicated that parathyroidectomy is associated with a decline in left ventricular mass and that higher levels of PTH pre-operatively predict a greater cardiovascular benefit. However, dissociating disease severity from study design (RCTs included individuals with lower levels of calcium and PTH than those included in observational studies) was not possible (175).

 

Vascular Function

Conflicting data exist regarding whether intima media thickness is increased in PHPT (176-180). Multiple studies have reported increased vascular stiffness, sometimes associated with PTH levels, in mild PHPT, but its reversibility with parathyroidectomy is inconsistent (180-183). Given conflicting data, most experts do not consider cardiovascular disease to be an indication for parathyroidectomy (137).

 

NEUROLOGICAL, PSYCHOLOGICAL, AND COGNITIVE FEATURES 

Descriptions of classical PHPT do indeed indicate neuropsychological features (2,184). The extent to which these features remain a part of the modern picture of PHPT as well as the exact mechanisms underlying them is unclear. Perhaps the most common complaints have been those of weakness and easy fatigability (67). Classic PHPT was formerly associated with a distinct neuromuscular syndrome characterized by type II muscle cell atrophy (185). Originally described by Vicale in 1949 (186), the syndrome consisted of easy fatigability, symmetric proximal muscle weakness, and muscle atrophy. Both the clinical and electromyographic features of this disorder were reversible after parathyroid surgery (187,188). In the milder, less symptomatic form of the disease that is common today, this disorder is rarely seen (189). In a group of 42 patients with mild disease, none had complaints consistent with the classic neuromuscular dysfunction described previously. Although more than half of all patients expressed nonspecific complaints of paresthesias and muscle cramps, electromyographic studies did not confirm the picture of past observations.

 

The “psychic groans” described by early observers of patients with classic PHPT remain a source of controversy today. Patients with PHPT often report some degree of behavioral and/or psychiatric symptomatology. A retrospective inquiry of patients with more severe disease showed a 23% incidence of psychiatric symptomatology (n = 441) (190). A number of studies suggest, however, that even ‘mild PHPT’ (serum calcium <12 mg/dl) is associated with non-specific symptoms such as depression, anxiety, fatigue, decreased quality of life (QOL), sleep disturbance, and cognitive dysfunction. Many, but not all, observational studies have indicated these features improve after parathyroidectomy (191). Three RCTs have investigated the reversibility of reduced QOL and psychiatric symptoms (192-194). Despite being of similar design and using similar assessment tools, all three randomized controlled trials came to different conclusions; one randomized controlled trial suggested parathyroidectomy prevents worsening of QOL and improves psychiatric symptoms (193); another randomized controlled trial indicated no benefit; and the third randomized controlled trial demonstrated improvement in QOL (192,194). One randomized controlled trial investigated changes in cognition after parathyroidectomy, but its small size precluded definitive conclusions being drawn (195).

 

While less well studied, some, but not all, studies have demonstrated reduced memory or impairment in other cognitive domains (195-201). It is unclear if cognition improves after parathyroidectomy because results of studies in which longitudinal control groups are compared to those undergoing parathyroidectomy are inconsistent (197,199,202-204). Recent work has turned to the potential mechanisms that contribute to cognitive dysfunction in PHPT. Our latest studies have addressed this issue. We hypothesized that cerebrovascular dysfunction (i.e., vascular stiffness) might underlie cognitive changes in patients with PHPT. While PTH correlated with cerebrovascular function as measured by transcranial Doppler, there was no consistent association between cerebrovascular function and cognitive performance (205). In a separate study, we utilized functional magnetic resonance imaging to assess if cerebral activation was altered by PHPT. Functional magnetic resonance imaging, or fMRI, is a non-invasive tool that maps brain function based on changes in blood flow (206). We found that PHPT was associated with differences in task-related neural activation patterns but no difference in cognitive performance. This may indicate compensation to maintain the same cognitive function, but there was no clear improvement in neural activation after parathyroidectomy (206). At present, most experts do not recognize cognitive or psychiatric symptoms as a sole indication for parathyroidectomy. Reasons for this include the failure to clearly demonstrate reversibility in randomized controlled trials, the inability to predict which patients might improve and a lack of a clear mechanism (137).

 

GASTROINTESTINAL MANIFESTATIONS  

Primary hyperparathyroidism has long been considered associated with an increased incidence of peptic ulcer disease. Most recent studies suggest that the incidence of peptic ulcer disease in PHPT is approximately 10%, a figure similar to its percentage in the general population. An increased incidence of peptic ulcer disease is seen in patients with PHPT resulting from MEN1, in which approximately 40% of patients have clinically apparent gastrinomas (Zollinger-Ellison syndrome). In those patients, PHPT is associated with increased clinical severity of gastrinoma, and treatment of the associated PHPT has been reported to benefit patients with Zollinger-Ellison syndrome (207,208). Despite this, current recommendations (Consensus Conference Guidelines for Therapy of MEN1) state that the coexistence of Zollinger-Ellison syndrome does not represent sufficient indication for parathyroidectomy, because medical therapy is so successful (208).

 

Although hypercalcemia can underlie pancreatitis, most large series have not reported an increased incidence of pancreatitis in patients with PHPT with serum calcium levels less than 12 mg/dL. The Mayo Clinic experience from 1950 to 1975 showed that only 1.5% of those with PHPT exhibited coexisting pancreatitis, and alternative explanations for pancreatitis were found for several patients. Regarding pancreatitis in pregnancy in patients with PHPT, these conditions may coexist, but there is no evidence for a causal relationship between the disorders (209).

 

OTHER SYSTEMIC INVOLVEMENT

Many organ systems were affected by the hyperparathyroid state in the past. Anemia, band keratopathy, and loose teeth are no longer part of the clinical syndrome of PHPT. Gout and pseudogout are seen infrequently, and their etiologic relationship to PHPT is not clear.

 

Unusual Presentations

NEONATAL PRIMARY HYPERPARATHYROIDISM

Neonatal PHPT is a rare form of the disorder caused by homozygous inactivation of the calcium-sensing receptor (210). When present in a heterozygous form, it is a benign hypercalcemic state known as familial hypercalciuric hypercalcemia (FHH). However, in the homozygous neonatal form, hypercalcemia is severe and the outcome is fatal unless recognized early. The treatment of choice is early subtotal parathyroidectomy to remove the majority of hyperplastic parathyroid tissue.

 

PRIMARY HYPERPARATHYROISM IN PREGNANCY

Primary hyperparathyroidism in pregnancy is primarily of concern for its potential effect on the fetus and neonate (211,212). Potential complications of PHPT in pregnancy include spontaneous abortion, low birth weight, supravalvular aortic stenosis, and neonatal tetany. The last condition is a result of fetal parathyroid gland suppression by high levels of maternal calcium, which readily crosses the placenta during pregnancy. These infants, accustomed to hypercalcemia in utero, have functional hypoparathyroidism after birth, and can develop hypocalcemia and tetany in the first few days of life. Today, with most patients (pregnant or not) presenting with a mild form of PHPT, an individualized approach to the management of the pregnant patient with PHPT is advised. A recent retrospective study suggested that PHPT did not increase the risk of abortion, birth weight, length, or Apgar score (213). Thus, many of those with mild disease can be followed safely, with successful neonatal outcomes without surgery. However, parathyroidectomy during the second trimester remains the traditional recommendation for this condition.

 

ACUTE PRIMARY HYPERPARATHYROIDISM

Acute PHPT is known variously as parathyroid crisis, parathyroid poisoning, parathyroid intoxication, and parathyroid storm. Acute PHPT describes an episode of life-threatening hypercalcemia of sudden onset in a patient with PHPT (214,215). Clinical manifestations of acute PHPT are mainly those associated with severe hypercalcemia. Nephrocalcinosis or nephrolithiasis is frequently seen. Radiologic evidence of subperiosteal bone resorption is also commonly present. Laboratory evaluation is remarkable not only for very high serum calcium levels but also for extremely high levels in PTH to approximately 20 times normal (215). In this way, acute PHPT resembles, in biochemical terms, parathyroid carcinoma. A history of persistent mild hypercalcemia has been reported in 25% of patients. However, given the rarity of this condition, the risk of developing acute PHPT in a patient with mild asymptomatic PHPT is very low. Intercurrent medical illness with immobilization may precipitate acute PHPT. Early diagnosis, with aggressive medical management followed by surgical cure, is essential for a successful outcome. The initial impression of patients who present in this manner, without an antecedent history of hypercalcemia, is often mistaken for malignancy. However, the parathyroid hormone level usually quickly clarifies the diagnosis to PHPT in most situations.

 

PARATHYROID CANCER

An indolent yet potentially fatal disease, parathyroid carcinoma accounts for less than 0.5% of cases of PHPT. In contrast to patients with PHPT due to benign parathyroid pathology, patients with parathyroid carcinoma typically have marked elevations in serum calcium and PHT. The cause of the disease is unknown, and no clear risk factors have been identified except for hereditary syndromes. There is no evidence to support the malignant degeneration of previously benign parathyroid adenomas (216). Parathyroid carcinoma has been reported particularly in hyperparathyroidism-jaw tumor (HPT-JT) syndrome (217-221), a rare autosomal disorder in which as many as 15% of patients will have malignant parathyroid disease. Because cystic changes are common, this disorder has also been referred to as cystic parathyroid adenomatosis (222). In HPT-JT, ossifying fibromas of the maxilla and mandible are seen in 30% of cases. Less commonly, kidney lesions, including cysts, polycystic disease, hamartomas, or Wilms’tumors, can be present (223). Parathyroid carcinoma has also been reported in familial isolated hyperparathyroidism (218,224). Parathyroid carcinoma, as defined pathologically, has been reported in MEN1 syndrome and with somatic MEN1 mutations (225-227). However, recurrent parathyroid disease in MEN1 may mimic but might not actually be a result of malignancy. Only one case of parathyroid carcinoma has been reported in the MEN2A syndrome (228).

 

Loss of the retinoblastoma tumor suppressor gene was formerly considered a marker for parathyroid cancer (229), but more recent studies do not unequivocally support this impression (230). Work by Shattuck et al (231,232) has provided new insights into the molecular pathogenesis of parathyroid cancer. Parathyroid carcinomas from 10 of 15 patients with sporadic parathyroid cancer carried a mutation in the HRPT2 gene. The HRPT2 gene encodes for the parafibromin protein that was shown to be mutated in a substantial number of patients with parathyroid cancer. Marcocci et al (216) have reviewed this topic, pointing out a potential role for parafibromin in parathyroid cancer. In three of 15 patients with parathyroid cancer, Shattuck et al (231) showed that the mutation was in the germline. The presence of the mutation in the germline suggests that this disease might be related in some way to the HPT-JT syndrome, in which this gene has been implicated (231). In addition, there is an increased risk of parathyroid cancer in the HPT-JT syndrome. In fact, certain clinical features in patients with a germline mutation and in their relatives are indicative of the HPT-JT syndrome or phenotypic variants (220,223,224).

 

Manifestations of hypercalcemia are the primary effects of parathyroid cancer. The disease tends not to have a bulk tumor effect, spreading slowly in the neck. Metastatic disease is a late finding, with lung (40%), liver (10%), and lymph node (30%) involvement seen most commonly. The clinical profile of parathyroid cancer differs from that of benign PHPT in several important ways (216). First, no female predominance is seen among patients with carcinoma. Second, elevations in serum calcium and PTH are far greater. Consequently, the hyperparathyroid disease tends to be much more severe, with the classic targets of PTH excess involved in most cases. Nephrolithiasis or nephrocalcinosis is seen in up to 60% of patients; overt radiologic evidence of skeletal involvement is seen in 35% to 90% of patients. A palpable neck mass, distinctly unusual in benign PHPT, has been reported in 30% to 76% of patients with parathyroid cancer (233). Grossly, malignant glands are large, often exceeding 12 g. They tend to be adherent to adjacent structures. Microscopically, thick, fibrous bands divide the trabecular arrangement of the tumor cells. Capsular and blood vessel invasion is common by these cells, which often contain mitotic figures (233). Treatment is reviewed below.

 

Parathyromatosis

Originally reported in 1975, fewer than one-hundred cases of parathyromatosis have been described in the literature (234,235).  The condition is characterized histologically by small collections or nodules of parathyroid cells embedded within surrounding soft tissue outside the parathyroid gland capsule margins (24,236). Parathyromatosis may rarely be embryologic in origin or, more often, is secondary to tissue seeding during parathyroid surgery or fine needle aspiration (24,234,237).  The majority of cases have been described in those who have undergone parathyroid surgery for secondary hyperparathyroidism associated with end-stage renal disease (24,235). While clinically and biochemically similar to primary hyperparathyroidism, parathyromatosis is associated with recurrent or persistentdisease (24). The diagnosis is typically made at the time of surgery, although pre-operative imaging has been reported to be diagnostically helpful (238,239). Management is challenging and complete cure is uncommon. Treatment involves complete surgical excision of all parathyromatosis nodules and/or parathyroid tissue (24,240). Intra-operative parathyroid hormone level monitoring and pathologic review of frozen sections at the time of surgery may be helpful to increase surgical success. Successful accounts of medical therapy with calcimimetics and bisphosphonates have been reported (24,235).

 

EVALUATION

The diagnosis of PHPT is confirmed by demonstrating an elevated or inappropriately normal PTH level in the face of hypercalcemia. Further biochemical assessment should include serum phosphorus, alkaline phosphatase activity, vitamin D metabolites, albumin, and creatinine. A morning 2-hour or 24-hour urine collection should be obtained for calcium and creatinine. A urinary bone resorption marker such as serum CTX or urinary N-telopeptide can be helpful. Bone densitometry is performed in all patients. It is important to obtain densitometry at three sites: the lumbar spine, the hip, and the distal third of the radius. Because of the differing amounts of cortical and cancellous bone at the three sites and the different effects of PTH on cortical and cancellous bone, measurement at all three sites gives the most accurate clinical assessment of skeletal involvement in PHPT. Bone biopsy is not routinely obtained in the evaluation of PHPT, but is essential in research. In the most recent guidelines, spinal imaging is recommended to assess for clinically silent vertebral fractures (137). This can be vertebral X-rays, vertebral fracture assessment or TBS score, the latter two obtained by the DXA image. While symptomatic kidney stones are present in 15% to 20% of patients by history, the finding that many more have clinically silent nephrolithiasis has led to the recommendation to obtain renal ultrasound, CT, or abdominal x-ray to assess for either nephrolithiasis or nephrocalcinosis. 

 

NATURAL HISTORY

Since the early 1990s, new knowledge of the natural history of PHPT with or without surgery has been very helpful in guiding decisions regarding surgery in patients with asymptomatic PHPT. The authors and their colleagues have conducted the longest prospective observational trial (52,241). This project began in 1984 in an effort to define the natural history of asymptomatic PHPT. The study included detailed analyses of pathophysiologic, densitometric, histomorphometric, and other skeletal features of PHPT (52,241). Much of the information gleaned from that study has been summarized already in this chapter. The 15-year follow-up to this study constitutes the longest natural-history study of this disorder (241).

 

Recommendations for surgery or observation were made based on the 1990 set of National Institutes of Health guidelines, but both groups included patients who did or did not meet surgical guidelines. This is because some patients opted for surgery even if they did not meet the guidelines, whereas others opted for a conservative approach even if they did meet guidelines for surgery. As will be described in the following sections, this imperfect design was followed by three studies that were truly randomized but were of much shorter duration. The results with regard to natural history from all studies are remarkably concordant.

 

Natural History with Surgery

Successful parathyroidectomy results in permanent normalization of the serum calcium and PTH levels. Postoperatively, there is  a marked improvement in BMD at all sites (lumbar spine, femoral neck, and distal one-third radius) amounting to gains greater than 10% (52) (Figure 3), The improvement is most rapid at the lumbar spine, followed by gains at the hip regions and the distal 1/3 radius during the 15-year follow-up (241). The improvements were seen in those who met and did not meet surgical criteria at study entry, confirming the salutary effect of parathyroidectomy in this regard on all patients.

Figure 3. Improvement in bone density after parathyroid surgery. Data shown are the cumulative percentage changes from baseline over 15 years of follow-up in patients who underwent parathyroidectomy.

Natural History Without Surgery

In subjects who did not undergo parathyroid surgery, serum calcium remained stable for about 12 years, with a tendency for the serum calcium level to rise in years 13 to 15 (241). Other biochemical indices such as PTH, vitamin D metabolites, and urinary calcium did not change for the entire 15 years of follow-up in the group as a whole. Bone density at all three sites remained stable for the first 8 to 10 years. However, after this period of stability, declining cortical BMD was seen at the hip and more dramatically at the distal one-third radius site. Although the numbers became limiting after 10 years of follow-up, it is noteworthy that a small majority of the subjects lost more than 10% of their BMD during the 15 years of observation. Even though this decline was observed in the majority of subjects, only 37% of subjects met one or more guidelines for surgery after the 15 years of follow-up.

 

Randomized Studies of the Natural History of Asymptomatic Primary Hyperparathyroidism

The long natural history study of asymptomatic PHPT has added much to our knowledge about this disease throughout time. Subsequent randomized trials confirm the observational data, but are limited by their shorter duration. In 2004, Rao et al (242) reported on 53 subjects, assigned either to parathyroid surgery (n = 25) or to no surgery (n = 28). The follow-up lasted for at least 2 years. There was a significant effect of parathyroidectomy on BMD at the hip and femoral neck but not the spine or forearm. Bollerslev et al (192) reported in 2007 the interim results of their randomized trial of parathyroidectomy versus no surgery. This study from three Scandinavian countries was larger, with 191 patients who were randomized to medical observation or to surgery. After surgery, biochemical indices normalized and BMD increased. In the group that did not undergo parathyroid surgery, BMD did not change. Also, in 2007, Ambrogini et al (194) reported the results of their randomized controlled trial of parathyroidectomy versus observation. Surgery was associated with a significant increase in BMD of the lumbar spine and hip after 1 year.

 

Whether fracture risk decreases after parathyroidectomy is not clear. The study by Bollerslev reported on vertebral fracture risk reduction at 5 years after initial treatment allocation. That study indicated that successful parathyroidectomy versus observation was associated with a reduction in vertebral fracture risk that was of borderline statistical significance (243).

 

GUIDELINES FOR PARATHYROIDECTOMY

Parathyroidectomy remains the only currently available option to cure PHPT. As the disease profile has changed, questions have arisen concerning the advisability of surgery in asymptomatic patients. If asymptomatic patients have a benign natural history, the surgical alternative is not an attractive one. On the other hand, asymptomatic patients may display levels of hypercalcemia or hypercalciuria that cause concern for the future. Similarly, bone-mass measurements can be frankly low. In an effort to address such issues, there have been four consensus development conferences (in 1991, 2002, 2008, and 2013) on the management of asymptomatic PHPT (89,137,244-247). The most recent guidelines that emerged from the 2013 conference are helpful to the clinician faced with the asymptomatic hyperparathyroid patient: All symptomatic patients are advised to undergo parathyroidectomy. Surgery is advised in asymptomatic patients with (1) serum calcium greater than 1 mg/dL higher than the upper limit of normal; (2) renal guidelines: reduction in creatinine clearance to less than 60 mL/min; urinary calcium excretion >400 mg/24 h with increased stone risk; or presence of nephrolithiasis or nephrocalcinosis on renal imaging; (3) skeletal guidelines: reduced bone density T-score < –2.5 at any site; or vertebral compression fracture on an imaging study; and (4) age younger than 50 years. The most recent guidelines are shown in Table 5. A noteworthy change in the guidelines reflects the fact that asymptomatic kidney stones and vertebral compression fractures are now considered as indications for parathyroidectomy.

 

Table 5. Comparison of New and Old Guidelines for Surgery in Asymptomatic Primary Hyperparathyroidism

 

 

1990 NIH Consensus Conference

2002 NIH Workshop

2008 International Workshop

2013 International Workshop

Serum calcium

1-1.6 mg/dL elevation

1.0 mg/dL elevation

1.0 mg/dL elevation

1.0 mg/dL elevation

Renal

24-h urine calcium >400 mg
Creatinine Cl reduced by 30%

24-h urine calcium >400 mg
Creatinine Cl reduced by 30%

No 24-h urine
Creatinine clearance: <60 mL/min

24-h urine for FHH/stone risk
U Ca >400 mg/day
Creatinine clearance: <60 mL/min
Calcification on renal imaging

Bone

Z-score < −2.0 in forearm

T-score < −2.5 at any site

T-score < −2.5
Fragility fracture

T-score < −2.5
Vertebral fracture on imaging

Age

<50

<50

<50

<50

FHH, Familial hypercalciuric hypercalcemia. Columns 3 and 4 modified from Bilezikian JP, Brandi ML, Eastell R, et al: Guidelines for the management of asymptomatic primary hyperparathyroidism: summary statement from the Fourth International Workshop, J Clin Endocrinol Metab. 2014; 99:3561-3569

 

A number of points were discussed that did not lead to specific recommendations, including the issues of the neurocognitive and cardiovascular aspects of PHPT. The workshop panel also acknowledged a potential role of vitamin D deficiency in fueling processes associated with abnormal parathyroid glandular activity. Finally, the panel also reaffirmed the entity of normocalcemic PHPT, but noted that there are insufficient data to provide evidence-based guidelines for management.

 

SURGERY

A large percentage of those patients who meet the surgical guidelines listed in Table 5 are asymptomatic. Some asymptomatic patients who meet surgical guidelines elect not to have surgery for varying reasons including personal choice, intercurrent medical conditions, and previous unsuccessful parathyroid surgery. Conversely, there are patients who meet none of the NIH guidelines for parathyroidectomy but opt for surgery nevertheless. Physician and patient input remain important factors in the decision regarding parathyroid surgery.

 

Preoperative Localization of Hyperfunctioning Parathyroid Tissue

A number of imaging tests have been developed and have been applied singly or in combination to address the challenge of preoperative localization. The rationale for locating abnormal parathyroid tissue before surgery is that the glands can be notoriously unpredictable in their location. Although most parathyroid adenomas are identified in regions proximate to their embryologically intended position (the four poles of the thyroid gland), many are not. In such situations, previous surgical experience and skill are needed to locate the ectopic parathyroid gland. In such hands, 95% of abnormal parathyroid glands will be discovered and removed at the time of initial parathyroid surgery. However, in the patient with previous neck surgery, even expert parathyroid surgeons do not generally achieve such high success rates. Preoperative localization of the abnormal parathyroid tissue can be extremely helpful under these circumstances. Preoperative imaging is also necessary for any patient who will undergo parathyroidectomy using a minimally invasive approach. Imaging studies should not be used for the diagnosis of PHPT because the sensitivity and specificity of various imaging modalities varies with some having false-positive rates as high as 25% (248).

 

NONINVASIVE IMAGING

Noninvasive parathyroid imaging studies include technetium (Tc)-99m sestamibi scintigraphy, ultrasound, computed tomography (CT) scanning, magnetic resonance imaging (MRI), and positron emission tomography (PET) scanning. Tc-99m sestamibi is generally regarded to be the most sensitive and specific imaging modality, especially when it is combined with single-photon emission CT (SPECT). For the single parathyroid adenoma, sensitivity has ranged from 80% to 100%, with a 5% to 10% false-positive rate. On the other hand, sestamibi scintigraphy and the other localization tests have a relatively poor record in the context of multiglandular disease (249). The success of ultrasonography is highly operator dependent (250). In centers where there is great expertise, this noninvasive approach is most attractive. Abnormalities identified by ultrasound as possible parathyroid tissue may prove to be a thyroid nodule or lymph node, which underscores the importance of the skill and experience of the ultrasonographer. Rapid spiral thin-slice CT scanning of the neck and mediastinum with evaluation of axial, coronal, and sagittal views can add much to the search for elusive parathyroid tissue, albeit with attendant higher radiation exposure (251). Four-dimensional (4D) CT has emerged as a promising method and consists of multiphase CT acquired at non-contrast, contrast-enhanced, arterial and delayed phases. In a recent study, 4D CT was superior compared with sestamibi SPECT/CT (252). MRI can also identify abnormal parathyroid tissue, but it is time consuming and expensive. It is also less sensitive than the other noninvasive modalities. It can nonetheless be useful when the search with these other noninvasive approaches has been unsuccessful. PET with or without simultaneous CT scan (PET/CT) can be used, but like MRI, it is expensive and does not have the kind of experiential basis that make it attractive. There are also specificity issues because FDG, the scanning agent, accumulates in the thyroid, making differentiation between parathyroid adenoma and thyroid nodules difficult. Recently, 18F-fluorocholine (FCH) positron emission tomography (PET) has been employed for the detection of parathyroid adenomas.

 

INVASIVE IMAGING 

Parathyroid Fine-Needle Aspiration

Fine-needle aspiration (FNA) of a parathyroid gland, identified by any of the aforementioned modalities, can be performed and the aspirate analyzed for PTH. This technique is not recommended for routine de novo cases.(253) A theoretical concern with this approach is the possibility that parathyroid cells could be deposited outside the parathyroid gland in the course of the aspiration. Autoseeding of parathyroid tissue would be an unwanted consequence of this procedure if it were to occur.

 

Arteriography and Selective Venous Sampling for Parathyroid Hormone

In situations where the gland has not been identified by any of the techniques described, the combination of arteriography and selective venous sampling can provide both anatomic and functional localization of abnormal parathyroid tissue. This approach, however, is costly and requires an experienced interventional radiologist. It is also performed in only a few centers in the United States. This approach is reserved for those individuals who have undergone previous unsuccessful parathyroid surgery in whom all other localization techniques have failed (254).

 

Surgical Approach

In the hands of an expert parathyroid surgeon, parathyroidectomy is a successful with infrequent complications. A standard surgical approach is the four-gland parathyroid exploration under general or local anesthesia, with or without preoperative localization. This approach has been reported to lead to surgical cure in more than 95% of cases (255). Several alternative approaches have emerged that focus on the single gland and not the total four-gland neck exploration that was routinely used in the past. Unilateral approaches are appealing for a disease in which most often only a single gland is involved. These procedures include a unilateral operation in which the gland on the same side that harbors the adenoma is ascertained to be normal. Because multiple parathyroid adenomas are unusual, a normal parathyroid gland is considered by some to be sufficient evidence for single-gland disease. Another limited surgical approach that has emerged in many centers as the approach of choice is the minimally invasive parathyroidectomy (MIP) (256,257). Preoperative parathyroid imaging is necessary, and the procedure is directed only to the site where the abnormal parathyroid gland has been visualized (258). Preoperative blood is obtained for comparison of the PTH concentration with an intraoperative sample(s) obtained after removal of the “abnormal” parathyroid gland. The availability of a rapid PTH assay in or near the operating room is necessary for this procedure. If the ten-minute post-excision PTH level falls by more than 50% compared to baseline, and into the normal range, the gland that has been removed is considered to be the sole source of overactive parathyroid tissue, and the operation is terminated. If the PTH level does not fall by more than 50%, into the normal range, the operation is extended to a more traditional one in a search for other overactive parathyroid tissue. There is a risk (albeit small) that the minimally invasive procedure may miss other overactive gland(s) that are suppressed in the presence of a dominant gland. The MIP procedure seems to be as successful, in the range of 95% to 98%, as more standard approaches (259,260). According to a recent meta-analysis that included more than 12,000 patients, MIP was associated with similar rates of success, disease recurrence, persistence, overall failure, and reoperation (261). The operative time was significantly shorter, with a lower overall complication rate for MIP compared to bilateral neck exploration. In Europe, MIP is being performed with an endoscopic camera, but this does not offer any advantage other than a smaller incision (262,263). Yet another variation on this theme is the use of preoperative sestamibi scanning with an intraoperative gamma probe to help locate enlarged parathyroid glands.

 

Immediate Postoperative Course

After surgery, biochemical indices return rapidly to normal (52,264). Serum calcium and PTH levels normalize, and urinary calcium excretion falls by as much as 50%. Serum calcium levels no longer fall rapidly into the hypocalcemic range, a situation characteristic of an earlier time when PHPT was a symptomatic disease with overt skeletal involvement. The acute reversal of PHPT was associated with a robust deposition of calcium into the skeleton at a pace that could not be compensated for by supplemental calcium. Thus, postoperative hypocalcemia was routine and was sometimes a serious short-term complication (“hungry bone syndrome”). Occasionally, postoperative hypocalcemia still occurs, especially if preoperative bone turnover markers are markedly elevated or there is concomitant vitamin D deficiency. More typically, however, the early postoperative course is not complicated by symptomatic hypocalcemia.

 

After successful parathyroid surgery, biochemical indices of the disease return to normal and BMD improves, as mentioned. The capacity of the skeleton to restore itself is seen dramatically in young patients with severe PHPT. Kulak et al (265) reported two patients with osteitis fibrosa cystica who experienced increases in bone density that ranged from 260% to 430% in a period of 3 to 4 years following surgery. Tritos and Hartzband (266) and DiGregorio (267) have made similar observations.

 

MEDICAL MANAGEMENT

Patients who do not meet any surgical guidelines are often followed without intervention. The most recent guidelines for management of asymptomatic PHPT restated the position that it is reasonable to pursue a nonsurgical course of management for those who do not meet criteria for surgery, at least for a period of years. In those patients who are not going to have parathyroid surgery, the Workshop (137) suggested a set of monitoring steps that are summarized in Table 6. This includes annual measurements of the serum calcium concentration, a calculated creatinine clearance, and regular monitoring of BMD.

 

Table 6. Comparison of New and Old Management Guidelines for Patients with Asymptomatic Primary Hyperparathyroidism Who Do Not Undergo Parathyroid Surgery

 

Measurement

Older Guidelines

Newer Guidelines

Serum calcium

Semiannually

Annually

24-h urinary calcium

Annually

Not recommended

Creatinine clearance

Annually

Not recommended

Serum creatinine

Annually

Annually

Bone density

Annually

Annually or biannually

Abdominal x-ray

Annually

Not recommended

From Bilezikian JP, Brandi ML, Eastell R, et al: Guidelines for the management of asymptomatic primary hyperparathyroidism: summary statement from the Fourth International Workshop, J Clin Endocrinol Metab. 2014; 99:3561-3569.

 

Ideal medical therapy of PHPT would provide the equivalent to a medical parathyroidectomy. Such an agent would normalize serum calcium and PTH levels as well as urinary calcium excretion, increase BMD and lower fracture risk, and reduce the risk of kidney stones. Unfortunately, no currently available single drug meets all these criteria. The following medications can achieve some of these goals and might be considered in patients not having surgery in whom it is desirable to lower serum or urinary calcium levels or increase BMD.

 

General Measures

Patients should be instructed to remain well hydrated and to avoid, if possible, medications that can increase serum calcium (e.g.  thiazide diuretics). Prolonged immobilization, which can raise the serum calcium concentration further and induce hypercalciuria, should also be avoided.

 

DIET AND SUPPLEMENTS

Dietary management of PHPT has long been an area of controversy. Many patients are advised to limit their dietary calcium intake because of the hypercalcemia. However, it is well known that low dietary calcium can lead to increased PTH levels in normal individuals (268-270). In patients with PHPT, even though the abnormal PTH tissue is not as sensitive to slight perturbations in the circulating calcium concentration, it is still possible that PTH levels will rise when dietary calcium is tightly restricted. Conversely, diets enriched in calcium could suppress PTH levels in PHPT, as shown by Insogna et al (271). Dietary calcium could also be variably influenced by ambient levels of 1,25-dihydroxyvitamin d. In patients with normal levels of 1,25-dihydroxyvitamin d, Locker et al (272) noted no difference in urinary calcium excretion between those on high (1000 mg/day) and low (500 mg/day) calcium intake diets. On the other hand, in those with elevated levels of 1,25-dihydroxyvitamin d, high calcium diets were associated with worsening hypercalciuria. This observation suggests that dietary calcium intake in patients can be liberalized to 1000 mg/day if 1,25-dihydroxyvitamin d levels are not increased but should be more tightly controlled if 1,25-dihydroxyvitamin d levels are elevated. Although calcium supplements are not specifically recommended in those with PHPT and osteoporosis, small doses do not seem to exacerbate hypercalcemia or hypercalciuria if the diet is deficient (273).  Most experts recommend that patients with PHPT who are going to be followed without surgery have an intake of calcium that is consistent with nutritional guidelines for a normal population. 

 

Recent guidelines recommend maintaining 25-hydroxyvitamin D to levels of 21–30 ng/ml with conservative doses of vitamin D (600–1000 IU daily) based on data showing that vitamin D repletion lowers PTH levels (274). Higher levels of vitamin D might be beneficial. A 2014 RCT of cholecalciferol (2,800 IU daily versus placebo) indicated that treatment increased 25-hydroxyvitamin d levels from 20 ng/ml to 37.8 ng/ml, lowered levels of PTH, and increased lumbar spine BMD without having a deleterious effect on serum or urinary calcium levels (275).

 

PHARMACEUTICALS

Phosphate

Oral phosphate can lower the serum calcium by up to 1 mg/dL (276,277). A complex interplay of mechanisms leads to this moderating effect of oral phosphate. First, calcium absorption falls in the presence of intestinal phosphorus. Second, concomitant increases in serum phosphorus will tend to reduce circulating 1,25-dihydroxyvitamin d levels. Third, phosphate can be an antiresorptive agent. Finally, increased serum phosphorus reciprocally lowers serum calcium. Problems with oral phosphate include limited gastrointestinal tolerance, possible further increase in PTH levels, and the possibility of soft-tissue calcifications after long-term use. It is essentially not used any longer in the management of PHPT.

 

Estrogens and Selective Estrogen-Receptor Modulators

The earliest studies on the use of estrogen replacement therapy in PHPT date back to the early 1970s. A 0.5 to 1.0 mg/dL reduction in total serum calcium levels in postmenopausal women with PHPT who received estrogen was seen along with a lowering of urinary calcium (278,279). Most studies indicated no change in PTH (279-281). A randomized controlled trial of conjugated estrogen (0.625 mg daily plus medroxyprogesterone 5 mg daily) versus placebo indicated that hormone-replacement therapy effectively increases BMD at all skeletal sites in patients with PHPT, with the greatest increases at the lumbar spine (281). This randomized controlled trials, however, did not confirm the calcium-lowering effect of earlier uncontrolled studies (274). In view of concerns expressed about chronic estrogen use in the Women’s Health Study, estrogen use is not often recommended for medical management of hyperparathyroidism.

 

Raloxifene, a selective estrogen-receptor modulator, has been studied in PHPT, but the data are sparse. In a short-term (8-week) trial of 18 postmenopausal women, raloxifene (60 mg/day) was associated with a statistically significant although small (0.5 mg/dL) reduction in the serum calcium concentration and in markers of bone turnover (282). No long-term data or data on bone density are available.

 

Bisphosphonates and Denosumab

Bisphosphonates are conceptually attractive in PHPT because they are antiresorptive agents with an overall effect of reducing bone turnover. Although they do not affect PTH secretion directly, bisphosphonates could reduce serum and urinary calcium levels. Early studies with the first-generation bisphosphonates were disappointing. Etidronate has no effect (283). Clodronate use was associated in several studies with a reduction in serum and urinary calcium (284), but the effect was transient.

 

Alendronate has been studied most extensively in PHPT. Studies by Rossini et al (285) and Hassani et al (286) were followed by those of Chow et al (287), Parker et al (288), and Kahn et al (289). These studies were all characterized by a randomized, controlled design. Typically, BMD of the lumbar spine and hip regions increases along with reductions in bone turnover markers (Figure 4). Except for the study of Chow et al (287), serum calcium was unchanged. These results suggest that bisphosphonates may be useful in patients with low bone density in whom parathyroid surgery is not to be performed. One small study suggests that denosumab increases BMD in women with PHPT to a greater extent than patients without PHPT but with osteoporosis (290).

Figure 4. The effect of alendronate on bone mineral density in primary hyperparathyroidism. With alendronate, bone mineral density increases significantly after 1 year, while the placebo group shows no change until it is crossed over to alendronate in year 2. (Modified from reference Khan AA, Bilezikian JP, Kung AWC, et al: Alendronate in primary hyperparathyroidism: a double-blind, randomized, placebo-controlled trial. J Clin Endocrinol Metab 2004;89:3319-3325).

Inhibition of Parathyroid Hormone

The most specific pharmacologic approach to PHPT is to inhibit the synthesis and secretion of PTH from the parathyroid glands, such as those that act on the parathyroid cell calcium-sensing receptor. This G protein–coupled receptor recognizes calcium as its cognate ligand (291-293). When activated by increased extracellular calcium, the calcium-sensing receptor signals the cell via a G protein–transducing pathway to raise the intracellular calcium concentration, which inhibits PTH secretion. Molecules that mimic the effect of extracellular calcium by altering the affinity of calcium for the receptor could activate this receptor and inhibit parathyroid cell function. The phenylalkylamine (R)-N(3-methoxy-a-phenylethyl)-3-(2-chlorophenyl)-1-propylamine (R-568) is one such calcimimetic compound. R-568 was found to increase cytoplasmic calcium and to reduce PTH secretion in vitro, as well as in normal postmenopausal women (294,295). This drug was also shown to inhibit PTH secretion in postmenopausal women with PHPT (296). A second-generation ligand, cinacalcet, has been the subject of more extensive investigations in PHPT and is now approved for the treatment of severe hypercalcemia in PHPT when surgery cannot be pursued. Studies conducted by the authors and their colleagues (297-299) indicate that this drug can reduce the serum calcium concentration to normal in PHPT, but despite normalization of the serum calcium concentration, PTH levels do not return to normal; they do fall by 35% to 50% after administration of the drug. Urinary calcium excretion does not change; serum phosphorus levels increase but are maintained in the lower range of normal; and 1,25-dihydroxyvitamin D levels do not change. The average BMD does not change, even after 5 years of administration of cinacalcet (300). Marcocci et al (299) have shown that cinacalcet is effective in subjects with intractable PHPT. Silverberg et al (301) have shown that cinacalcet reduces calcium levels effectively in inoperable parathyroid carcinoma.

 

Hydrochlorothiazide

Though avoiding agents that exacerbate hypercalcemia is generally recommended in patients with PHPT, a recent study has recently reexamined the role of thiazides in patients with PHPT. This retrospective analysis of 72 patients suggested that thiazides might not increase serum levels of calcium in PHPT as they can do in normal individuals. Hydrochlorothiazide (12.5–50 mg daily for 3.1 years on average) was associated with a decrease in urinary calcium excretion but no change in serum levels of calcium (302). Smaller and cross-sectional studies have suggested similar results, although it is unclear if hydrochlorothiazide reduces the risk of nephrolithiasis (303,304). Given the heterogeneity of doses used, and the absence of larger, (preferably) randomized trial data, recommending thiazide use routinely in PHPT is premature. However, thiazides could be considered in those who refuse surgery or are poor surgical candidates but at high risk of nephrolithiasis in whom the benefit is thought to outweigh the risk as long as serum levels of calcium are monitored regularly.

 

TREATMENT OF PARATHYROID CANCER

Surgery is the only effective therapy currently available for parathyroid cancer. The greatest chance for cure occurs with the first operation. After the disease recurs, cure is unlikely, although the disease may smolder for many years thereafter. The tumor is not radiosensitive, although there are isolated reports of tumor regression with localized radiation therapy. Traditional chemotherapeutic agents have not been useful. When metastasis occurs, isolated removal is an option, especially if only one or two nodules are found in the lung. Such isolated metastasectomies are never curative but they can lead to prolonged remissions, sometimes lasting for several years. Similarly, local debulking of tumor tissue in the neck can be palliative, although malignant tissue is invariably left behind.

 

Chemotherapy has had a very limited role in this disease. Bradwell and Harvey (305) have attempted an immunotherapeutic approach by injecting a patient who had severe hypercalcemia resulting from parathyroid cancer with antibodies raised to their own circulating PTH. Coincident with a rise in antibody titer to PTH, previously refractory hypercalcemia fell impressively. A more recent report (306) provided evidence of the antitumor effect in a single case of PTH immunization in metastatic parathyroid cancer.

 

Attention has been focused instead on control of hypercalcemia. Intravenous bisphosphonates have been used to treat severe hypercalcemia. Although efficacious in the short term, they do not provide an approach that allows long-term outpatient normalization of serum calcium levels. Denosumab has more recently been reported to treat resistant hypercalcemia in patients with parathyroid carcinoma (307,308).

 

The calcimimetic agents offer a newer approach. Our group (309) reported on a single patient treated with the calcimimetic, R-568; despite widely metastatic disease, the patient showed serum calcium levels that were maintained within a range that allowed him to return to normal functioning for nearly 2 years. A wider experience by Silverberg et al (301) showed that cinacalcet is useful in the management of parathyroid cancer. The U.S. Food and Drug Administration approved this calcimimetic for the treatment of hypercalcemia in patients with parathyroid cancer. Use of this agent in parathyroid cancer led to improvement in serum calcium levels and a decrease in symptoms of nausea, vomiting, and mental lethargy, which are common concomitants of marked hypercalcemia. There are no data on the effect of cinacalcet on tumor growth in parathyroid cancer. Similarly, there are no data on the use of a combination of cinacalcet and bisphosphonates in parathyroid cancer, the former used to decrease PTH secretion from the cancer, and the latter used to decrease release of calcium from the skeleton. Cinacalcet offers an option for control of intractable hypercalcemia when surgical removal of cancerous tissue is no longer an option.

 

SUMMARY

This chapter has presented a comprehensive summary of the modern-day presentation of PHPT. Typically, an asymptomatic disorder in countries that are economically more developed, the disorder’s presentation has raised issues regarding the extent to which such patients may nevertheless show subclinical target organ involvement, who should be recommended for parathyroid surgery, who can be safely followed without surgical intervention, as well as questions regarding the role of medical therapy. Questions about the natural history and pathophysiology of the disorder continue to be of great interest. Inasmuch as this disorder continues to evolve, it is clear that additional careful studies are required continually to gain new insights into this disease.

 

ACKNOWLEDGEMENTS

This work was supported in part by National Institutes of Health grants NIDDK 32333, DK084986, RR 00645, and R21DK104105. With permission, this chapter is adapted from:

Walker MD, Bilezikian JP. Primary hyperparathyroidism. IN: Endocrinology, 8th edition (Jameson JL, Robertson P, eds) Saunders, Elsevier (in press), 2021.

 

REFERENCES 

  1. Albright F, Bauer W, Claflin D, Cockrill JR. STUDIES IN PARATHYROID PHYSIOLOGY: III. The Effect of Phosphate Ingestion in Clinical Hyperparathyroidism. J Clin Invest 1932; 11:411-435
  2. Albright F, Aub J, Bauer W. Hyperparathyroidism: common and polymorphic condition as illustrated by seventeen proven cases in one clinic. JAMA 1934; 102:1276
  3. Bauer W, Federman DD. Hyperparathyroidism epitomized: the case of Captain Charles E. Martell. Metabolism 1962; 11:21-29
  4. Silverberg S, Bilezikian JP. Clinical presentation of primary hyperparathyroidism in the United States. New York: Academic Press.
  5. Heath H, 3rd, Hodgson SF, Kennedy MA. Primary hyperparathyroidism. Incidence, morbidity, and potential economic impact in a community. N Engl J Med 1980; 302:189-193
  6. Mundy GR, Cove DH, Fisken R. Primary hyperparathyroidism: changes in the pattern of clinical presentation. Lancet 1980; 1:1317-1320
  7. Scholz DA, Purnell DC. Asymptomatic primary hyperparathyroidism. 10-year prospective study. Mayo Clin Proc 1981; 56:473-478
  8. Silverberg SJ, Bilezikian JP. The diagnosis and management of asymptomatic primary hyperparathyroidism. Nat Clin Pract Endocrinol Metab 2006; 2:494-503
  9. Misiorowski W, Czajka-Oraniec I, Kochman M, Zgliczynski W, Bilezikian JP. Osteitis fibrosa cystica-a forgotten radiological feature of primary hyperparathyroidism. Endocrine 2017; 58:380-385
  10. Bendz H, Sjodin I, Toss G, Berglund K. Hyperparathyroidism and long-term lithium therapy--a cross-sectional study and the effect of lithium withdrawal. J Intern Med 1996; 240:357-365
  11. Rao SD, Frame B, Miller MJ, Kleerekoper M, Block MA, Parfitt AM. Hyperparathyroidism following head and neck irradiation. Arch Intern Med 1980; 140:205-207
  12. Nordenstrom J, Strigard K, Perbeck L, Willems J, Bagedahl-Strindlund M, Linder J. Hyperparathyroidism associated with treatment of manic-depressive disorders by lithium. Eur J Surg 1992; 158:207-211
  13. Vaidya A, Curhan GC, Paik JM, Wang M, Taylor EN. Body Size and the Risk of Primary Hyperparathyroidism in Women: A Cohort Study. J Bone Miner Res 2017; 32:1900-1906
  14. Paik JM, Curhan GC, Taylor EN. Calcium intake and risk of primary hyperparathyroidism in women: prospective cohort study. BMJ 2012; 345:e6390
  15. Golden SH, Robinson KA, Saldanha I, Anton B, Ladenson PW. Clinical review: Prevalence and incidence of endocrine and metabolic disorders in the United States: a comprehensive review. J Clin Endocrinol Metab 2009; 94:1853-1878
  16. Attie JN, Bock G, Auguste LJ. Multiple parathyroid adenomas: report of thirty-three cases. Surgery 1990; 108:1014-1019; discussion 1019-1020
  17. Barczynski M, Branstrom R, Dionigi G, Mihai R. Sporadic multiple parathyroid gland disease--a consensus report of the European Society of Endocrine Surgeons (ESES). Langenbecks Arch Surg 2015; 400:887-905
  18. Bilezikian JP, Bandeira L, Khan A, Cusano NE. Hyperparathyroidism. Lancet 2018; 391:168-178
  19. Nudelman IL, Deutsch AA, Reiss R. Primary hyperparathyroidism due to mediastinal parathyroid adenoma. Int Surg 1987; 72:104-108
  20. Sloane JA, Moody HC. Parathyroid adenoma in submucosa of esophagus. Arch Pathol Lab Med 1978; 102:242-243
  21. Joseph MP, Nadol JB, Pilch BZ, Goodman ML. Ectopic parathyroid tissue in the hypopharyngeal mucosa (pyriform sinus). Head Neck Surg 1982; 5:70-74
  22. Gilmour JR. Some developmental abnormalities of the thymus and parathyroids. J Pathol Bacteriol 1941; 52:213-218
  23. Fleischer J, Becker C, Hamele-Bena D, Breen TL, Silverberg SJ. Oxyphil parathyroid adenoma: a malignant presentation of a benign disease. J Clin Endocrinol Metab 2004; 89:5948-5951
  24. Jain M, Krasne DL, Singer FR, Giuliano AE. Recurrent primary hyperparathyroidism due to Type 1 parathyromatosis. Endocrine 2017; 55:643-650
  25. Wermers RA, Khosla S, Atkinson EJ, Hodgson SF, O'Fallon WM, Melton LJ, 3rd. The rise and fall of primary hyperparathyroidism: a population-based study in Rochester, Minnesota, 1965-1992. Ann Intern Med 1997; 126:433-440
  26. Melton LJ, 3rd. The epidemiology of primary hyperparathyroidism in North America. J Bone Miner Res 2002; 17 Suppl 2:N12-17
  27. Wermers RA, Khosla S, Atkinson EJ, Achenbach SJ, Oberg AL, Grant CS, Melton LJ, 3rd. Incidence of primary hyperparathyroidism in Rochester, Minnesota, 1993-2001: an update on the changing epidemiology of the disease. J Bone Miner Res 2006; 21:171-177
  28. Griebeler ML, Kearns AE, Ryu E, Hathcock MA, Melton LJ, 3rd, Wermers RA. Secular trends in the incidence of primary hyperparathyroidism over five decades (1965-2010). Bone 2015; 73:1-7
  29. Yeh MW, Ituarte PH, Zhou HC, Nishimoto S, Liu IL, Harari A, Haigh PI, Adams AL. Incidence and prevalence of primary hyperparathyroidism in a racially mixed population. J Clin Endocrinol Metab 2013; 98:1122-1129
  30. Lowe H, McMahon DJ, Rubin MR, Bilezikian JP, Silverberg SJ. Normocalcemic primary hyperparathyroidism: further characterization of a new clinical phenotype. J Clin Endocrinol Metab 2007; 92:3001-3005
  31. Cusano NE, Maalouf NM, Wang PY, Zhang C, Cremers SC, Haney EM, Bauer DC, Orwoll ES, Bilezikian JP. Normocalcemic hyperparathyroidism and hypoparathyroidism in two community-based nonreferral populations. J Clin Endocrinol Metab 2013; 98:2734-2741
  32. Schini M, Jacques RM, Oakes E, Peel NFA, Walsh JS, Eastell R. Normocalcemic Hyperparathyroidism: Study of its Prevalence and Natural History. J Clin Endocrinol Metab 2020; 105
  33. Jacobs TP, Bilezikian JP. Clinical review: Rare causes of hypercalcemia. J Clin Endocrinol Metab 2005; 90:6316-6322
  34. Eastell R, Arnold A, Brandi ML, Brown EM, D'Amour P, Hanley DA, Rao DS, Rubin MR, Goltzman D, Silverberg SJ, Marx SJ, Peacock M, Mosekilde L, Bouillon R, Lewiecki EM. Diagnosis of asymptomatic primary hyperparathyroidism: proceedings of the third international workshop. J Clin Endocrinol Metab 2009; 94:340-350
  35. Nesbit MA, Hannan FM, Howles SA, Babinsky VN, Head RA, Cranston T, Rust N, Hobbs MR, Heath H, 3rd, Thakker RV. Mutations affecting G-protein subunit alpha11 in hypercalcemia and hypocalcemia. N Engl J Med 2013; 368:2476-2486
  36. Nesbit MA, Hannan FM, Howles SA, Reed AA, Cranston T, Thakker CE, Gregory L, Rimmer AJ, Rust N, Graham U, Morrison PJ, Hunter SJ, Whyte MP, McVean G, Buck D, Thakker RV. Mutations in AP2S1 cause familial hypocalciuric hypercalcemia type 3. Nat Genet 2013; 45:93-97
  37. Souberbielle JC, Cormier C, Kindermans C, Gao P, Cantor T, Forette F, Baulieu EE. Vitamin D status and redefining serum parathyroid hormone reference range in the elderly. J Clin Endocrinol Metab 2001; 86:3086-3090
  38. Walker MD, Cong E, Lee JA, Kepley A, Zhang C, McMahon DJ, Silverberg SJ. Vitamin D in Primary Hyperparathyroidism: Effects on Clinical, Biochemical, and Densitometric Presentation. J Clin Endocrinol Metab 2015; 100:3443-3451
  39. Tassone F, Gianotti L, Emmolo I, Ghio M, Borretta G. Glomerular filtration rate and parathyroid hormone secretion in primary hyperparathyroidism. J Clin Endocrinol Metab 2009; 94:4458-4461
  40. Lepage R, Roy L, Brossard JH, Rousseau L, Dorais C, Lazure C, D'Amour P. A non-(1-84) circulating parathyroid hormone (PTH) fragment interferes significantly with intact PTH commercial assay measurements in uremic samples. Clin Chem 1998; 44:805-809
  41. Quarles LD, Lobaugh B, Murphy G. Intact parathyroid hormone overestimates the presence and severity of parathyroid-mediated osseous abnormalities in uremia. J Clin Endocrinol Metab 1992; 75:145-150
  42. John MR, Goodman WG, Gao P, Cantor TL, Salusky IB, Juppner H. A novel immunoradiometric assay detects full-length human PTH but not amino-terminally truncated fragments: implications for PTH measurements in renal failure. J Clin Endocrinol Metab 1999; 84:4287-4290
  43. Gao P, Scheibel S, D'Amour P, John MR, Rao SD, Schmidt-Gayk H, Cantor TL. Development of a novel immunoradiometric assay exclusively for biologically active whole parathyroid hormone 1-84: implications for improvement of accurate assessment of parathyroid function. J Bone Miner Res 2001; 16:605-614
  44. Slatopolsky E, Finch J, Clay P, Martin D, Sicard G, Singer G, Gao P, Cantor T, Dusso A. A novel mechanism for skeletal resistance in uremia. Kidney Int 2000; 58:753-761
  45. Silverberg SJ, Gao P, Brown I, LoGerfo P, Cantor TL, Bilezikian JP. Clinical utility of an immunoradiometric assay for parathyroid hormone (1-84) in primary hyperparathyroidism. J Clin Endocrinol Metab 2003; 88:4725-4730
  46. D'Amour P, Brossard JH, Rousseau L, Roy L, Gao P, Cantor T. Amino-terminal form of parathyroid hormone (PTH) with immunologic similarities to hPTH(1-84) is overproduced in primary and secondary hyperparathyroidism. Clin Chem 2003; 49:2037-2044
  47. Rubin MR, Silverberg SJ, D'Amour P, Brossard JH, Rousseau L, Sliney J, Jr., Cantor T, Bilezikian JP. An N-terminal molecular form of parathyroid hormone (PTH) distinct from hPTH(1 84) is overproduced in parathyroid carcinoma. Clin Chem 2007; 53:1470-1476
  48. Silverberg SJ, Shane E, Dempster DW, Bilezikian JP. The effects of vitamin D insufficiency in patients with primary hyperparathyroidism. Am J Med 1999; 107:561-567
  49. Hagag P, Revet-Zak I, Hod N, Horne T, Rapoport MJ, Weiss M. Diagnosis of normocalcemic hyperparathyroidism by oral calcium loading test. J Endocrinol Invest 2003; 26:327-332
  50. Silverberg SJ, Bilezikian JP. "Incipient" primary hyperparathyroidism: a "forme fruste" of an old disease. J Clin Endocrinol Metab 2003; 88:5348-5352
  51. Vieth R, Bayley TA, Walfish PG, Rosen IB, Pollard A. Relevance of vitamin D metabolite concentrations in supporting the diagnosis of primary hyperparathyroidism. Surgery 1991; 110:1043-1046; discussion 1046-1047
  52. Silverberg SJ, Shane E, Jacobs TP, Siris E, Bilezikian JP. A 10-year prospective study of primary hyperparathyroidism with or without parathyroid surgery. N Engl J Med 1999; 341:1249-1255
  53. Lumb GA, Stanbury SW. Parathyroid function in human vitamin D deficiency and vitamin D deficiency in primary hyperparathyroidism. Am J Med 1974; 56:833-839
  54. Liu JM, Cusano NE, Silva BC, Zhao L, He XY, Tao B, Sun LH, Zhao HY, Fan WW, Romano ME, Ning G, Bilezikian JP. Primary Hyperparathyroidism: A Tale of Two Cities Revisited - New York and Shanghai. Bone research 2013; 1:162-169
  55. Malabu UH, Founda MA. Primary hyperparathyroidism in Saudi Arabia: a review of 46 cases. The Medical journal of Malaysia 2007; 62:394-397
  56. Paruk IM, Esterhuizen TM, Maharaj S, Pirie FJ, Motala AA. Characteristics, management and outcome of primary hyperparathyroidism in South Africa: a single-centre experience. Postgraduate medical journal 2013; 89:626-631
  57. Shah VN, Bhadada S, Bhansali A, Behera A, Mittal BR. Changes in clinical & biochemical presentations of primary hyperparathyroidism in India over a period of 20 years. Indian J Med Res 2014; 139:694-699
  58. Zhao L, Liu JM, He XY, Zhao HY, Sun LH, Tao B, Zhang MJ, Chen X, Wang WQ, Ning G. The changing clinical patterns of primary hyperparathyroidism in Chinese patients: data from 2000 to 2010 in a single clinical center. J Clin Endocrinol Metab 2013; 98:721-728
  59. Castellano E, Attanasio R, Boriano A, Pellegrino M, Garino F, Gianotti L, Borretta G. Sex Difference in the Clinical Presentation of Primary Hyperparathyroidism: Influence of Menopausal Status. J Clin Endocrinol Metab 2017; 102:4148-4152
  60. Pradeep PV, Mishra A, Agarwal G, Agarwal A, Verma AK, Mishra SK. Long-term outcome after parathyroidectomy in patients with advanced primary hyperparathyroidism and associated vitamin D deficiency. World J Surg 2008; 32:829-835
  61. Bandeira F, Cassibba S. Hyperparathyroidism and Bone Health. Current rheumatology reports 2015; 17:48
  62. Harinarayan CV, Gupta N, Kochupillai N. Vitamin D status in primary hyperparathyroidism in India. Clin Endocrinol (Oxf) 1995; 43:351-358
  63. Meng X, Xing X, Liu S. [The diagnosis of primary hyperparathyroidism--analysis of 134 cases]. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 1994; 16:13-19
  64. Luong KV, Nguyen LT. Coexisting hyperthyroidism and primary hyperparathyroidism with vitamin D-deficient osteomalacia in a Vietnamese immigrant. Endocr Pract 1996; 2:250-254
  65. Usta A, Alhan E, Cinel A, Turkyilmaz S, Erem C. A 20-year study on 190 patients with primary hyperparathyroidism in a developing country: Turkey experience. Int Surg 2015; 100:648-655
  66. Silverberg SJ, Bilezikian JP. Primary Hyperparathyroidism. Primer on Metabolic Bone Diseases and Disorders of Mineral Metbolism. Washington D. C.: American Society for Bone and Mineral Research:302-306.
  67. Silverberg SJ. Non-classical target organs in primary hyperparathyroidism. J Bone Miner Res 2002; 17 Suppl 2:N117-125
  68. Ringe JD. Reversible hypertension in primary hyperparathyroidism--pre- and posteroperative blood pressure in 75 cases. Klin Wochenschr 1984; 62:465-469
  69. Broulik PD, Horky K, Pacovsky V. Blood pressure in patients with primary hyperparathyroidism before and after parathyroidectomy. Exp Clin Endocrinol 1985; 86:346-352
  70. Rapado A. Arterial hypertension and primary hyperparathyroidism. Incidence and follow-up after parathyroidectomy. Am J Nephrol 1986; 6 Suppl 1:49-50
  71. Bilezikian JP, Connor TB, Aptekar R, Freijanes J, Aurbach GD, Pachas WN, Wells SA, Decker JL. Pseudogout after parathyroidectomy. Lancet 1973; 1:445-446
  72. Geelhoed GW, Kelly TR. Pseudogout as a clue and complication in primary hyperparathyroidism. Surgery 1989; 106:1036-1041, discussion 1041-1032
  73. Ludvigsson JF, Kampe O, Lebwohl B, Green PH, Silverberg SJ, Ekbom A. Primary hyperparathyroidism and celiac disease: a population-based cohort study. J Clin Endocrinol Metab 2012; 97:897-904
  74. Silverberg SJ, Bilezikian JP. Primary Hyperparathyroidism. In: Seibel MJ, Robins SP, Bilezikian JP, eds. Dynamics of bone and cartilage metabolism. San Diego: Elsevier:767–778.
  75. Silverberg SJ, Gartenberg F, Jacobs TP, Shane E, Siris E, Staron RB, Bilezikian JP. Longitudinal measurements of bone density and biochemical indices in untreated primary hyperparathyroidism. J Clin Endocrinol Metab 1995; 80:723-728
  76. Seibel MJ. Molecular markers of bone metabolism in primary hyperparathyroidism. In: Bilezikian JP, ed. The parathyroids: basic and clinical concepts. New York: Academic Press:399-410.
  77. Price PA, Parthemore JG, Deftos LJ. New biochemical marker for bone metabolism. Measurement by radioimmunoassay of bone GLA protein in the plasma of normal subjects and patients with bone disease. J Clin Invest 1980; 66:878-883
  78. Deftos LJ, Parthemore JG, Price PA. Changes in plasma bone GLA protein during treatment of bone disease. Calcif Tissue Int 1982; 34:121-124
  79. Ebeling PR, Peterson JM, Riggs BL. Utility of type I procollagen propeptide assays for assessing abnormalities in metabolic bone diseases. J Bone Miner Res 1992; 7:1243-1250
  80. van Lierop AH, Witteveen JE, Hamdy NA, Papapoulos SE. Patients with primary hyperparathyroidism have lower circulating sclerostin levels than euparathyroid controls. Eur J Endocrinol 2010; 163:833-837
  81. Costa AG, Cremers S, Rubin MR, McMahon DJ, Sliney J, Jr., Lazaretti-Castro M, Silverberg SJ, Bilezikian JP. Circulating sclerostin in disorders of parathyroid gland function. J Clin Endocrinol Metab 2011; 96:3804-3810
  82. Ardawi MS, Al-Sibiany AM, Bakhsh TM, Rouzi AA, Qari MH. Decreased serum sclerostin levels in patients with primary hyperparathyroidism: a cross-sectional and a longitudinal study. Osteoporos Int 2011;
  83. Seibel MJ, Gartenberg F, Silverberg SJ, Ratcliffe A, Robins SP, Bilezikian JP. Urinary hydroxypyridinium cross-links of collagen in primary hyperparathyroidism. J Clin Endocrinol Metab 1992; 74:481-486
  84. Seibel MJ, Woitge HW, Pecherstorfer M, Karmatschek M, Horn E, Ludwig H, Armbruster FP, Ziegler R. Serum immunoreactive bone sialoprotein as a new marker of bone turnover in metabolic and malignant bone disease. J Clin Endocrinol Metab 1996; 81:3289-3294
  85. Guo CY, Thomas WE, al-Dehaimi AW, Assiri AM, Eastell R. Longitudinal changes in bone mineral density and bone turnover in postmenopausal women with primary hyperparathyroidism. J Clin Endocrinol Metab 1996; 81:3487-3491
  86. Tanaka Y, Funahashi H, Imai T, Tominaga Y, Takagi H. Parathyroid function and bone metabolic markers in primary and secondary hyperparathyroidism. Semin Surg Oncol 1997; 13:125-133
  87. Minisola S, Romagnoli E, Scarnecchia L, Rosso R, Pacitti MT, Scarda A, Mazzuoli G. Serum carboxy-terminal propeptide of human type I procollagen in patients with primary hyperparathyroidism: studies in basal conditions and after parathyroid surgery. Eur J Endocrinol 1994; 130:587-591
  88. Ohe MN, Bonansea TCP, Santos RO, Neves MCD, Santos LM, Rosano M, Kunii IS, Castro ML, Vieira JGH. Prediction of bone mass changes after successful parathyroidectomy using biochemical markers of bone metabolism in primary hyperparathyroidism: is it clinically useful? Arch Endocrinol Metab 2019; 63:394-401
  89. NIH conference. Diagnosis and management of asymptomatic primary hyperparathyroidism: consensus development conference statement. Ann Intern Med 1991; 114:593-597
  90. Silverberg SJ, Shane E, de la Cruz L, Dempster DW, Feldman F, Seldin D, Jacobs TP, Siris ES, Cafferty M, Parisien MV, et al. Skeletal disease in primary hyperparathyroidism. J Bone Miner Res 1989; 4:283-291
  91. Bilezikian JP, Silverberg SJ, Shane E, Parisien M, Dempster DW. Characterization and evaluation of asymptomatic primary hyperparathyroidism. J Bone Miner Res 1991; 6 Suppl 2:S85-89; discussion S121-124
  92. Dempster DW, Cosman F, Parisien M, Shen V, Lindsay R. Anabolic actions of parathyroid hormone on bone. Endocr Rev 1993; 14:690-709
  93. Bilezikian JP, Rubin MR, Finkelstein JS. Parathyroid hormone as an anabolic therapy for women and men. J Endocrinol Invest 2005; 28:41-49
  94. Canalis E, Giustina A, Bilezikian JP. Mechanisms of anabolic therapies for osteoporosis. N Engl J Med 2007; 357:905-916
  95. Silverberg SJ, Locker FG, Bilezikian JP. Vertebral osteopenia: a new indication for surgery in primary hyperparathyroidism. J Clin Endocrinol Metab 1996; 81:4007-4012
  96. Walker MD, Saeed I, Lee JA, Zhang C, Hans D, Lang T, Silverberg SJ. Effect of concomitant vitamin D deficiency or insufficiency on lumbar spine volumetric bone mineral density and trabecular bone score in primary hyperparathyroidism. Osteoporos Int 2016; 27:3063-3071
  97. Walker MD, Nishiyama KK, Zhou B, Cong E, Wang J, Lee JA, Kepley A, Zhang C, Guo XE, Silverberg SJ. Effect of Low Vitamin D on Volumetric Bone Mineral Density, Bone Microarchitecture, and Stiffness in Primary Hyperparathyroidism. J Clin Endocrinol Metab 2016; 101:905-913
  98. Parisien M, Silverberg SJ, Shane E, de la Cruz L, Lindsay R, Bilezikian JP, Dempster DW. The histomorphometry of bone in primary hyperparathyroidism: preservation of cancellous bone structure. J Clin Endocrinol Metab 1990; 70:930-938
  99. Parfitt AM. Accelerated cortical bone loss: primary and secondary hyperparathyroidism. In: Uhthoff H, Stahl E, eds. Current Concepts in Bone Fragility. New York: Mary Ann Liebert:7-14.
  100. van Doorn L, Lips P, Netelenbos JC, Hackeng WH. Bone histomorphometry and serum concentrations of intact parathyroid hormone (PTH(1-84)) in patients with primary hyperparathyroidism. Bone Miner 1993; 23:233-242
  101. Dempster DW, Parisien M, Silverberg SJ, Liang XG, Schnitzer M, Shen V, Shane E, Kimmel DB, Recker R, Lindsay R, Bilezikian JP. On the mechanism of cancellous bone preservation in postmenopausal women with mild primary hyperparathyroidism. J Clin Endocrinol Metab 1999; 84:1562-1566
  102. Parisien M, Cosman F, Mellish RW, Schnitzer M, Nieves J, Silverberg SJ, Shane E, Kimmel D, Recker RR, Bilezikian JP, et al. Bone structure in postmenopausal hyperparathyroid, osteoporotic, and normal women. J Bone Miner Res 1995; 10:1393-1399
  103. Parisien M, Mellish RW, Silverberg SJ, Shane E, Lindsay R, Bilezikian JP, Dempster DW. Maintenance of cancellous bone connectivity in primary hyperparathyroidism: trabecular strut analysis. J Bone Miner Res 1992; 7:913-919
  104. Lindsay R, Nieves J, Formica C, Henneman E, Woelfert L, Shen V, Dempster D, Cosman F. Randomised controlled study of effect of parathyroid hormone on vertebral-bone mass and fracture incidence among postmenopausal women on oestrogen with osteoporosis. Lancet 1997; 350:550-555
  105. Dempster DW, Cosman F, Kurland ES, Zhou H, Nieves J, Woelfert L, Shane E, Plavetic K, Muller R, Bilezikian J, Lindsay R. Effects of daily treatment with parathyroid hormone on bone microarchitecture and turnover in patients with osteoporosis: a paired biopsy study. J Bone Miner Res 2001; 16:1846-1853
  106. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, Hodsman AB, Eriksen EF, Ish-Shalom S, Genant HK, Wang O, Mitlak BH. Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med 2001; 344:1434-1441
  107. Dempster DW, Muller R, Zhou H, Kohler T, Shane E, Parisien M, Silverberg SJ, Bilezikian JP. Preserved three-dimensional cancellous bone structure in mild primary hyperparathyroidism. Bone 2007; 41:19-24
  108. Roschger P, Dempster DW, Zhou H, Paschalis EP, Silverberg SJ, Shane E, Bilezikian JP, Klaushofer K. New observations on bone quality in mild primary hyperparathyroidism as determined by quantitative backscattered electron imaging. J Bone Miner Res 2007; 22:717-723
  109. Zoehrer R, Dempster DW, Bilezikian JP, Zhou H, Silverberg SJ, Shane E, Roschger P, Paschalis EP, Klaushofer K. Bone quality determined by Fourier transform infrared imaging analysis in mild primary hyperparathyroidism. J Clin Endocrinol Metab 2008; 93:3484-3489
  110. Bilezikian JP. Bone strength in primary hyperparathyroidism. Osteoporos Int 2003; 14 Suppl 5:S113-115; discussion S115-117
  111. Hans D, Goertzen AL, Krieg MA, Leslie WD. Bone microarchitecture assessed by TBS predicts osteoporotic fractures independent of bone density: the Manitoba study. J Bone Miner Res 2011; 26:2762-2769
  112. Silva BC, Boutroy S, Zhang C, McMahon DJ, Zhou B, Wang J, Udesky J, Cremers S, Sarquis M, Guo XD, Hans D, Bilezikian JP. Trabecular bone score (TBS)--a novel method to evaluate bone microarchitectural texture in patients with primary hyperparathyroidism. J Clin Endocrinol Metab 2013; 98:1963-1970
  113. Stein EM, Silva BC, Boutroy S, Zhou B, Wang J, Udesky J, Zhang C, McMahon DJ, Romano M, Dworakowski E, Costa AG, Cusano N, Irani D, Cremers S, Shane E, Guo XE, Bilezikian JP. Primary hyperparathyroidism is associated with abnormal cortical and trabecular microstructure and reduced bone stiffness in postmenopausal women. J Bone Miner Res 2013; 28:1029-1040
  114. Hansen S, Hauge EM, Rasmussen L, Jensen JE, Brixen K. Parathyroidectomy improves bone geometry and microarchitecture in female patients with primary hyperparathyroidism. A 1-year prospective controlled study using high resolution peripheral quantitative computed tomography. J Bone Miner Res 2012;
  115. Vu TD, Wang XF, Wang Q, Cusano NE, Irani D, Silva BC, Ghasem-Zadeh A, Udesky J, Romano ME, Zebaze R, Jerums G, Boutroy S, Bilezikian JP, Seeman E. New insights into the effects of primary hyperparathyroidism on the cortical and trabecular compartments of bone. Bone 2013; 55:57-63
  116. Stein EM, Silva BC, Boutroy S, Zhou B, Wang J, Udesky J, Zhang C, McMahon DJ, Romano M, Dworakowski E, Costa AG, Cusano N, Irani D, Cremers S, Shane E, Guo XE, Bilezikian JP. Primary hyperparathyroidism is associated with abnormal cortical and trabecular microstructure and reduced bone stiffness in postmenopausal women. J Bone Miner Res 2012;
  117. Khosla S, Melton LJ, 3rd, Wermers RA, Crowson CS, O'Fallon W, Riggs B. Primary hyperparathyroidism and the risk of fracture: a population-based study. J Bone Miner Res 1999; 14:1700-1707
  118. Vignali E, Viccica G, Diacinti D, Cetani F, Cianferotti L, Ambrogini E, Banti C, Del Fiacco R, Bilezikian JP, Pinchera A, Marcocci C. Morphometric vertebral fractures in postmenopausal women with primary hyperparathyroidism. J Clin Endocrinol Metab 2009; 94:2306-2312
  119. Mosekilde L. Primary hyperparathyroidism and the skeleton. Clin Endocrinol (Oxf) 2008; 69:1-19
  120. De Geronimo S, Romagnoli E, Diacinti D, D'Erasmo E, Minisola S. The risk of fractures in postmenopausal women with primary hyperparathyroidism. Eur J Endocrinol 2006; 155:415-420
  121. Dauphine RT, Riggs BL, Scholz DA. Back pain and vertebral crush fractures: an unemphasized mode of presentation for primary hyperparathyroidism. Ann Intern Med 1975; 83:365-367
  122. Kochersberger G, Buckley NJ, Leight GS, Martinez S, Studenski S, Vogler J, Lyles KW. What is the clinical significance of bone loss in primary hyperparathyroidism? Arch Intern Med 1987; 147:1951-1953
  123. Wilson RJ, Rao S, Ellis B, Kleerekoper M, Parfitt AM. Mild asymptomatic primary hyperparathyroidism is not a risk factor for vertebral fractures. Ann Intern Med 1988; 109:959-962
  124. Liu M, Williams J, Kuo J, Lee JA, Silverberg SJ, Walker MD. Risk factors for vertebral fracture in primary hyperparathyroidism. Endocrine 2019; 66:682-690
  125. Ejlsmark-Svensson H, Bislev LS, Lajlev S, Harslof T, Rolighed L, Sikjaer T, Rejnmark L. Prevalence and Risk of Vertebral Fractures in Primary Hyperparathyroidism: A Nested Case-Control Study. J Bone Miner Res 2018; 33:1657-1664
  126. Larsson K, Ljunghall S, Krusemo UB, Naessen T, Lindh E, Persson I. The risk of hip fractures in patients with primary hyperparathyroidism: a population-based cohort study with a follow-up of 19 years. J Intern Med 1993; 234:585-593
  127. Silverberg SJ, Shane E, Jacobs TP, Siris ES, Gartenberg F, Seldin D, Clemens TL, Bilezikian JP. Nephrolithiasis and bone involvement in primary hyperparathyroidism. Am J Med 1990; 89:327-334
  128. Pak CY, Oata M, Lawrence EC, Snyder W. The hypercalciurias. Causes, parathyroid functions, and diagnostic criteria. J Clin Invest 1974; 54:387-400
  129. Kaplan RA, Haussler MR, Deftos LJ, Bone H, Pak CY. The role of 1 alpha, 25-dihydroxyvitamin D in the mediation of intestinal hyperabsorption of calcium in primary hyperparathyroidism and absorptive hypercalciuria. J Clin Invest 1977; 59:756-760
  130. Broadus AE, Horst RL, Lang R, Littledike ET, Rasmussen H. The importance of circulating 1,25-dihydroxyvitamin D in the pathogenesis of hypercalciuria and renal-stone formation in primary hyperparathyroidism. N Engl J Med 1980; 302:421-426
  131. Pak CY, Nicar MJ, Peterson R, Zerwekh JE, Snyder W. A lack of unique pathophysiologic background for nephrolithiasis of primary hyperparathyroidism. J Clin Endocrinol Metab 1981; 53:536-542
  132. Pak CY. Effect of parathyroidectomy on crystallization of calcium salts in urine of patients with primary hyperparathyroidism. Invest Urol 1979; 17:146-148
  133. Klugman VA, Favus M, Pak CY. Nephrolithiasis in primary hyperparathyroidism. In: Bilezikian JP, ed. The parathyroids: basic and clinical concepts. New York: Academic Press:437-450.
  134. Cipriani C, Biamonte F, Costa AG, Zhang C, Biondi P, Diacinti D, Pepe J, Piemonte S, Scillitani A, Minisola S, Bilezikian JP. Prevalence of kidney stones and vertebral fractures in primary hyperparathyroidism using imaging technology. J Clin Endocrinol Metab 2015; 100:1309-1315
  135. Rejnmark L, Vestergaard P, Mosekilde L. Nephrolithiasis and renal calcifications in primary hyperparathyroidism. J Clin Endocrinol Metab 2011; 96:2377-2385
  136. Cassibba S, Pellegrino M, Gianotti L, Baffoni C, Baralis E, Attanasio R, Guarnieri A, Borretta G, Tassone F. Silent renal stones in primary hyperparathyroidism: prevalence and clinical features. Endocr Pract 2014; 20:1137-1142
  137. Bilezikian JP, Brandi ML, Eastell R, Silverberg SJ, Udelsman R, Marcocci C, Potts JT, Jr. Guidelines for the management of asymptomatic primary hyperparathyroidism: summary statement from the Fourth International Workshop. J Clin Endocrinol Metab 2014; 99:3561-3569
  138. Tay YD, Liu M, Bandeira L, Bucovsky M, Lee JA, Silverberg SJ, Walker MD. Occult urolithiasis in asymptomatic primary hyperparathyroidism. Endocr Res 2018; 43:106-115
  139. Albright F, Reifenstein EC. The parathyroid glands and metabolic bone disease. Baltimore: Williams and Wilkins.
  140. Dent CE, Hartland BV, Hicks J, Sykes ED. Calcium intake in patients with primary hyperparathyroidism. Lancet 1961; 2:336-338
  141. Berger AD, Wu W, Eisner BH, Cooperberg MR, Duh QY, Stoller ML. Patients with primary hyperparathyroidism--why do some form stones? J Urol 2009; 181:2141-2145
  142. Odvina CV, Sakhaee K, Heller HJ, Peterson RD, Poindexter JR, Padalino PK, Pak CY. Biochemical characterization of primary hyperparathyroidism with and without kidney stones. Urological research 2007; 35:123-128
  143. Saponaro F, Cetani F, Mazoni L, Apicella M, Di Giulio M, Carlucci F, Scalese M, Pardi E, Borsari S, Bilezikian JP, Marcocci C. Hypercalciuria: its value as a predictive risk factor for nephrolithiasis in asymptomatic primary hyperparathyroidism? J Endocrinol Invest 2020; 43:677-682
  144. Saponaro F, Marcocci C, Apicella M, Mazoni L, Borsari S, Pardi E, Di Giulio M, Carlucci F, Scalese M, Bilezikian JP, Cetani F. Hypomagnesuria is Associated With Nephrolithiasis in Patients With Asymptomatic Primary Hyperparathyroidism. J Clin Endocrinol Metab 2020; 105
  145. Walker MD, Nickolas T, Kepley A, Lee JA, Zhang C, McMahon DJ, Silverberg SJ. Predictors of renal function in primary hyperparathyroidism. J Clin Endocrinol Metab 2014; 99:1885-1892
  146. Walker MD, Dempster DW, McMahon DJ, Udesky J, Shane E, Bilezikian JP, Silverberg SJ. Effect of renal function on skeletal health in primary hyperparathyroidism. J Clin Endocrinol Metab 2012; 97:1501-1507
  147. Rao DS, Wilson RJ, Kleerekoper M, Parfitt AM. Lack of biochemical progression or continuation of accelerated bone loss in mild asymptomatic primary hyperparathyroidism: evidence for biphasic disease course. J Clin Endocrinol Metab 1988; 67:1294-1298
  148. Symons C, Fortune F, Greenbaum RA, Dandona P. Cardiac hypertrophy, hypertrophic cardiomyopathy, and hyperparathyroidism--an association. Br Heart J 1985; 54:539-542
  149. Stefenelli T, Mayr H, Bergler-Klein J, Globits S, Woloszczuk W, Niederle B. Primary hyperparathyroidism: incidence of cardiac abnormalities and partial reversibility after successful parathyroidectomy. Am J Med 1993; 95:197-202
  150. Palmer M, Adami HO, Bergstrom R, Akerstrom G, Ljunghall S. Mortality after surgery for primary hyperparathyroidism: a follow-up of 441 patients operated on from 1956 to 1979. Surgery 1987; 102:1-7
  151. Ronni-Sivula H. Causes of death in patients previously operated on for primary hyperparathyroidism. Ann Chir Gynaecol 1985; 74:13-18
  152. Hedback G, Tisell LE, Bengtsson BA, Hedman I, Oden A. Premature death in patients operated on for primary hyperparathyroidism. World J Surg 1990; 14:829-835; discussion 836
  153. Ljunghall S, Jakobsson S, Joborn C, Palmer M, Rastad J, Akerstrom G. Longitudinal studies of mild primary hyperparathyroidism. J Bone Miner Res 1991; 6 Suppl 2:S111-116; discussion S121-114
  154. Hedback G, Oden A, Tisell LE. The influence of surgery on the risk of death in patients with primary hyperparathyroidism. World J Surg 1991; 15:399-405; discussion 406-397
  155. Soreide JA, van Heerden JA, Grant CS, Yau Lo C, Schleck C, Ilstrup DM. Survival after surgical treatment for primary hyperparathyroidism. Surgery 1997; 122:1117-1123
  156. Wermers RA, Khosla S, Atkinson EJ, Grant CS, Hodgson SF, O'Fallon WM, Melton LJ, 3rd. Survival after the diagnosis of hyperparathyroidism: a population-based study. Am J Med 1998; 104:115-122
  157. Nilsson IL, Yin L, Lundgren E, Rastad J, Ekbom A. Clinical presentation of primary hyperparathyroidism in Europe--nationwide cohort analysis on mortality from nonmalignant causes. J Bone Miner Res 2002; 17 Suppl 2:N68-74
  158. Hedback G, Oden A. Increased risk of death from primary hyperparathyroidism--an update. Eur J Clin Invest 1998; 28:271-276
  159. Vestergaard P, Mollerup CL, Frokjaer VG, Christiansen P, Blichert-Toft M, Mosekilde L. Cardiovascular events before and after surgery for primary hyperparathyroidism. World J Surg 2003; 27:216-222
  160. Bradley EL, 3rd, Wells JO. Primary hyperparathyroidism and hypertension. Am Surg 1983; 49:569-570
  161. Dominiczak AF, Lyall F, Morton JJ, Dargie HJ, Boyle IT, Tune TT, Murray G, Semple PF. Blood pressure, left ventricular mass and intracellular calcium in primary hyperparathyroidism. Clin Sci (Lond) 1990; 78:127-132
  162. Nainby-Luxmoore JC, Langford HG, Nelson NC, Watson RL, Barnes TY. A case-comparison study of hypertension and hyperparathyroidism. J Clin Endocrinol Metab 1982; 55:303-306
  163. Lind L, Jacobsson S, Palmer M, Lithell H, Wengle B, Ljunghall S. Cardiovascular risk factors in primary hyperparathyroidism: a 15-year follow-up of operated and unoperated cases. J Intern Med 1991; 230:29-35
  164. Lind L, Skarfors E, Berglund L, Lithell H, Ljunghall S. Serum calcium: a new, independent, prospective risk factor for myocardial infarction in middle-aged men followed for 18 years. J Clin Epidemiol 1997; 50:967-973
  165. Kamycheva E, Sundsfjord J, Jorde R. Serum parathyroid hormone level is associated with body mass index. The 5th Tromso study. Eur J Endocrinol 2004; 151:167-172
  166. Roberts WC, Waller BF. Effect of chronic hypercalcemia on the heart. An analysis of 18 necropsy patients. Am J Med 1981; 71:371-384
  167. Streeten EA, Munir K, Hines S, Mohamed A, Mangano C, Ryan KA, Post W. Coronary artery calcification in patients with primary hyperparathyroidism in comparison with control subjects from the multi-ethnic study of atherosclerosis. Endocr Pract 2008; 14:155-161
  168. Kepez A, Harmanci A, Hazirolan T, Isildak M, Kocabas U, Ates A, Ciftci O, Tokgozoglu L, Gurlek A. Evaluation of subclinical coronary atherosclerosis in mild asymptomatic primary hyperparathyroidism patients. Int J Cardiovasc Imaging 2009; 25:187-193
  169. Nilsson IL, Aberg J, Rastad J, Lind L. Maintained normalization of cardiovascular dysfunction 5 years after parathyroidectomy in primary hyperparathyroidism. Surgery 2005; 137:632-638
  170. Iwata S, Walker MD, Di Tullio MR, Hyodo E, Jin Z, Liu R, Sacco RL, Homma S, Silverberg SJ. Aortic valve calcification in mild primary hyperparathyroidism. J Clin Endocrinol Metab 2012; 97:132-137
  171. Walker MD, Rundek T, Homma S, DiTullio M, Iwata S, Lee JA, Choi J, Liu R, Zhang C, McMahon DJ, Sacco RL, Silverberg SJ. Effect of parathyroidectomy on subclinical cardiovascular disease in mild primary hyperparathyroidism. Eur J Endocrinol 2012; 167:277-285
  172. Nuzzo V, Tauchmanova L, Fonderico F, Trotta R, Fittipaldi MR, Fontana D, Rossi R, Lombardi G, Trimarco B, Lupoli G. Increased intima-media thickness of the carotid artery wall, normal blood pressure profile and normal left ventricular mass in subjects with primary hyperparathyroidism. Eur J Endocrinol 2002; 147:453-459
  173. Nilsson IL, Aberg J, Rastad J, Lind L. Left ventricular systolic and diastolic function and exercise testing in primary hyperparathyroidism-effects of parathyroidectomy. Surgery 2000; 128:895-902
  174. Walker MD, Silverberg SJ. Cardiovascular aspects of primary hyperparathyroidism. J Endocrinol Invest 2008; 31:925-931
  175. McMahon DJ, Carrelli A, Palmeri N, Zhang C, DiTullio M, Silverberg SJ, Walker MD. Effect of Parathyroidectomy Upon Left Ventricular Mass in Primary Hyperparathyroidism: A Meta-Analysis. J Clin Endocrinol Metab 2015; 100:4399-4407
  176. Nilsson IL, Aberg J, Rastad J, Lind L. Endothelial vasodilatory dysfunction in primary hyperparathyroidism is reversed after parathyroidectomy. Surgery 1999; 126:1049-1055
  177. Kosch M, Hausberg M, Vormbrock K, Kisters K, Rahn KH, Barenbrock M. Studies on flow-mediated vasodilation and intima-media thickness of the brachial artery in patients with primary hyperparathyroidism. Am J Hypertens 2000; 13:759-764
  178. Lumachi F, Ermani M, Frego M, Pilon F, Filosa T, Di Cristofaro L, De Lotto F, Fallo F. Intima-media thickness measurement of the carotid artery in patients with primary hyperparathyroidism. A prospective case-control study and long-term follow-up. In Vivo 2006; 20:887-890
  179. Fallo F, Camporese G, Capitelli E, Andreozzi GM, Mantero F, Lumachi F. Ultrasound evaluation of carotid artery in primary hyperparathyroidism. J Clin Endocrinol Metab 2003; 88:2096-2099
  180. Walker MD, Fleischer J, Rundek T, McMahon DJ, Homma S, Sacco R, Silverberg SJ. Carotid vascular abnormalities in primary hyperparathyroidism. J Clin Endocrinol Metab 2009; 94:3849-3856
  181. Rosa J, Raska I, Jr., Wichterle D, Petrak O, Strauch B, Somloova Z, Zelinka T, Holaj R, Widimsky J, Jr. Pulse wave velocity in primary hyperparathyroidism and effect of surgical therapy. Hypertens Res 2011; 34:296-300
  182. Schillaci G, Pucci G, Pirro M, Monacelli M, Scarponi AM, Manfredelli MR, Rondelli F, Avenia N, Mannarino E. Large-artery stiffness: a reversible marker of cardiovascular risk in primary hyperparathyroidism. Atherosclerosis 2011; 218:96-101
  183. Rubin MR, Maurer MS, McMahon DJ, Bilezikian JP, Silverberg SJ. Arterial stiffness in mild primary hyperparathyroidism. J Clin Endocrinol Metab 2005; 90:3326-3330
  184. Cope O. The study of hyperparathyroidism at the Massachusetts General Hospital. N Engl J Med 1966; 274:1174-1182
  185. Patten BM, Bilezikian JP, Mallette LE, Prince A, Engel WK, Aurbach GD. Neuromuscular disease in primary hyperparathyroidism. Ann Intern Med 1974; 80:182-193
  186. Vicale CT. Diagnostic features of muscular syndrome resulting from hyperparathyroidism, osteomalacia owing to renal tubular acidosis and perhaps related disorders of calcium metabolism. Trans Am Neurol Assoc 1949; 74:143-147
  187. Frame B, Heinze EG, Jr., Block MA, Manson GA. Myopathy in primary hyperparathyroidism. Observations in three patients. Ann Intern Med 1968; 68:1022-1027
  188. Rollinson RD, Gilligan BS. Primary hyperparathyroidism presenting as a proximal myopathy. Aust N Z J Med 1977; 7:420-421
  189. Turken SA, Cafferty M, Silverberg SJ, De La Cruz L, Cimino C, Lange DJ, Lovelace RE, Bilezikian JP. Neuromuscular involvement in mild, asymptomatic primary hyperparathyroidism. Am J Med 1989; 87:553-557
  190. Joborn C, Hetta J, Johansson H, Rastad J, Agren H, Akerstrom G, Ljunghall S. Psychiatric morbidity in primary hyperparathyroidism. World J Surg 1988; 12:476-481
  191. Walker MD, Silverberg SJ. Parathyroidectomy in asymptomatic primary hyperparathyroidism: improves "bones" but not "psychic moans". J Clin Endocrinol Metab 2007; 92:1613-1615
  192. Bollerslev J, Jansson S, Mollerup CL, Nordenstrom J, Lundgren E, Torring O, Varhaug JE, Baranowski M, Aanderud S, Franco C, Freyschuss B, Isaksen GA, Ueland T, Rosen T. Medical observation, compared with parathyroidectomy, for asymptomatic primary hyperparathyroidism: a prospective, randomized trial. J Clin Endocrinol Metab 2007; 92:1687-1692
  193. Talpos GB, Bone HG, 3rd, Kleerekoper M, Phillips ER, Alam M, Honasoge M, Divine GW, Rao DS. Randomized trial of parathyroidectomy in mild asymptomatic primary hyperparathyroidism: patient description and effects on the SF-36 health survey. Surgery 2000; 128:1013-1020;discussion 1020-1011
  194. Ambrogini E, Cetani F, Cianferotti L, Vignali E, Banti C, Viccica G, Oppo A, Miccoli P, Berti P, Bilezikian JP, Pinchera A, Marcocci C. Surgery or surveillance for mild asymptomatic primary hyperparathyroidism: a prospective, randomized clinical trial. J Clin Endocrinol Metab 2007; 92:3114-3121
  195. Perrier ND, Balachandran D, Wefel JS, Jimenez C, Busaidy N, Morris GS, Dong W, Jackson E, Weaver S, Gantela S, Evans DB, Grubbs EG, Lee JE. Prospective, randomized, controlled trial of parathyroidectomy versus observation in patients with "asymptomatic" primary hyperparathyroidism. Surgery 2009; 146:1116-1122
  196. Walker MD, McMahon DJ, Inabnet WB, Lazar RM, Brown I, Vardy S, Cosman F, Silverberg SJ. Neuropsychological features in primary hyperparathyroidism: a prospective study. J Clin Endocrinol Metab 2009; 94:1951-1958
  197. Roman SA, Sosa JA, Mayes L, Desmond E, Boudourakis L, Lin R, Snyder PJ, Holt E, Udelsman R. Parathyroidectomy improves neurocognitive deficits in patients with primary hyperparathyroidism. Surgery 2005; 138:1121-1128; discussion 1128-1129
  198. Benge JF, Perrier ND, Massman PJ, Meyers CA, Kayl AE, Wefel JS. Cognitive and affective sequelae of primary hyperparathyroidism and early response to parathyroidectomy. J Int Neuropsychol Soc 2009; 15:1002-1011
  199. Chiang CY, Andrewes DG, Anderson D, Devere M, Schweitzer I, Zajac JD. A controlled, prospective study of neuropsychological outcomes post parathyroidectomy in primary hyperparathyroid patients. Clin Endocrinol (Oxf) 2005; 62:99-104
  200. Numann PJ, Torppa AJ, Blumetti AE. Neuropsychologic deficits associated with primary hyperparathyroidism. Surgery 1984; 96:1119-1123
  201. Babinska D, Barczynski M, Stefaniak T, Oseka T, Babinska A, Babinski D, Sworczak K, Lachinski AJ, Nowak W, Sledzinski Z. Evaluation of selected cognitive functions before and after surgery for primary hyperparathyroidism. Langenbecks Arch Surg 2012; 397:825-831
  202. Lourida I, Thompson-Coon J, Dickens CM, Soni M, Kuzma E, Kos K, Llewellyn DJ. Parathyroid hormone, cognitive function and dementia: a systematic review. PLoS One 2015; 10:e0127574
  203. Dotzenrath CM, Kaetsch AK, Pfingsten H, Cupisti K, Weyerbrock N, Vossough A, Verde PE, Ohmann C. Neuropsychiatric and cognitive changes after surgery for primary hyperparathyroidism. World J Surg 2006; 30:680-685
  204. Goyal A, Chumber S, Tandon N, Lal R, Srivastava A, Gupta S. Neuropsychiatric manifestations in patients of primary hyperparathyroidism and outcome following surgery. Indian J Med Sci 2001; 55:677-686
  205. Liu M, Sum M, Cong E, Colon I, Bucovsky M, Williams J, Kepley A, Kuo J, Lee JA, Lazar RM, Marshall R, Silverberg S, Walker MD. Cognition and cerebrovascular function in primary hyperparathyroidism before and after parathyroidectomy. J Endocrinol Invest 2020; 43:369-379
  206. Gazes Y, Liu M, Sum M, Cong E, Kuo J, Lee JA, Silverberg S, Stern Y, Walker M. Functional magnetic resonance imaging in primary hyperparathyroidism. Eur J Endocrinol 2020; 183:21-30
  207. Marx S. Multiple endocrine neoplasia type 1. In: Bilezikian JP, ed. The parathyroids. New York: Academic Press:535-584.
  208. Brandi ML, Gagel RF, Angeli A, Bilezikian JP, Beck-Peccoz P, Bordi C, Conte-Devolx B, Falchetti A, Gheri RG, Libroia A, Lips CJ, Lombardi G, Mannelli M, Pacini F, Ponder BA, Raue F, Skogseid B, Tamburrano G, Thakker RV, Thompson NW, Tomassetti P, Tonelli F, Wells SA, Jr., Marx SJ. Guidelines for diagnosis and therapy of MEN type 1 and type 2. J Clin Endocrinol Metab 2001; 86:5658-5671
  209. Khoo TK, Vege SS, Abu-Lebdeh HS, Ryu E, Nadeem S, Wermers RA. Acute pancreatitis in primary hyperparathyroidism: a population-based study. J Clin Endocrinol Metab 2009; 94:2115-2118
  210. Marx SJ, Fraser D, Rapoport A. Familial hypocalciuric hypercalcemia. Mild expression of the gene in heterozygotes and severe expression in homozygotes. Am J Med 1985; 78:15-22
  211. Kristoffersson A, Dahlgren S, Lithner F, Jarhult J. Primary hyperparathyroidism in pregnancy. Surgery 1985; 97:326-330
  212. Lowe DK, Orwoll ES, McClung MR, Cawthon ML, Peterson CG. Hyperparathyroidism and pregnancy. Am J Surg 1983; 145:611-619
  213. Abood A, Vestergaard P. Pregnancy outcomes in women with primary hyperparathyroidism. Eur J Endocrinol 2014; 171:69-76
  214. Fitzpatrick LA, Bilezikian JP. Acute primary hyperparathyroidism. Am J Med 1987; 82:275-282
  215. Bayat-Mokhtari F, Palmieri GM, Moinuddin M, Pourmand R. Parathyroid storm. Arch Intern Med 1980; 140:1092-1095
  216. Marcocci C, Cetani F, Rubin MR, Silverberg SJ, Pinchera A, Bilezikian JP. Parathyroid carcinoma. J Bone Miner Res 2008; 23:1869-1880
  217. Streeten EA, Weinstein LS, Norton JA, Mulvihill JJ, White BJ, Friedman E, Jaffe G, Brandi ML, Stewart K, Zimering MB, et al. Studies in a kindred with parathyroid carcinoma. J Clin Endocrinol Metab 1992; 75:362-366
  218. Wassif WS, Moniz CF, Friedman E, Wong S, Weber G, Nordenskjold M, Peters TJ, Larsson C. Familial isolated hyperparathyroidism: a distinct genetic entity with an increased risk of parathyroid cancer. J Clin Endocrinol Metab 1993; 77:1485-1489
  219. Yoshimoto K, Endo H, Tsuyuguchi M, Tanaka C, Kimura T, Iwahana H, Kato G, Sano T, Itakura M. Familial isolated primary hyperparathyroidism with parathyroid carcinomas: clinical and molecular features. Clin Endocrinol (Oxf) 1998; 48:67-72
  220. Marx SJ, F. SW, K. AS. Hyperparathyroidism in hereditary syndromes: special expressions and special managements. Journal of Bone and Mineral Research 2002; 17:N37-N43
  221. Chen JD, Morrison C, Zhang C, Kahnoski K, Carpten JD, Teh BT. Hyperparathyroidism-jaw tumour syndrome. J Intern Med 2003; 253:634-642
  222. Mallette LE, Malini S, Rappaport MP, Kirkland JL. Familial cystic parathyroid adenomatosis. Ann Intern Med 1987; 107:54-60
  223. Carpten JD, Robbins CM, Villablanca A, Forsberg L, Presciuttini S, Bailey-Wilson J, Simonds WF, Gillanders EM, Kennedy AM, Chen JD, Agarwal SK, Sood R, Jones MP, Moses TY, Haven C, Petillo D, Leotlela PD, Harding B, Cameron D, Pannett AA, Hoog A, Heath H, 3rd, James-Newton LA, Robinson B, Zarbo RJ, Cavaco BM, Wassif W, Perrier ND, Rosen IB, Kristoffersson U, Turnpenny PD, Farnebo LO, Besser GM, Jackson CE, Morreau H, Trent JM, Thakker RV, Marx SJ, Teh BT, Larsson C, Hobbs MR. HRPT2, encoding parafibromin, is mutated in hyperparathyroidism-jaw tumor syndrome. Nat Genet 2002; 32:676-680
  224. Simonds WF, James-Newton LA, Agarwal SK, Yang B, Skarulis MC, Hendy GN, Marx SJ. Familial isolated hyperparathyroidism: clinical and genetic characteristics of 36 kindreds. Medicine (Baltimore) 2002; 81:1-26
  225. Dionisi S, Minisola S, Pepe J, De Geronimo S, Paglia F, Memeo L, Fitzpatrick LA. Concurrent parathyroid adenomas and carcinoma in the setting of multiple endocrine neoplasia type 1: presentation as hypercalcemic crisis. Mayo Clin Proc 2002; 77:866-869
  226. Agha A, Carpenter R, Bhattacharya S, Edmonson SJ, Carlsen E, Monson JP. Parathyroid carcinoma in multiple endocrine neoplasia type 1 (MEN1) syndrome: two case reports of an unrecognised entity. J Endocrinol Invest 2007; 30:145-149
  227. Haven CJ, van Puijenbroek M, Tan MH, Teh BT, Fleuren GJ, van Wezel T, Morreau H. Identification of MEN1 and HRPT2 somatic mutations in paraffin-embedded (sporadic) parathyroid carcinomas. Clin Endocrinol (Oxf) 2007; 67:370-376
  228. Jenkins PJ, Satta MA, Simmgen M, Drake WM, Williamson C, Lowe DG, Britton K, Chew SL, Thakker RV, Besser GM. Metastatic parathyroid carcinoma in the MEN2A syndrome. Clin Endocrinol (Oxf) 1997; 47:747-751
  229. Cryns VL, Thor A, Xu HJ, Hu SX, Wierman ME, Vickery AL, Jr., Benedict WF, Arnold A. Loss of the retinoblastoma tumor-suppressor gene in parathyroid carcinoma. N Engl J Med 1994; 330:757-761
  230. Dotzenrath C, Teh BT, Farnebo F, Cupisti K, Svensson A, Toell A, Goretzki P, Larsson C. Allelic loss of the retinoblastoma tumor suppressor gene: a marker for aggressive parathyroid tumors? J Clin Endocrinol Metab 1996; 81:3194-3196
  231. Shattuck TM, Valimaki S, Obara T, Gaz RD, Clark OH, Shoback D, Wierman ME, Tojo K, Robbins CM, Carpten JD, Farnebo LO, Larsson C, Arnold A. Somatic and germ-line mutations of the HRPT2 gene in sporadic parathyroid carcinoma. N Engl J Med 2003; 349:1722-1729
  232. Weinstein LS, Simonds WF. HRPT2, a marker of parathyroid cancer. N Engl J Med 2003; 349:1691-1692
  233. LiVolsi V. Morphology of the parathyroid glands. In: Becker KL, ed. Principles and Practice of Endocrinology and Metabolism. Philadelphia: Lipincott Williams and Wilkins.
  234. Palmer JA, Brown WA, Kerr WH, Rosen IB, Watters NA. The surgical aspects of hyperparathyroidism. Arch Surg 1975; 110:1004-1007
  235. Hage MP, Salti I, El-Hajj Fuleihan G. Parathyromatosis: a rare yet problematic etiology of recurrent and persistent hyperparathyroidism. Metabolism 2012; 61:762-775
  236. Baloch ZW, LiVolsi VA. Pathology of the parathyroid glands in hyperparathyroidism. Semin Diagn Pathol 2013; 30:165-177
  237. Reddick RL, Costa JC, Marx SJ. Parathyroid hyperplasia and parathyromatosis. Lancet 1977; 1:549
  238. Pinnamaneni N, Shankar PR, Muthukrishnan A. (99m)Tc MIBI SPECT findings in parathyromatosis--a rare entity causing recurrent hyperparathyroidism. Clin Nucl Med 2013; 38:e443-445
  239. Tublin ME, Yim JH, Carty SE. Recurrent hyperparathyroidism secondary to parathyromatosis: clinical and imaging findings. J Ultrasound Med 2007; 26:847-851
  240. Kollmorgen CF, Aust MR, Ferreiro JA, McCarthy JT, van Heerden JA. Parathyromatosis: a rare yet important cause of persistent or recurrent hyperparathyroidism. Surgery 1994; 116:111-115
  241. Rubin MR, Bilezikian JP, McMahon DJ, Jacobs T, Shane E, Siris E, Udesky J, Silverberg SJ. The natural history of primary hyperparathyroidism with or without parathyroid surgery after 15 years. J Clin Endocrinol Metab 2008; 93:3462-3470
  242. Rao DS, Phillips ER, Divine GW, Talpos GB. Randomized controlled clinical trial of surgery versus no surgery in patients with mild asymptomatic primary hyperparathyroidism. J Clin Endocrinol Metab 2004; 89:5415-5422
  243. Lundstam K, Heck A, Mollerup C, Godang K, Baranowski M, Pernow Y, Varhaug JE, Hessman O, Rosen T, Nordenstrom J, Jansson S, Hellstrom M, Bollerslev J, Group SS. Effects of parathyroidectomy versus observation on the development of vertebral fractures in mild primary hyperparathyroidism. J Clin Endocrinol Metab 2015; 100:1359-1367
  244. Bilezikian JP, Potts JT, Jr., Fuleihan Gel H, Kleerekoper M, Neer R, Peacock M, Rastad J, Silverberg SJ, Udelsman R, Wells SA. Summary statement from a workshop on asymptomatic primary hyperparathyroidism: a perspective for the 21st century. J Clin Endocrinol Metab 2002; 87:5353-5361
  245. Khan AA, Bilezikian JP, Potts JT, Jr. The diagnosis and management of asymptomatic primary hyperparathyroidism revisited. J Clin Endocrinol Metab 2009; 94:333-334
  246. Bilezikian JP, Khan AA, Potts JT, Jr. Guidelines for the management of asymptomatic primary hyperparathyroidism: summary statement from the third international workshop. J Clin Endocrinol Metab 2009; 94:335-339
  247. Silverberg SJ, Clarke BL, Peacock M, Bandeira F, Boutroy S, Cusano NE, Dempster D, Lewiecki EM, Liu JM, Minisola S, Rejnmark L, Silva BC, Walker MD, Bilezikian JP. Current issues in the presentation of asymptomatic primary hyperparathyroidism: proceedings of the Fourth International Workshop. J Clin Endocrinol Metab 2014; 99:3580-3594
  248. Cheung K, Wang TS, Farrokhyar F, Roman SA, Sosa JA. A meta-analysis of preoperative localization techniques for patients with primary hyperparathyroidism. Ann Surg Oncol 2012; 19:577-583
  249. Civelek AC, Ozalp E, Donovan P, Udelsman R. Prospective evaluation of delayed technetium-99m sestamibi SPECT scintigraphy for preoperative localization of primary hyperparathyroidism. Surgery 2002; 131:149-157
  250. Van Husen R, Kim LT. Accuracy of surgeon-performed ultrasound in parathyroid localization. World J Surg 2004; 28:1122-1126
  251. Mortenson MM, Evans DB, Lee JE, Hunter GJ, Shellingerhout D, Vu T, Edeiken BS, Feng L, Perrier ND. Parathyroid exploration in the reoperative neck: improved preoperative localization with 4D-computed tomography. J Am Coll Surg 2008; 206:888-895; discussion 895-886
  252. Yeh R, Tay YD, Tabacco G, Dercle L, Kuo JH, Bandeira L, McManus C, Leung DK, Lee JA, Bilezikian JP. Diagnostic Performance of 4D CT and Sestamibi SPECT/CT in Localizing Parathyroid Adenomas in Primary Hyperparathyroidism. Radiology 2019; 291:469-476
  253. Maser C, Donovan P, Santos F, Donabedian R, Rinder C, Scoutt L, Udelsman R. Sonographically guided fine needle aspiration with rapid parathyroid hormone assay. Ann Surg Oncol 2006; 13:1690-1695
  254. Udelsman R, Donovan PI. Remedial parathyroid surgery: changing trends in 130 consecutive cases. Ann Surg 2006; 244:471-479
  255. Clark OH. How should patients with primary hyperparathyroidism be treated? J Clin Endocrinol Metab 2003; 88:3011-3014
  256. Udelsman R. Six hundred fifty-six consecutive explorations for primary hyperparathyroidism. Ann Surg 2002; 235:665-670; discussion 670-662
  257. Irvin GL, 3rd, Solorzano CC, Carneiro DM. Quick intraoperative parathyroid hormone assay: surgical adjunct to allow limited parathyroidectomy, improve success rate, and predict outcome. World J Surg 2004; 28:1287-1292
  258. Udelsman R, Donovan PI, Sokoll LJ. One hundred consecutive minimally invasive parathyroid explorations. Ann Surg 2000; 232:331-339
  259. Westerdahl J, Bergenfelz A. Unilateral versus bilateral neck exploration for primary hyperparathyroidism: five-year follow-up of a randomized controlled trial. Ann Surg 2007; 246:976-980; discussion 980-971
  260. Russell CF, Dolan SJ, Laird JD. Randomized clinical trial comparing scan-directed unilateral versus bilateral cervical exploration for primary hyperparathyroidism due to solitary adenoma. Br J Surg 2006; 93:418-421
  261. Jinih M, O'Connell E, O'Leary DP, Liew A, Redmond HP. Focused Versus Bilateral Parathyroid Exploration for Primary Hyperparathyroidism: A Systematic Review and Meta-analysis. Ann Surg Oncol 2017; 24:1924-1934
  262. Miccoli P, Berti P, Materazzi G, Ambrosini CE, Fregoli L, Donatini G. Endoscopic bilateral neck exploration versus quick intraoperative parathormone assay (qPTHa) during endoscopic parathyroidectomy: A prospective randomized trial. Surg Endosc 2008; 22:398-400
  263. Henry JF, Sebag F, Tamagnini P, Forman C, Silaghi H. Endoscopic parathyroid surgery: results of 365 consecutive procedures. World J Surg 2004; 28:1219-1223
  264. Silverberg SJ, Gartenberg F, Jacobs TP, Shane E, Siris E, Staron RB, McMahon DJ, Bilezikian JP. Increased bone mineral density after parathyroidectomy in primary hyperparathyroidism. J Clin Endocrinol Metab 1995; 80:729-734
  265. Kulak CA, Bandeira C, Voss D, Sobieszczyk SM, Silverberg SJ, Bandeira F, Bilezikian JP. Marked improvement in bone mass after parathyroidectomy in osteitis fibrosa cystica. J Clin Endocrinol Metab 1998; 83:732-735
  266. Tritos NA, Hartzband P. Rapid improvement of osteoporosis following parathyroidectomy in a premenopausal woman with acute primary hyperparathyroidism. Arch Intern Med 1999; 159:1495-1498
  267. DiGregorio S. Hiperparatiroidismo primario: dramatico incremento de la masa ostea post paratiroidectomia. Diagn Osteol 1999; 1:11-15
  268. Dawson-Hughes B, Stern DT, Shipp CC, Rasmussen HM. Effect of lowering dietary calcium intake on fractional whole body calcium retention. J Clin Endocrinol Metab 1988; 67:62-68
  269. Barger-Lux MJ, Heaney RP. Effects of calcium restriction on metabolic characteristics of premenopausal women. J Clin Endocrinol Metab 1993; 76:103-107
  270. Calvo MS, Kumar R, Heath H. Persistently elevated parathyroid hormone secretion and action in young women after four weeks of ingesting high phosphorus, low calcium diets. J Clin Endocrinol Metab 1990; 70:1334-1340
  271. Insogna KL, Mitnick ME, Stewart AF, Burtis WJ, Mallette LE, Broadus AE. Sensitivity of the parathyroid hormone-1,25-dihydroxyvitamin D axis to variations in calcium intake in patients with primary hyperparathyroidism. N Engl J Med 1985; 313:1126-1130
  272. Locker FG, Silverberg SJ, Bilezikian JP. Optimal dietary calcium intake in primary hyperparathyroidism. Am J Med 1997; 102:543-550
  273. Jorde R, Szumlas K, Haug E, Sundsfjord J. The effects of calcium supplementation to patients with primary hyperparathyroidism and a low calcium intake. Eur J Nutr 2002; 41:258-263
  274. Marcocci C, Bollerslev J, Khan AA, Shoback DM. Medical management of primary hyperparathyroidism: proceedings of the fourth International Workshop on the Management of Asymptomatic Primary Hyperparathyroidism. J Clin Endocrinol Metab 2014; 99:3607-3618
  275. Rolighed L, Rejnmark L, Sikjaer T, Heickendorff L, Vestergaard P, Mosekilde L, Christiansen P. Vitamin D treatment in primary hyperparathyroidism: a randomized placebo controlled trial. J Clin Endocrinol Metab 2014; 99:1072-1080
  276. Purnell DC, Scholz DA, Smith LH, Sizemore GW, Black MB, Goldsmith RS, Arnaud CD. Treatment of primary hyperparathyroidism. Am J Med 1974; 56:800-809
  277. Broadus AE, Magee JS, Mallette LE, Horst RL, Lang R, Jensen PS, Gertner JM, Baron R. A detailed evaluation of oral phosphate therapy in selected patients with primary hyperparathyroidism. J Clin Endocrinol Metab 1983; 56:953-961
  278. Gallagher JC, Nordin BE. Treatment with with oestrogens of primary hyperparathyroidism in post-menopausal women. Lancet 1972; 1:503-507
  279. Marcus R, Madvig P, Crim M, Pont A, Kosek J. Conjugated estrogens in the treatment of postmenopausal women with hyperparathyroidism. Ann Intern Med 1984; 100:633-640
  280. Selby PL, Peacock M. Ethinyl estradiol and norethindrone in the treatment of primary hyperparathyroidism in postmenopausal women. N Engl J Med 1986; 314:1481-1485
  281. Grey AB, Stapleton JP, Evans MC, Tatnell MA, Reid IR. Effect of hormone replacement therapy on bone mineral density in postmenopausal women with mild primary hyperparathyroidism. A randomized, controlled trial. Ann Intern Med 1996; 125:360-368
  282. Rubin MR, Lee KH, McMahon DJ, Silverberg SJ. Raloxifene lowers serum calcium and markers of bone turnover in postmenopausal women with primary hyperparathyroidism. J Clin Endocrinol Metab 2003; 88:1174-1178
  283. Kaplan RA, Geho WB, Poindexter C, Haussler M, Dietz GW, Pak CY. Metabolic effects of diphosphonate in primary hyperparathyroidism. J Clin Pharmacol 1977; 17:410-419
  284. Shane E, Baquiran DC, Bilezikian JP. Effects of dichloromethylene diphosphonate on serum and urinary calcium in primary hyperparathyroidism. Ann Intern Med 1981; 95:23-27
  285. Rossini M, Gatti D, Isaia G, Sartori L, Braga V, Adami S. Effects of oral alendronate in elderly patients with osteoporosis and mild primary hyperparathyroidism. J Bone Miner Res 2001; 16:113-119
  286. Hassani S, Braunstein GD, Seibel MJ, Brickman AS, Geola F, Pekary AE, Hershman JM. Alendronate therapy of primary hyperparathyroidism. Endocrinologist 2001; 11:459-464
  287. Chow CC, Chan WB, Li JK, Chan NN, Chan MH, Ko GT, Lo KW, Cockram CS. Oral alendronate increases bone mineral density in postmenopausal women with primary hyperparathyroidism. J Clin Endocrinol Metab 2003; 88:581-587
  288. Parker CR, Blackwell PJ, Fairbairn KJ, Hosking DJ. Alendronate in the treatment of primary hyperparathyroid-related osteoporosis: a 2-year study. J Clin Endocrinol Metab 2002; 87:4482-4489
  289. Khan AA, Bilezikian JP, Kung AW, Ahmed MM, Dubois SJ, Ho AY, Schussheim D, Rubin MR, Shaikh AM, Silverberg SJ, Standish TI, Syed Z, Syed ZA. Alendronate in primary hyperparathyroidism: a double-blind, randomized, placebo-controlled trial. J Clin Endocrinol Metab 2004; 89:3319-3325
  290. Eller-Vainicher C, Palmieri S, Cairoli E, Goggi G, Scillitani A, Arosio M, Falchetti A, Chiodini I. Protective Effect of Denosumab on Bone in Older Women with Primary Hyperparathyroidism. J Am Geriatr Soc 2018; 66:518-524
  291. Nemeth EF, Scarpa A. Rapid mobilization of cellular Ca2+ in bovine parathyroid cells evoked by extracellular divalent cations. Evidence for a cell surface calcium receptor. J Biol Chem 1987; 262:5188-5196
  292. Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, Kifor O, Sun A, Hediger MA, Lytton J, Hebert SC. Cloning and characterization of an extracellular Ca(2+)-sensing receptor from bovine parathyroid. Nature 1993; 366:575-580
  293. 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
  294. Fox J, Hadfield S, Petty BA, Nemeth EF. A first generation calcimimetic compound (NPS R-568) that acts on the parathyroid cell calcium receptor: a novel therapeutic approach for hyperparathyroidism. J Bone Miner Res 1993; 8
  295. Heath H, Sanguinetti EL, Oglseby S, Marriott TB. Inhibition of human parathyroid hormone secretion in vivo by NPS R-568, a calcimimetic drug that targets the parathyroid cell surface calcium receptor. Bone 1995; 16:85S
  296. Silverberg SJ, Bone HG, 3rd, Marriott TB, Locker FG, Thys-Jacobs S, Dziem G, Kaatz S, Sanguinetti EL, Bilezikian JP. Short-term inhibition of parathyroid hormone secretion by a calcium-receptor agonist in patients with primary hyperparathyroidism. N Engl J Med 1997; 337:1506-1510
  297. Shoback DM, Bilezikian JP, Turner SA, McCary LC, Guo MD, Peacock M. The calcimimetic cinacalcet normalizes serum calcium in subjects with primary hyperparathyroidism. J Clin Endocrinol Metab 2003; 88:5644-5649
  298. Peacock M, Bilezikian JP, Klassen PS, Guo MD, Turner SA, Shoback D. Cinacalcet hydrochloride maintains long-term normocalcemia in patients with primary hyperparathyroidism. J Clin Endocrinol Metab 2005; 90:135-141
  299. Marcocci C, Chanson P, Shoback D, Bilezikian J, Fernandez-Cruz L, Orgiazzi J, Henzen C, Cheng S, Sterling LR, Lu J, Peacock M. Cinacalcet reduces serum calcium concentrations in patients with intractable primary hyperparathyroidism. J Clin Endocrinol Metab 2009; 94:2766-2772
  300. Peacock M, Bolognese MA, Borofsky M, Scumpia S, Sterling LR, Cheng S, Shoback D. Cinacalcet treatment of primary hyperparathyroidism: biochemical and bone densitometric outcomes in a five-year study. J Clin Endocrinol Metab 2009; 94:4860-4867
  301. Silverberg SJ, Rubin MR, Faiman C, Peacock M, Shoback DM, Smallridge RC, Schwanauer LE, Olson KA, Klassen P, Bilezikian JP. Cinacalcet hydrochloride reduces the serum calcium concentration in inoperable parathyroid carcinoma. J Clin Endocrinol Metab 2007; 92:3803-3808
  302. Tsvetov G, Hirsch D, Shimon I, Benbassat C, Masri-Iraqi H, Gorshtein A, Herzberg D, Shochat T, Shraga-Slutzky I, Diker-Cohen T. Thiazide Treatment in Primary Hyperparathyroidism - A New Indication for an Old Medication? J Clin Endocrinol Metab 2016:jc20162481
  303. Riss P, Kammer M, Selberherr A, Bichler C, Kaderli R, Scheuba C, Niederle B. The influence of thiazide intake on calcium and parathyroid hormone levels in patients with primary hyperparathyroidism. Clin Endocrinol (Oxf) 2016; 85:196-201
  304. Farquhar CW, Spathis GS, Barron JL, Levin GE. Failure of thiazide diuretics to increase plasma calcium in mild primary hyperparathyroidism. Postgraduate medical journal 1990; 66:714-716
  305. Bradwell AR, Harvey TC. Control of hypercalcaemia of parathyroid carcinoma by immunisation. Lancet 1999; 353:370-373
  306. Betea D, Bradwell AR, Harvey TC, Mead GP, Schmidt-Gayk H, Ghaye B, Daly AF, Beckers A. Hormonal and biochemical normalization and tumor shrinkage induced by anti-parathyroid hormone immunotherapy in a patient with metastatic parathyroid carcinoma. J Clin Endocrinol Metab 2004; 89:3413-3420
  307. Vellanki P, Lange K, Elaraj D, Kopp PA, El Muayed M. Denosumab for management of parathyroid carcinoma-mediated hypercalcemia. J Clin Endocrinol Metab 2014; 99:387-390
  308. Nadarasa K, Theodoraki A, Kurzawinski TR, Carpenter R, Bull J, Chung TT, Drake WM. Denosumab for management of refractory hypercalcaemia in recurrent parathyroid carcinoma. Eur J Endocrinol 2014; 171:L7-8
  309. Collins MT, Skarulis MC, Bilezikian JP, Silverberg SJ, Spiegel AM, Marx SJ. Treatment of hypercalcemia secondary to parathyroid carcinoma with a novel calcimimetic agent. J Clin Endocrinol Metab 1998; 83:1083-1088

Diabetes Mellitus and Tuberculosis

ABSTRACT

 

The converging epidemics of non-communicable disease like DM (DM) and an infectious disease like tuberculosis (TB) is a double burden. DM is increasing in the same population that is at high risk for developing TB. There is a two-to-four-fold higher risk of active TB in individuals with DM and up to 30% of individuals with TB are likely to have DM. Immune deficiency either in absolute or relative quantities are sufficient for re-activation of latent TB. From a 10% risk of reactivation over the whole lifetime of an immunocompetent individual, the risk of reactivation increases to 10% every year in immune-deficient individuals. DM impairs cell mediated immunity and poor glycemic control affects cytokine response and alters the defenses in the alveolar macrophages. Fever, hemoptysis, extensive parenchymal lesions, and lung cavities are more common in those with DM particularly heavier and older males. DM increases the risk of treatment failure, death, and relapse. Evidence collected from meta-analysis conclude that DM can increase the odds of developing Multi Drug resistant TB (MDR-TB). The synergism between DM and TB necessitates bi-directional screening. Sputum examination for Ziehl-Neelsen staining is both a sensitive and specific screening test. Rapid molecular diagnostic tests like cartridge based nucleic acid amplification tests (CB-NAAT) are useful in cases where there is a high-index of suspicion and difficulty in arriving at a definitive diagnosis exists. Random plasma glucose and HbA1c (glycosylated Hemoglobin) measurements are convenient tests for DM screening that can be done in non-fasting individuals. Screening for DM more than once during the course of illness is sensible so that transient DM and new-onset DM can be identified. Anti-TB drugs affect glycemic control as they interact with anti-diabetic drugs by either stimulating or inhibiting the metabolizing enzymes. They may also aggravate metabolic, ocular, and neuropathic complications of DM. Insulin is the preferred drug in most instances. The presence of renal and hepatic dysfunction affects TB and hyperglycemic management.

 

INTRODUCTION

 

Tuberculosis (TB) and diabetes mellitus (DM) are two diverse conditions of immense public health importance existing for centuries. TB was traditionally identified with poverty while DM was considered as an entity associated with prosperity. TB is today one of the commonest and widespread communicable infectious diseases largely but not necessarily confined to low-economic groups. DM on the other hand spearheads the group of chronic non-communicable diseases affecting people across all socio-economic strata. Contrary to previous beliefs, a larger number of people with DM are living in middle- and low-income countries. Unfortunately, these are the countries where DM is expected to increase in the near future (1). Both DM and TB have been associated with significant morbidity and mortality from time immemorial. Advancements in modern medical science over the years has definitely improved the outcome in both these conditions. But the magnitude of these two diseases has not waned and both are collaborative in worsening each other. In fact, the increase in the population affected with DM is sustaining the TB epidemic.

 

TB is associated with the endocrine system in different ways. The effects of TB on the endocrine system are discussed in detail in another Endotext chapter (2).  The interaction of TB and DM is discussed in this chapter.

 

TUBERCULOSIS

 

TB is a global health threat, particularly to the poor and the susceptible. It is estimated that on average approximately 9 to 10 million people are affected by TB and around 1 to 2 million succumb to it annually (3). In 2019, 1.3 million people died due to TB. In developed countries, TB has slipped down in ranking among the global list of top 10 diseases causing mortality. However, in the underdeveloped regions it still remains among the top 10 diseases with high mortality (up to 30%). When in concurrence with retroviral infections, the risk of active TB is 12 to 20 times higher and the mortality is higher even in developed countries (4). Multi drug resistant TB is another rising problem which requires expensive second line drugs and a longer duration of treatment. A large proportion of the global population is at risk and the true prevalence and the annual incidence also depends on access to health care facilities and the laboratory-based testing capacity of the regions. Individuals who are in close contact with affected individuals, people living in crowded places, immigration to a country or area with a high prevalence of TB, and children less than 5 years of age are considered vulnerable to getting infected by the TB bacilli (Table 1). Bacillary load in the sputum of the infectious individual and close proximity to the infectious persons are two important external determinants for infection in any individual after exposure.

 

Table 1. Risk Factors for TB Infection, Disease, and Outcome

Stage of TB

Intrinsic Factors

Extrinsic factors

 

Exposure to infection

Closeness of contact

Duration of contact

Load of Bacilli

 

 

Overcrowding and lack of ventilation

Indoor pollution

Community prevalence of TB

Tobacco Use, Drug abuse

Alcohol

Migration

 

 

Infection to disease progression

Altered Immune status (disease or drug induced)

Lack of BCG vaccination

Nourishment

DM

Malignancy

Respiratory diseases like Silicosis

Age

Male

Disease Outcome  

Female sex, Social Stigma, Immune status, Malnourishment, DM, Malignancy, Age, MDR-TB,

Barriers to health care access:   Cultural, Geographical, Economical, Weak social support, Weak health care support

 

Latent Tuberculosis

 

In most of the exposed individuals the infection is quelled by the immune system and the bacilli are fenced inside a granuloma or tubercle, immunologically aborting an active disease. This is a subclinical disease (LTBI – Latent TB infection) which doesn’t have symptoms and can last for weeks or decades. This latent infection is seen in nearly one third of the world population. Even though non-infectious, they carry the risk (about 10%) of re-activation into active TB later (primary progressive TB) (5). Such re-activation occurs in immunocompromised as in HIV infection, those on immunosuppressive agents (such as post organ transplant, autoimmune diseases, and allergic diseases), conditions like DM, alcoholism, substance abuse, silicosis, malnutrition, steroid therapy, renal failure, malignancies, indoor air pollution, and smoking. The World Health Organization (WHO) has guidelines on the approach to latent TB especially in countries where the burden of TB is low (an incidence of < 100 / 1, 00,000 per year). It strongly recommends screening and treatment of latent TB in high-risk individuals in these countries (6).

 

TB is an airborne infectious disease which spreads by droplets. TB affects the lungs primarily (Pulmonary TB) and when it affects the pleura, bones/joints, abdominal organs, lymph nodes, and meninges it is called extra-pulmonary TB. Mycobacterium tuberculosis, the causative agent has a thick mycolic acid cell wall that enables its survival in the environment and in its host. External to its cell membrane it has a peptidoglycan polymer which makes it impermeable. Its cell wall also contains lipoarabinomannan which enables its phagocytosis by macrophages and facilitates its survival inside macrophages in airways especially alveoli (7). The ability of the host’s defense system then determines whether the outcome is a progressive primary pulmonary disease or a latent state.

 

DIABETES MELLITUS

 

The prevalence of DM is rapidly increasing to justify it to be termed as an epidemic disease. According to WHO, the global prevalence of DM has doubled from 4.7 % in 1980 to 8.5% in 2014 (8). From an estimated prevalence of 463 million in 2019, it is estimated to increase to 578 million in 2030, and 700 million in 2045. For every diagnosed individual with DM there is another undiagnosed person with DM (9). The differences in the prevalence of DM between high and middle-income countries and similarly between rural and urban population are decreasing.

 

According to WHO, non-communicable diseases constitute 7 out of the top 10 leading causes of death and DM is prominent among them. In 2019 the estimated number of deaths to have occurred due to DM globally is approximately 4 million (10). DM is associated with significant morbidity due to its microvascular and macrovascular complications and high cardiovascular mortality. DM is a major cause of cardiac ischemia, stroke, renal failure, blindness, and amputations.

 

 

The high prevalence of DM and TB being in epidemic proportions has rightly earned them the names ‘the converging epidemics’ and ‘double burden’ (11,12). Due to rapid changes in lifestyle, urbanization, and epidemiological changes, DM is increasingly seen in low- and medium-income groups, and in younger individuals more frequently than before. The prevalence of DM is increasing faster where TB is endemic already. Unfortunately, these are the regions in the world where health care facilities are less common. According to International Diabetes Federation, the 50-55% increase in the prevalence of DM over the next 2 decades will occur predominantly in the continent of Africa (10). A longitudinal, multi-national study involving low-income countries concluded that an odds ratio of 4.7 for prevalence and 8.1 for incidence of TB is highly likely in those counties where DM has increased over the last decade (13). The WHO states that younger age group individuals are three times more likely to get infected with TB (14). There is a two-to four-fold higher risk of active TB in individuals with DM compared to non-diabetic individuals (15). According to a meta-analysis by Wilkinson et al, around 4 % of people with type 2 DM develop TB (16). Since the number of individuals with undiagnosed DM in the world is expected to be more than 50%, the proportion of TB in DM also should be much higher. Analytical models that try to project the future burden of TB in DM predict that the increase in DM counteracts the decreasing incidence of TB by at least 3% over the next 15 years (17).

 

Increased chances of finding undetected and uncontrolled hyperglycemia in close household contacts of subjects with TB have been mentioned in studies from Asian counties. In fact, a systemic analysis of a group of heterogenous studies on bi-directional screening i.e., screening for TB in DM affected individuals and vice versa has shown some evidence to support active screening for both DM and TB in the affected individuals and family members (18). Up to 35% of TB patients may have DM and the quoted figures are variable in literature (19). Jean Jacques Noubiap et al in their systematic review and meta-analysis of data from 2.3 million people with TB world-wide estimated that the prevalence of DM in patients with TB is around 15 % and was twice that of the general population (20). These data clearly give epidemiological evidence for the co-existence of DM and TB as a syndemic. A potentially lethal combination of a communicable disease and a non-communicable disease having a synergistic effect is a challenge on the public health system.

 

Epidemiology of Diabetes Among Patients with Active Tuberculosis

 

The WHO collaborative framework recommends for a joint plan for DM and TB related activities which have to be reflected in the national plans on non-communicable diseases and TB respectively (21). As per the WHO approximately15% of TB cases in the world are associated with DM. Of these 15%, India accounts for more than 40% of the cases (19). Estimates of the burden of DM and TB co-existence has primarily come from studies looking at prevalence of DM and glucose intolerance among newly diagnosed patients with TB diagnosed in TB clinics. The community prevalence of these two disorders co-existing are not clear. Many of these studies have been done in hospitals where sicker patients with TB are treated which would probably account for a higher percentage of patients having glucose intolerance and DM. In a study from five randomly selected TB care clinics in the southern state of Tamil Nadu, around 25% of patients with TB had coexisting DM. Around 10% of these patients had newly diagnosed DM. Risk factors for having DM in this group of patients included older age, higher basal metabolic index (BMI), family history of DM among first degree relatives, and following a sedentary lifestyle (22). Close to Tamil Nadu in the southern state of Kerala, the prevalence of DM in patients with TB was nearly double that observed in the previous study. Among the patients with TB in Kerala, 44% had DM. Among them about half (21%) had newly diagnosed DM (23). The risk of DM was higher among those with sputum positive TB in both of these studies. In a study from Odisha on the Eastern part of India, 13.9% of tribal patients with TB had DM suggestive of a significant burden of disease even among poorer regions of the country (24). In a retrospective study, among 1000 patients with TB from the northern state of Punjab, 11.6% had DM and TB coexistence (25). In the same state the authors found 30% of patients with TB diagnosed in a tertiary referral hospital had DM (26).

 

To better understand the prevalence of DM among newly diagnosed patients with TB a large multicentric study involving five centers is in progress (GIANT Study - Glucose Intolerance Among New patients with TB - Clinical Trial Registry India - CTRI/2019/05/019396). This study incorporates simultaneous OGTT and HbA1c determinations at three different time points to ascertain if HbA1c can replace the standard oral glucose tolerance test (OGTT) in this population and avoid the inconvenience of performing an OGTT.

 

A recent systemic review and meta-analysis ascertained the worldwide prevalence of DM among active cases of TB. This meta-analysis involved over 200 studies that included a little under 2.3 million patients with active TB. The overall pooled prevalence was similar to the WHO estimate of 15% prevalence of DM in patients with active TB. However, this varied from 0.1% in Latvia to 45% in the Marshall Islands. The high prevalence areas as per the International Diabetes Federation included North America, Western Pacific (which includes Australia and China), South East Asia, North Africa, and Middle East Asia (27). Figure 1 summarizes the information on the prevalence of DM and active TB in 7 regions of the world.

Figure 1. Prevalence of DM among patients with active TB in the seven International Diabetes Federation (IDF) regions of the world. The areas in red have the high burden of DM co-existing with active TB. The low burden areas are marked in blue. (adapted from ref 27)

DIABETES PREDISPOSING TO TUBERCULOSIS

 

Absolute or relative immune deficiency is sufficient for re-activation of latent TB. The association of increased prevalence of TB in immune deficient diseases like HIV is well established. The wider prevalence of DM makes DM a more important risk factor for developing TB than retroviral diseases. The higher incidence of multi-drug resistant TB reported to be significant in some studies (Odds Ratio of 2.1) reiterates the role of immune dysregulation in DM (28). The immune response in DM to TB is supposed to be hyper-reactive but ineffective and even deleterious as it may produce pulmonary tissue damage.

 

Chronic hyperglycemia impairs immunity (both innate and adaptive). DM impairs cell mediated immunity and poor glycemic control affects cytokine response and alters the defenses in the alveolar macrophages. Hyperglycemia disrupts the recruitment of neutrophils, chemotactic movement of monocytes, and phagocytic action of alveolar macrophages. Also, the antigen-specific interferon-gamma release is affected as the T-helper cell activation is ineffective. In addition, altered pulmonary microvasculature and micronutrient deficiency facilitate the invasion and establishment of TB as surveillance and nutrition is compromised. The chronic immunosuppression or ineffective immune response predisposes the individual for TB infection and with a higher bacilli load. This is summarized in Figure 2.

Figure 2. DM is associated with increase in active TB and relapse in TB which both may be a result of the direct effect of diabetes. There is increased death in DM and TB which may be secondary to TB or due to the inherent excess mortality of DM due to cardiovascular disease

CLINICAL PRESENTATION

 

The manifestations of tuberculous infection in patients with DM have been documented to be different from those without diabetes. Fever and hemoptysis in diabetic population is more common compared to the general population. Radiographic differences include higher than usual parenchymal lesions and lung cavities (30). There are reports of a higher incidence of lower lobe involvement in individuals with DM in contrast to the classical upper lobe involvement in the general population. Also, a higher rate of other atypical presentations like a reduced rate of sputum conversion (low quality evidence), higher probability of treatment failure and death (moderate quality evidences) is known to occur when DM occurs with TB. A higher rate of recurrence and reactivation of latent TB infection (OR=1.83) has been documented (31). Subjects affected with TB and DM are found to be heavier and older males compared to those without diabetes. More pulmonary than extra-pulmonary involvement is seen in TB with DM (32).

 

Outcome

 

Negative smear or culture on two separate occasions while on treatment and on completion of treatment defines a cured TB (Table 2). Apart from being a risk factor for increased incidence of active TB, co-existence of DM worsens the outcome even in treated patients. In the pre-insulin era, the commonest cause of death in DM apart from diabetic coma was the co-affection with TB (33). DM increases the risk of treatment failure, death and relapse. The risks are likely to be an underestimation as loss of follow-up or unreported death has been a major problem in data collection on outcome in TB management. In a systematic review and meta-analysis by Baker et al, the risk ratio for combined treatment failure and death was 1.69. The risk ratio for death was 1.89 when unadjusted and went up to 4.95 when adjusted for age and other confounding factors.

 

Table 2. Terminology and Definitions

Terminology

Definitions

Treatment completed

Bacteriologically confirmed TB patient who has completed treatment without evidence of failure, but not yet completed sputum test to prove negative result in the last month of treatment and on at least one previous occasion

Cured

Bacteriologically confirmed TB in whom smear- or culture-was negative in the last month of treatment and on at least one previous occasion

Treatment Success

The sum of cured and treatment completed

Treatment Failed

Sputum smear or culture positive at or beyond 5th month of treatment

Died

A proven patient who dies for any reason before or during the course of treatment

Lost to follow-up

A proven patient who did not start treatment or who has interrupted treatment for 2 or more consecutive months

Not Evaluated

A proven patient for whom no treatment outcome is assigned. Includes cases “transferred out” to another treatment unit as well as cases for which the treatment outcome is unknown to the reporting unit

Terminology and Definitions adopted from RNTCP, Revised National TB Control Programme, Training Course for Programme Manager (Modules 1-4), 2011.Training modules. Central TB Division. https://tbcindia.gov.in.

 

The risk ratio for relapse was 3.89 but no additional risk of TB relapse in those with multidrug resistant TB was demonstrated. In their analysis, Baker et al found that the effect of co-existing DM on sputum conversion 2 to 3 months after treatment was variable (0.79 to 3.25) and wide (34). Persistence of sputum positivity i.e., delay in sputum conversion has been shown in a few studies. The authors conclude that advancing age and underlying co-morbidity contribute to death and is not due to drug resistant TB or severity of hyperglycemia (35).

 

BI-DIRECTIONAL SCREENING FOR DIABETES AND TUBERCULOSIS

 

The synergism between DM and TB in terms of epidemiology and outcome necessitates bi-directional screening for the presence of either TB or DM in the presence of the other disorder. The Collaborative Framework for Care and Control of TB and DM proposed by the World Health Organization (WHO) along with the International Union against TB and Lung is an effort towards bi-directional screening and management of both these conditions (36). Studies implementing bi-directional screening point towards its feasibility and effectiveness (18).

 

Screening for Active TB Among Patients with Diabetes

 

The diagnosis and treatment of TB is affected by substantial delay which occurs at multiple levels a) between the onset of symptoms and clinical presentation b) clinical presentation and suspicion of TB c) Clinical suspicion of TB and its confirmation. This is due to variability in symptoms, host immunity, lack of knowledge, paucity of access to medical care, and lack of rapid and reliable diagnostic tools. The average delay even in resource- rich countries after presentation to heath care system is 3 weeks (37). According to WHO, patients with suspected TB should be promptly sent to TB diagnostic and treatment centers and evaluated accordingly.

 

The higher risk of TB in diabetic population compels intensive screening for detection of TB at the earliest time so as to reduce transmission, morbidity, and mortality. WHO recommends for TB surveillance among patients with DM in settings with medium to high TB burdens. The practical difficulties are the non-availability of TB screening tools in all DM clinics. Also, in areas of low TB burden, the cost-effectiveness is low. The number needed to screen to detect one case of active TB depends on the prevalence in that area. Screening of all patients with clinical history during their visit to diabetic clinic and additional testing in symptomatic and high-risk patients should be undertaken. Also screening for TB whenever there is unexplained worsening of metabolic control would help detect occult cases. Different modalities (clinical, radiological, sputum microbiology) alone or in combination are used for screening individuals with DM for the presence of active TB.

 

Clinical Assessment

 

It is inexpensive and requires minimum time. Fever, cough of more than 2 weeks duration, hemoptysis, weight loss, night sweats, and exposure to a case of active TB are the clinical clues to suspect pulmonary TB. Lymphadenopathy, fever with altered sensorium, neck stiffness, abdominal symptoms like ascites, intestinal obstruction etc. all favor the possibilities of extra-pulmonary TB. But clinical symptoms lack both sensitivity and specificity as it excludes asymptomatic patients and relies on the presence of symptoms.

 

Radiography of the Chest

 

It has good sensitivity to pick up asymptomatic pulmonary cases, but there can be false positive results. Inconsistent evidence exists on the presence of atypical findings of TB in the chest x-rays of individuals with diabetes. The presence of clinical symptoms of fever, cough, hemoptysis, and weight loss with an abnormal chest-x-ray helps in the presumptive diagnosis of TB. Figure 3, 4, and 5 show different radiological presentations of pulmonary TB.

Figure 3. Chest Radiographs suggesting fibrocavitatory lesions. Post primary infections and reactivation of pulmonary TB are more likely to cavitate. They are most common in the posterior segments of the upper lobes (85%) as seen in Picture A. Red arrow pointing to the cavity. The other common site is the superior segment of the lower lobe (Picture B) Yellow arrow pointing to the cavity (Picture courtesy- Prof Mary John, Christian Medical College and Hospital, Ludhiana)

Figure 4. Chest Radiographs suggesting lobar consolidation. (Picture courtesy- Prof Mary John& Dr Neeru Mittal, Christian Medical College and Hospital, Ludhiana)

Figure 5. Chest Radiographs suggesting miliary TB. It represents hematogenous dissemination of an uncontrolled tuberculous infection. Although implants are seen throughout the body, the lungs are usually the easiest location to image. Miliary deposits appear as 1-3 mm diameter nodules uniformly distributed in the lung parenchyma. (Picture courtesy- Prof Mary John & Dr Neeru Mittal, Christian Medical College and Hospital, Ludhiana)

Microscopic Examination 

 

Sputum collected for Ziehl-Neelsen staining and examination for acid -fast bacilli is both sensitive and specific. Even-though they are the commonly used confirmatory tests, most diabetes-oriented clinics are unlikely to have standard laboratory facilities for sputum tests even though having a radiography unit for screening TB appears feasible (32). Sputum tests have limitations in cases of scanty sputum or salivary contamination especially in children. If sputum availability is scanty then sputum is induced by saline nebulization, which if not helpful, can be followed by bronchoscopy assisted lavage or trans-bronchial pulmonary biopsy.

 

Sputum Culture

 

The gold standard test is sputum culture for TB bacilli but is both time consuming (turnaround time 8 weeks) and expensive. It cannot be used for all individuals attending the DM clinic and is reserved for those patients where the index of suspicion is high and in difficult cases when other available tests are not contributory for diagnosis. Sputum culture is also useful for assessing response in multi-drug resistant TB.

 

Rapid Molecular Diagnostic Tests

 

Tests like cartridge based nucleic acid amplification test (CB-NAAT) (Figure 6) or rapid automated molecular test Expert MTB/ RIF assay using polymerase chain reaction have a quick turnaround time of two hours and additional advantage of using a single sputum sample. They also detect the presence of rifampicin resistance. Being expensive it cannot be used for screening all patients with DM even though it has high sensitivity and specificity. But they are useful in cases with high-index of suspicion and difficulty in arriving at a definitive diagnosis.

Figure 6. All district hospitals in India have been provided with Cartridge Based-Nucleic Acid Amplification Testing equipment under the RNTPC program. Picture of the equipment at Civil Hospital, Ludhiana (Picture courtesy- Dr Ashish Chawla, Civil Hospital, Ludhiana)

Screening for Latent TB Among Patients with Diabetes

 

As mentioned earlier, Identification and treatment of latent TB infection to prevent its progression to active disease is necessary to prevent morbidity, mortality, and spread of TB. The WHO AND USPSTF (US preventive services task force) have issued strong guidelines for the screening and treatment of latent TB infection in high risk adults aged more than 18 years in countries with low incidence of TB (38,39).The high risk groups include persons hailing from countries with high TB prevalence, persons residing in homeless shelters and correctional facilities, immunocompromised individuals (HIV, those on immunosuppressants including post-organ transplant), silicosis, those receiving dialysis, those receiving anti-TNF-alfa inhibitor treatment, previously treated TB, and persons who come in contact with those active TB (household contacts and health care workers). The Mantoux tuberculin skin test (TST) and interferon-gamma release assays (IGRAs) are the two screening tests used and they are moderately sensitive and highly specific (40,41). In the tuberculin test, purified protein derivative (PPD) is injected intradermally and assessed within 48 to 72 hours for the presence of induration which is a palpable hard swelling (a diameter of more than 10 mm is considered positive) over the injected area (42,43). In the IGRA a single venous blood sample is taken for the assay and the reports are available within a day. They are particularly useful in those who are unlikely to return for TST test reading and BCG vaccinees.

 

There is no consensus on the issue of screening for latent TB infection in DM as a high-risk group. The results of studies done previously have been inconsistent. Studies have shown a variable prevalence of latent TB infection and there are no randomized controlled trials demonstrating the benefits of screening. Similarly, there are no recommendations for chemoprophylaxis of latent TB infection in individuals with DM due to the lack of randomized controlled trials that show benefit. The small added risk of hepatotoxicity with chemoprophylactic drugs given for TB in latent TB infection has been the only concern arguing against such measures.

 

Screening for Diabetes in Tuberculosis

 

In geographical regions with a high prevalence of diabetes, screening for hyperglycemia in TB affected individuals is highly recommended (14). Detection and monitoring of hyperglycemia is an essential part of infection management in any patient with an infectious disease. Chronic infectious diseases like TB thrive in hyperglycemic individuals and the outcome is unfavorable in a hyperglycemic milieu. Recognition of hyperglycemia during the entire course of the illness in TB affected individuals implies either monitoring of pre-existing hyperglycemia or new onset of transient or permanent hyperglycemia. Transient hyperglycemia is a manifestation secondary to the insulin resistance induced by the inflammation of TB infection. There is evidence that hyperglycemic status improves during the course of anti-TB treatment. The optimal time to screen for DM in TB patients on anti-TB treatment is thus unresolved particularly when it is well known that transient hyperglycemia exists during the course of TB. Many groups have advocated for screening more than once during the course of illness i.e., once at the initiation of treatment and at least once again either during and at the time of completion of TB treatment.

 

In regions with high prevalence of diabetes, younger age of onset of DM is on the rise and this is an emerging problem. There is a three times higher risk for younger individuals to get TB and a two-to four-fold higher risk in diabetic individuals compared to non-diabetic individuals. Hence screening for DM in all individuals 18 years of age or older appears logical.

 

The type of tests for screening patients with TB for DM depends on the availability of the local health care facilities, cost of the tests, and the ability of patients to come back for additional or repeat tests. Symptom based screening for DM has a low sensitivity. Risk score-based screening also is marred by low sensitivity and specificity. Random plasma glucose test and HbA1c (glycosylated Hb) are convenient tests that can be done in non-fasting individuals as screening tests. Glycosylated Hb test which doesn’t require a fasting blood sample helps to differentiate stress induced hyperglycemia from spontaneous onset pre-existing diabetes. Oral glucose tolerance test (OGTT) with 75 gm helps in identifying impaired glucose tolerance (IGT) and frank DM. A FBG ≥126 mg/dL or random plasma glucose ≥200 mg/dl on two tests is diagnostic of diabetes; FBG 110to 125 mg/dL is considered as impaired fasting glucose and post glucose values between 141- to 199 mg/dl is taken as impaired glucose tolerance (43,44). In one study, the number of TB patients needed to screen (NNS) for detecting DM was on average 40. But in the same population it was lower among smear positive subjects (NNS = 23), in age less than 40 years (< 40 years vs. > 40 years NNS = 35 Vs 47), in males (male vs. female NNS = 31 vs. 116), smokers (smoker vs. non-smoker NNS = 27 vs. 68) and HIV positive (Positive vs. Negative 22 Vs 43) indicating that there are high risk individuals (46).

 

Currently, screening for DM in individuals with TB and screening for TB in patients with DM where the prevalence is > 100/1,00,000 population appears feasible. Once diagnosed as having diabetes, diet and drug therapy is initiated and the patients are followed up closely for assessing the glycemic response. Transient hyperglycemic situations improve either spontaneously or with minimal medical intervention. After completion of TB medications, regular follow-up with glycemic monitoring is recommended for all patients who had diabetes.

 

MANAGEMENT OF DIABETES AND TUBERCULOSIS 

 

Anti-Tuberculosis Therapy in Diabetes

 

Management of DM should be according to the existing global guidelines with adaptations according to the regional needs. The treatment of TB in DM is not different from the general population. DOTs (Directly Observed Treatment, short-course) is a patient-centered WHO strategy adopted to treat individuals with active TB. A trained health worker provides drugs and observes in-person the patient taking the drug. It guarantees compliance, completion of the treatment course and prevents transmission, treatment failure and development of drug resistance. For newly detected TB, 2 months of intensive phase with 4 drugs (INH, Rifampicin, Pyrazinamide and Ethambutol) and 4 months of continuation phase with 3 drugs (except pyrazinamide) is the standard regimen. For those with a relapse 3 months of intensive therapy with 5 drugs (streptomycin in addition) followed by 4 months of continuation phase with 3 drugs (except pyrazinamide and streptomycin) is administrated.

 

In Chronic Kidney Disease

 

All four first line drugs (RIF, INH, PZA and EMB) can be used in patients with CKD. Up to 50% dose reduction for EMB and PZA may be needed in patients with creatinine clearance <10 ml/min. Regular monitoring is advised to ensure optimal therapy.

 

Adverse Effects of Anti-TB Drugs

 

INH induced peripheral neuropathy may worsen the underlying diabetic neuropathy. Pyridoxine is supplemented to prevent this. INH is also associated with hepatitis. Ethambutol is known to produce optic nerve toxicity which may confound diabetic retinopathy. Also, ethambutol and rifampicin are known to affect the kidneys. Rifampicin can induce immune-allergic reactions. Pyrazinamide is rarely associated with liver injury but its more common side effect is hyperuricemia induced joint pain. Streptomycin is potentially associated with renal and cochleo-vestibular toxicity

 

Multi-Drug Resistant TB (MDR-TB)

 

Multi-drug resistant TB (MDR-TB) is an added medical and economical burden as it is much more difficult to treat, involves therapy with atypical anti-TB drugs for a longer duration of time, and requires referral to specialty centers. Infections caused by mycobacterial strains which are resistant to INH and rifampicin are called multi-drug resistant TB infections. If there is additional resistance to one fluroquinolone and one of the additional inject able drugs (Kanamycin, Capreomycin or Amikacin) then it is called extensive drug resistant TB (XDR-TB). It is associated with poor outcomes and risk of continued transmission (47). Treatment outcome always has been poor due to the complex drug regimen, non-availability of all the drugs, and the possible occurrence of XDR-TB. The reason for resistance is multi-factorial including patient’s non-compliance to therapy, incomplete or inadequate treatment of susceptible TB, decision error by the treating community, etc. Resistance to drugs arises from mutations which are spontaneous and restricted to specific gene loci making it detectable without much difficulty.

 

There is inconsistent data on the incidence of TB-drug resistance in diabetes. Tegegne et al in their meta-analysis concluded that DM can increase the odds of developing MDR-TB.  (47,48). Observational studies have shown delayed clearance of mycobacterium, failure of treatment, relapse and death in the presence of DM (49). The possible theoretical explanations pertaining to the influence of DM in developing resistance include lower drug concentrations, hyperglycemia induced acute and chronic effects of immune regulation, and the presence of more extensive disease in DM affected individuals (50).

 

Altered plasma concentrations of the anti-TB drug rifampicin has been demonstrated in the continuation phase (but not in the induction phase) of TB treatment in individuals with DM (51). The heavier body weight of insulin resistant individuals with DM is supposed to be one of the reasons because of the use of fixed-dose combination drugs. Giving an exact weight-based dose for a longer duration of time is suggested to overcome this hurdle (52). In addition, DM can influence the plasma concentrations of anti-TB drugs. The absorption, distribution, and metabolism of anti-TB drugs may be altered either due to local gastrointestinal causes (gastropathy, polypharmacy mediated interactions) or dysautonomia of DM predisposing to treatment failure.

 

After initiation of treatment all patients should be closely followed for evidence of resistance. Persistent sputum positivity at the end of 2-3 months of ATT therapy should prompt a look for evidence of drug resistance. CT chest features have been demonstrated to be different from non-DM subjects. Pulmonary segment consolidation and lobe consolidation seen as moth-eaten cavities without a wall and filled with fluid are the features mentioned in published data. They are accompanied by bronchial damage (53). Multiple moth-eaten cavities in chest CT while on ATT should prompt the suspicion of MDR-TB. Rapid molecular techniques like CB-NAAT or sputum culture for sensitivity should be used to look for drug resistance. All MDR-TB should be referred to specialized centers dealing with MDR-TB.

 

Drug Therapy in Multi-Drug Resistant Tuberculosis

 

The second line drugs are generally less efficacious and more toxic. In the presence of drug resistance second line agents are used in multiple combinations to address different pharmacological targets in the mycobacterium. In addition to one of the first line drugs to which there is retained susceptibility, an injectable agent, a fluroquinolone and class 4 and class 5 drugs are used in combination to combat MDR-TB (53). Bedaquiline and Delamanidare are new anti-TB drugs used in MDR-TB

 

Bedaquiline belongs to the diarylquinoline group. It inhibits mycobacterial ATP-synthase activity. A shorter time to sputum conversion compared to placebo has been demonstrated. The adverse effects include enzyme induction,electrolyte imbalance, QTc prolongation, and gastrointestinal toxicity (54).

 

Delamanid a dihydro-imidazooxazole that inhibits mycolic acid synthesis also has shown a shorter time period taken for sputum conversion while used in the regimen for MDR-TB. A dose dependent association with QT prolongation occurs (55).

 

Linezolid an oxazolidinone has demonstrated 87% sputum conversion but more than 80% of patients had adverse effects. Peripheral neuropathy, myelosuppression, optic neuropathy, and rhabdomyolysis have been documented in study subjects.

 

ANTI-DIABETES THERAPY IN TUBERCULOSIS    

 

The outcome of TB in DM is also dependent on good glycemic control. The management of hyperglycemia in TB depends on many factors like age, duration of diabetes, presence of complications of DM and co-morbidities, existing drugs, patient support and preferences, economic background and access to medical facilities.

 

Changes in the lifestyle pattern is once again reiterated when starting ATT. Adequate nutrition with high quality protein without affecting glycemic control is the corner stone of managing nutrition associated glycemic status in TB. In the presence of nephropathy or liver disease, the protein intake has to be modified appropriately and spurious use of protein is avoided. Vitamins particularly pyridoxine (B6), methylcobalamine (B12), vitamin A and Vitamin D should be adequately replaced. Tobacco use and alcohol consumptions have to be stopped. Moderate intensity exercise can help weight lose and improve glycemic control in overweight individuals.

 

Drug regimen, monitoring and follow-up should be individualized to maximize benefit with minimal discomfort and side effects like hypoglycemia, arrhythmias etc. Monitoring of capillary blood glucose at home, dose adjustment, monitoring of renal and liver functions are required during follow-up.

 

Anti-TB drugs can influence the metabolism of anti-diabetic medications (Table 3). Rifampicin, by cytochrome p450 enzyme induction, increases the metabolism of most oral anti-diabetic drugs, which may worsen the hyperglycemia. INH on the other hand inhibits cytochrome P450 enzymes and prolongs the effect of anti-diabetic drugs.

 

Table 3. Anti TB Drugs and Drug Metabolism (Ref 57-59)

Anti TB drug

Effect on Cytochrome P 450

Effect on anti- diabetic drugs

Rifampicin

Induces the cytochrome enzymes thereby accelerating the elimination of drugs like sulphonylureas, thiazolidinediones, and meglitinides

Reduced effect of sulphonylureas by one-third due to CYP2C-mediated accelerated metabolism leading to hyperglycemia

 

Reduced effect of thiazolidinedione by half due to CYP2C8-mediated accelerated metabolism leading to hyperglycemia

INH

Inhibits

Reduced elimination through action on CYP2C9; persistent effect and risk of hypoglycemia

Bedaquiline

Enzyme inducer

Can reduce the effect of anti- diabetic drugs

 

 

Aggressive therapy of DM is necessary for optimal response to TB therapy (Table 4). Insulin is the drug of choice in most illnesses including TB; insulin has the advantage of producing an anabolic effect, positive influence on appetite, and faster relief of hyperglycemic symptoms. It is the most suitable anti-hyperglycemic agent in cachexic and low BMI individuals. It is a mandatory therapeutic agent in Type 1 diabetes, pancreatic diabetes, severe DM and TB, coexisting renal or hepatic diseases, situations complicated by drug interactions and oral drug intolerance. Insulin is neutral in drug-to-drug interactions and achieves glycemic control faster than oral drugs. In the presence of fasting hyperglycemia of > 200 with ketonuria, Insulin is used to treat the hyperglycemia. It is also the preferred drug in renal impairment. The main disadvantage with insulin is that it has to be injected and more than twice a day in presence of infection/ stress. The indications for insulin use in patients with active TB and DM is summarized in Figure 7.

Figure 7. Indications of Insulin Use in patients with Type 2 DM and Active TB

Metformin has the advantage of not producing hypoglycemia when used alone. It can however reduce appetite and needs caution while being used in renal or hepatic dysfunction. It doesn’t interact much with ATT and doesn’t influence cytochrome enzyme induced metabolism. It can help to shorten the course of TB therapy. It modifies the immune response and inflammation. It acts on the mitochondrial respiratory chain and can reduce the intracellular growth by acting through AMPK pathway which has a negative impact on the inflammatory process.

 

Table 4. Anti-Diabetic Drug Use in Patients with Tuberculosis

Drug

Advantages

Disadvantages

Comment

 

Insulin

Increases appetite,

weight; anabolic effect; No drug interactions with ATT

Injectable

 

Needs supervision for change in requirement

Preferred in lean diabetes, secondary diabetes, severe DM with ketosis or hyperosmolar state

Metformin

Oral;

Cost-effective and easily available

Not influenced by ATT; positive anti-TB adjuvant action

Gastrointestinal disturbances;

Needs renal and hepatic function monitoring; Change in eGFR (< 35 ml/ min/l) or > 3 times raised liver enzymes necessitates stopping metformin;

Not suitable in hypoperfusion states (risk of lactic acidosis)

Not potent in severe hyperglycemic situations. Can be used in mild forms of hyperglycemia.

Sulphonylureas

Quick restoration of euglycemia

Long-acting drugs can induce hypoglycemia in anorexic people

Shorter acting sulphonylureas such as gliclazide and glipizide

DPP4- inhibitors

Less hypoglycemic potential

? Risk of immune dysregulation – respiratory

Infection

Selective use

Alpha glucosidase inhibitors

No hypoglycemia

GI intolerance

In mild post meal elevation

Thiazolidinediones

Against insulin resistance

Hepatic

Selective use

SGLT2

inhibitors

No hypoglycemia

 

Dehydration, DKA

Selective use

 

 

Metformin increases the host cell production of reactive oxygen species and acidification of mycobacterial phagosome (60). It has been found to downregulate oxidative phosphorylation, mammalian target of rapamycin (mTOR) signaling, and type I interferon response pathways (61).  Sulphonylureas can be used for quick glycemic control. The long acting sulphonylureas like glibenclamide and glimepride have a risk of prolonged hypoglycemia particularly seen in anorexic individuals. Their plasma concentration and their duration of action are modified by drugs acting on cytochrome P450 system and hence monitoring of glycemic status is essential while on these drugs. Thiazolidinediones (pioglitazone) do not produce GI symptoms and are non-hypoglycemic when used as monotherapy. Monitoring of hepatic enzymes is required particularly when TB drugs are co-administered. Alpha-glucosidase inhibitors may be helpful in mild postprandial hyperglycemias. SGLT-2 inhibitors have the risk of further weight loss, euglycemic ketoacidosis, worsening of dehydration in sick patients, and urinary tract infections. They should not be used for glycemic control in patients who are sick and cachexic. In individuals who have no contra-indications they can be continued selectively under close follow-up (62, 63).

 

CO-MORBIDITIES OF DIABETES

 

Cardiovascular Disease in Diabetes and Tuberculosis

 

The commonest cause of mortality in DM is cardiovascular disease due to atherosclerosis manifesting as coronary heart disease (myocardial infarction /cardiac failure), stroke and peripheral vascular disease. TB also has a possible role in chronic vascular inflammation, autoimmunity and inhabitation of TB bacilli atheromatous plaque (64). It also affects the myocardium (65). After successful initiation of TB treatment which is done on a priority basis, DM and cardiovascular status should be assessed. In addition to hyperglycemia management, lifestyle modification, antihypertensive treatment, lipid-lowering therapy, and anti-platelet therapy are the corner stones of management of cardiovascular disease in DM irrespective of the presence or absence of TB. In the presence of hemoptysis anti-platelet drugs are withdrawn or held back. Cessation of smoking and moderation of alcohol has to be counselled about. Anti-hypertensive therapy is initiated during review visits and titrated. Anti-lipid therapy using statins are added gradually and the liver enzymes should be monitored during the course of therapy while on ATT.

 

Renal Dysfunction in Diabetes and Tuberculosis

 

More than a third of individuals with DM develop renal complications due to diabetes. This complicates TB in many ways including increased susceptibility to TB and difficulty in the management of TB (66). In patients with chronic renal failure and on dialysis there is a 6.9‐ to 52.5‐fold risk of developing TB (67). Peritoneal TB is another risk for CKD patients on peritoneal dialysis. The altered immune function in chronic renal impairment increases susceptibility to TB (68). CKD adversely affects TB and its treatment. The anti-TB drugs (ethambutol and pyrazinamide) require dose reduction by up to 50%. Insulin is preferred in most instances for glycemic control. Short acting sulphonylureas or repaglinide can be used as an alternative.

 

Hepatic Dysfunction in Diabetes and Tuberculosis

 

DM liver pathology includes fatty liver disease, NASH, and cirrhosis. In TB, drug induced hepatitis is a concern. In such cases the drugs are withdrawn until resolution of hepatotoxicity. Pyrazinamide is withdrawn completely. Quinolones, ethambutol, and ofloxacin can be used instead of the first line agents. Anti-TB drugs are restarted when the liver enzymes are normalized. Insulin is the drug of choice in severe chronic liver disease with diabetes. Metformin is preferred in fatty liver but withdrawn in cirrhosis.

 

Tuberculosis and Diabetes in HIV – The Triangular Overlap

 

DM and HIV are two independent risk factors for developing TB. The wider prevalence of DM makes DM a more important risk factor for developing TB than retroviral diseases. A significantly higher preponderance of DM over HIV has been reported in cases of pulmonary TB while extra-pulmonary TB was predominant in HIV-TB patients (69). DM is increasing in areas where TB and HIV are rampant. Literature reports on the influence of HIV-co infection on DM with TB have been contradictory. Studies have reported reduced odds of developing DM in HIV infected TB patients (70).  A paradoxical protective effect of HIV on the development of TB was reported (71). However, they were cross-sectional single center studies of limited sample size and had used single random blood glucose tests for screening. But in a case-control study the association between DM and TB in HIV was found to be strong except when HbA1c was used for screening. This may be because of anemia of HIV compromising the true HbA1c value (72). HIV testing is mandatory in all presumptive TB and confirmatory assessment using Gene Xpert MTB/RIF assay for drug resistance.

 

PREVENTIVE METHODS

 

Aggressive DM screening among the population with TB and effective management of both TB and DM can improve outcome on an individual basis. The more effective approach to have an impact at the community level is to have a preventive strategy like vaccination against TB and aggressive prevention and management of DM (29).

 

SUMMARY

 

Epidemiological evidence for an uncharacteristic alliance between non-communicable disease like DM and a communicable disease like TB as a syndemic are overwhelming. A two-to four-fold higher risk of active TB in individuals with DM and twice the prevalence of DM in patients with TB explains the double burden. Ranked among the top ten mortal diseases, the geographical spread of both these diseases is unfortunately overlapping and the co-existence is progressive. They are increasing alarmingly in those regions where health care facilities are limited. DM impacts TB both from infection to disease stage and disease stage to progression stage. Chronic hyperglycemia compromises the alveolar defenses.

 

Latent TB infection is a dormant subclinical disease which lasts for weeks or decades. Immune deficiency either in absolute or relative quantities are sufficient for its re-activation. The clinical and radiological presentations of active TB have been reported to be pulmonary predominantly and atypical when DM co-exists with TB. DM increases the risk of treatment failure, death and relapse. The risk ratio for combined treatment failure and death was 1.69, unadjusted and adjusted risk ratios for death were 1.89 and 4.95 respectively and risk ratio for relapse was 3.89.

 

Bidirectional screening is recommended to improve the outcome in both diseases. Sputum microscopic examination, rapid molecular tests, random plasma glucose and glycosylated hemoglobin are the available among the screening tests with their own merits and limitations.

 

Anti-TB treatment has adverse impact on glycemic control and the complications of diabetes. The metabolism of some DM drugs is modified by ATT which affects glycemic control. Modifications of medications are required in co-morbid illnesses of DM like cardio-vascular diseases, renal, and hepatic dysfunction. Insulin is the drug of choice in lean diabetes, severe hyperglycemia, ketotic states, anorexic patients, and in drug intolerance. Metformin if tolerated has an advantage as a non-hypoglycemic agent with some favorable anti-TB activity.

 

Lifestyle modification, antihypertensive treatment, lipid-lowering therapy, and anti-platelet therapy are the cornerstones in the management of cardiovascular disease in DM and TB. Anti-platelet drugs are withdrawn in patients with hemoptysis. Chronic kidney diseases can predispose to TB and its management is complicated by drug toxicity and dose adjustment of ATT is required along with careful monitoring of response and renal functions. In case of ATT induced hepatotoxicity, ATT is withdrawn and second line agents are substituted. Once liver enzymes normalize the first line drugs are restarted cautiously.

 

Effective preventive strategies like vaccination against TB and aggressive prevention and management of DM will be the approach to be adopted till time throws new light on the means to fight these dual epidemics more effectively.

 

REFERENCES

 

  1. Whiting DR, Guariguata L, Weil C, Shaw J. IDF diabetes atlas: global estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Res Clin Pract. 2011; 94(3):311-321.
  2. Jacob JJ, Paul PAM. Infections in Endocrinology: Tuberculosis. In: Feingold KR, Anawalt B, Boyce A, et al., eds. Endotext. South Dartmouth (MA): MDText.com, Inc.; March 14, 2021.
  3. Lönnroth K, Castro KG, Chakaya JM, et al. Tuberculosis control and elimination 2010-50: cure, care, and social development. Lancet. 2010; 375(9728):1814-1829. doi:10.1016/S0140-6736(10)60483-7
  4. World Health Organization. Global Tuberculosis Report 2020. https://apps.who.int/iris/bitstream/handle/10665/336069/9789240013131-eng.pdf (Accessed 28 Nov 2020)
  5. Narasimhan P, Wood J, Macintyre CR, Mathai D. Risk factors for tuberculosis. Pulm Med. 2013; 2013:828939. doi:10.1155/2013/828939
  6. Getahun H, Matteelli A, Abubakar I, et al. Management of latent Mycobacterium tuberculosis infection: WHO guidelines for low tuberculosis burden countries. Eur Respir J. 2015; 46(6):1563-1576. doi:10.1183/13993003.01245-2015
  7. Lee RE, Li W, Chatterjee D, Lee RE. Rapid structural characterization of the arabinogalactan and lipoarabinomannan in live mycobacterial cells using 2D and 3D HR-MAS NMR: structural changes in the arabinan due to ethambutol treatment and gene mutation are observed. Glycobiology. 2005; 15(2):139-151. doi:10.1093/glycob/cwh150
  8. Emerging Risk Factors Collaboration, Sarwar N, Gao P, et al. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies [published correction appears in Lancet. 2010 Sep 18;376(9745):958. Hillage, H L [corrected to Hillege, H L]]. Lancet. 2010; 375(9733):2215-2222. Doi: 10.1016/S0140-6736(10)60484-9.
  9. International Diabetes Federation. IDF Diabetes Atlas, 8th edn. Brussels, Belgium:International Diabetes Federation, 2019. https://diabetesatlas.org/en/resources/(Accessed 8 March 2021)
  10. World Health Organization. Global tuberculosis report 2019: World Health Organization; 2019. https://www.who.int/tb/publications/global_report/en/ (Accessed 8 March 2021)
  11. Dooley KE, Chaisson RE. Tuberculosis and diabetes mellitus: convergence of two epidemics. Lancet Infect Dis. 2009 Dec; 9(12):737-46. Doi: 10.1016/S1473-3099(09)70282-8. PMID: 19926034; PMCID: PMC2945809.
  12. Viswanathan, V., Bajaj, S., Kalra, S. et al. RSSDI clinical practice recommendations for diagnosis, prevention, and control of the diabetes mellitus-tuberculosis double burden. Int J Diabetes Dev Ctries 2017; 37, 379–399
  13. Goldhaber-Fiebert JD, Jeon CY, Cohen T, Murray MB. Diabetes mellitus and tuberculosis in countries with high tuberculosis burdens: individual risks and social determinants. Int J Epidemiology. 2011 Apr; 40(2):417-28. Doi: 10.1093/ije/dyq238. Epub 2011 Jan 20. PMID: 21252210; PMCID: PMC3621385.
  14. World Health Organization/International Union Against Tuberculosis and Lung Disease. Provisional collaborative framework for care and control of tuberculosis and diabetes. WHO/HTM/TB/2011.15. Geneva, Switzerland: WHO, 2011.
  15. Hayashi, S., &Chandramohan, D. (2018). Risk of active tuberculosis among people with diabetes mellitus: systematic review and meta-analysis. Tropical medicine & international health: TM & IH, 23(10), 1058–1070. https://doi.org/10.1111/tmi.13133
  16. Workneh MH, Bjune GA, Yimer SA. Prevalence and associated factors of tuberculosis and diabetes mellitus co morbidity: A systematic review. PLoS One. 2017; 12(4):e0175925. Published 2017 Apr 21. doi:10.1371/journal.pone.0175925.
  17. Odone A, Houben RM, White RG, Lönnroth K. The effect of diabetes and under nutrition trends on reaching 2035 global tuberculosis targets. Lancet Diabetes Endocrinol. 2014; 2(9):754-764. doi:10.1016/S2213-8587(14)70164-0
  18. Jeon CY, Harries AD, Baker MA, et al. Bi-directional screening for tuberculosis and diabetes: a systematic review. Trop Med Int Health. 2010; 15(11):1300-1314. doi:10.1111/j.1365-3156.2010.02632.
  19. Lönnroth K, Roglic G, Harries AD. Improving tuberculosis prevention and care through addressing the global diabetes epidemic: from evidence to policy and practice. Lancet Diabetes Endocrinol. 2014; 2(9):730-739. doi:10.1016/S2213-8587(14)70109-3
  20. Noubiap JJ, Nansseu JR, Nyaga UF, et al. Global prevalence of diabetes in active tuberculosis: a systematic review and meta-analysis of data from 2·3 million patients with tuberculosis. Lancet Glob Health. 2019; 7(4):e448-e460. doi:10.1016/S2214-109X(18)30487
  21. Collaborative framework for care and control of tuberculosis and diabetes. WHO/HTM/TB/2011.15
  22. Viswanathan V, Kumpatla S, Aravindalochanan V, et al. Prevalence of diabetes and pre-diabetes and associated risk factors among tuberculosis patients in India. PLoS One. 2012;7(7):e41367.
  23. Balakrishnan S, Vijayan S, Nair S, et al. High diabetes prevalence among tuberculosis cases in Kerala, India. PLoS One. 2012;7(10):e46502.
  24. Manjareeka M, Palo SK, Swain S, Pati S, Pati S. Diabetes Mellitus among Newly Diagnosed Tuberculosis Patients in Tribal Odisha: An Exploratory Study. J Clin Diagn Res. 2016;10(10):LC06-LC08.
  25. Singh SP, Singh SP, Kishan J, Kaur S, Ramana S. Association of tuberculosis and diabetes Mellitus: an analysis of 1000 consecutively admitted cases in a tertiary care hospital of North India. Pan Afr Med J. 2016;24:4
  26. Kodiatte A, John M, Jacob JJ. Diabetes mellitus and prediabetes among patients with tuberculosis in a single north Indian tertiary care centre. J R Coll Physicians Edinb. 2020;50(3):242-246.
  27. Noubiap JJ, Nansseu JR, Nyaga UF, Nkeck JR, Endomba FT, Kaze AD, Agbor VN, Bigna JJ. Global prevalence of diabetes in active tuberculosis: a systematic review and meta-analysis of data from 2·3 million patients with tuberculosis. Lancet Glob Health. 2019 Apr;7(4): e448-e460.
  28. Alisjahbana B, Sahiratmadja E, Nelwan EJ, et al. The effect of type 2 diabetes mellitus on the presentation and treatment response of pulmonary tuberculosis. Clin Infect Dis. 2007; 45(4):428-435. doi:10.1086/519841
  29. Crevel RV, Critchley JA. The Interaction of Diabetes and Tuberculosis: Translating Research to Policy and Practice. Trop Med Infect Dis. 2021; 6(1):8. Published 2021 Jan 8. doi:10.3390/tropicalmed6010008
  30. Chiang CY, Lee JJ, Chien ST, et al. Glycemic control and radiographic manifestations of tuberculosis in diabetic patients. PLoS One. 2014; 9(4):e93397. Published 2014 Apr 3. doi:10.1371/journal.pone.0093397
  31. Jiménez-Corona ME, Cruz-Hervert LP, García-García L, et al. Association of diabetes and tuberculosis: impact on treatment and post-treatment outcomes. Thorax. 2013; 68(3):214-220. Doi: 10.1136/thoraxjnl-2012-201756.
  32. Riza AL, Pearson F, Ugarte-Gil C, et al. Clinical management of concurrent diabetes and tuberculosis and the implications for patient services. Lancet Diabetes Endocrinol. 2014; 2(9):740-753. doi:10.1016/S2213-8587(14)70110
  33. Restrepo BI, Schlesinger LS. Impact of diabetes on the natural history of tuberculosis. Diabetes Res Clin Pract. 2014; 106(2):191-199. doi:10.1016/j.diabres.2014.06.011.
  34. Baker MA Harries AD, Jeon CY, et al. The impact of diabetes on tuberculosis treatment outcomes: a systematic review. BMC Med. 2011; 9:81. Published 2011 Jul 1. doi:10.1186/1741-7015-9-81
  35. Chang JT, Dou HY, Yen CL, et al. Effect of type 2 diabetes mellitus on the clinical severity and treatment outcome in patients with pulmonary tuberculosis: a potential role in the emergence of multidrug-resistance. J Formos Med Assoc. 2011; 110(6):372-381. doi:10.1016/S0929-6646(11)60055-7
  36. Collaborative framework for care and control of tuberculosis and diabetes. WHO/HTM/TB/2011.15
  37. Sreeramareddy CT, Panduru KV, Menten J, Van den Ende J. Time delays in diagnosis of pulmonary tuberculosis: a systematic review of literature. BMC Infect Dis. 2009; 9:91. Published 2009 Jun 11. doi:10.1186/1471-2334-9-91
  38. Getahun H, Matteelli A, Abubakar I, et al. Management of latent Mycobacterium tuberculosis infection: WHO guidelines for low tuberculosis burden countries. Eur Respir J. 2015; 46(6):1563-1576. doi:10.1183/13993003.01245-2015]
  39. US Preventive Services Task Force, Bibbins-Domingo K, Grossman DC, et al. Screening for Latent Tuberculosis Infection in Adults: US Preventive Services Task Force Recommendation Statement. JAMA. 2016; 316(9):962-969. doi:10.1001/jama.2016.11046
  40. Mansilla Bermejo MJ, Sanz Gil MJ, Moraleda Velasco P, Alvárez Prado A, Carbayo García JJ, Mata Guijarro F. Lectura de la prueba de la tuberculinaenpacientesdiabéticos de un centro de salud [Tuberculin test in diabetic patients in a health centre]. Aten Primaria. 1995; 16(3):154-157.
  41. Vega Torres RA, Conde JG, Díaz M. Prevalence of tuberculin reactivity and risk factors for the development of active tuberculosis upon admission to a nursing home. P R Health Sci J. 1996; 15(4):275-277
  42. Nwabudike LC, Ionescu-Tîrgovişte C. Intradermal reactions to purified protein derivative in patients with diabetes mellitus. Rom J Intern Med. 2005; 43(1-2):127-132.
  43. Dabhi PA, Thangakunam B, Gupta R, et al. Screening for prevalence of current TB disease and latent TB infection in type 2 diabetes mellitus patients attending a diabetic clinic in an Indian tertiary care hospital. PLoS One. 2020; 15(6):e0233385. Published 2020 Jun 5. doi:10.1371/journal.pone.0233385
  44. Kumpatla S, Aravindalochanan V, Rajan R, Viswanathan V, Kapur A. Evaluation of performance of A1C and FPG tests for screening newly diagnosed diabetes defined by an OGTT among tuberculosis patients-a study from India. Diabetes Res Clin Pract. 2013; 102(1):60-64. doi:10.1016/j.diabres.2013.08.007]
  45. Sariko ML, Mpagama SG, Gratz J, Kisonga R, Saidi Q, Kibiki GS, Heysell SK. Glycated haemoglobin screening identifies patients admitted for retreatment of tuberculosis at risk for diabetes in Tanzania. J Infect Dev Ctries. 2016 Apr 28; 10(4):423-6. Doi: 10.3855/jidc.7324. PMID: 27131008; PMCID: PMC4869164.
  46. Naik B, Kumar AM, Satyanarayana S, et al. Is screening for diabetes among tuberculosis patients feasible at the field level? Public Health Action. 2013; 3(Suppl 1):S34-S37. doi:10.5588/pha.13.0022
  47. Tegegne BS, Habtewold TD, Mengesha MM, Burgerhof JG. Association between diabetes mellitus and multi-drug-resistant tuberculosis: a protocol for a systematic review and meta-analysis. Syst Rev. 2017; 6(1):6. Published 2017 Jan 14. Doi: 10.1186/s13643-017-0407-9.
  48. Pérez-Navarro LM, Fuentes-Domínguez F, Morales-Romero J, Zenteno-Cuevas R. Factoresasociados a tuberculosis pulmonarenpacientes con diabetes mellitus de Veracruz, México [Factors associated to pulmonary tuberculosis in patients with diabetes mellitus from Veracruz, México]. Gac Med Mex. 2011; 147(3):219-225.
  49. Kang YA, Kim SY, Jo KW, et al. Impact of diabetes on treatment outcomes and long-term survival in multidrug-resistant tuberculosis. Respiration. 2013; 86(6):472-478. doi:10.1159/000348374
  50. Ruslami R, Aarnoutse RE, Alisjahbana B, van der Ven AJ, van Crevel R. Implications of the global increase of diabetes for tuberculosis control and patient care. Trop Med Int Health. 2010; 15(11):1289-1299. doi:10.1111/j.1365-3156.2010.02625.
  51. Nijland HM, Ruslami R, Stalenhoef JE, et al. Exposure to rifampicin is strongly reduced in patients with tuberculosis and type 2 diabetes. Clin Infect Dis. 2006; 43(7):848-854. doi:10.1086/507543
  52. Van Ingen J, Aarnoutse RE, Donald PR, et al. Why Do We Use 600 mg of Rifampicin in Tuberculosis Treatment? Clin Infect Dis. 2011; 52(9):e194-e199. doi:10.1093/cid/cir184.
  53. Song Q, Zhang G, Jiang H, Ren Y, Lu X. Imaging Features of Pulmonary CT in Type 2 Diabetic Patients with Multidrug-Resistant Tuberculosis. PLoS One. 2016; 11(3):e0152507. Published 2016 Mar 29. doi:10.1371/journal.pone.0152507
  54. Kurz SG, Furin JJ, Bark CM. Drug-Resistant Tuberculosis: Challenges and Progress. Infect Dis Clin North Am. 2016; 30(2):509-522. doi:10.1016/j.idc.2016.02.010
  55. U.S. Food and Drug Administration. SIRTURO Prescribing Information.http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/204384s000lbl.
  56. Shimokawa Y, Sasahara K, Yoda N, Mizuno K, Umehara K. Delamanid does not inhibit or induce cytochrome p450 enzymes in vitro. Biol Pharm Bull. 2014; 37(11):1727-1735. doi:10.1248/bpb.b14-00311 ]
  57. Niemi M, Backman JT, Neuvonen M, Neuvonen PJ, Kivistö KT. Effects of rifampin on the pharmacokinetics and pharmacodynamics of glyburide and glipizide. Clin Pharmacol Ther. 2001;69(6):400-406. doi:10.1067/mcp.2001.115822
  58. Niemi M, Backman JT, Neuvonen PJ. Effects of trimethoprim and rifampin on the pharmacokinetics of the cytochrome P450 2C8 substrate rosiglitazone. Clin Pharmacol Ther. 2004;76(3):239-249. doi:10.1016/j.clpt.2004.05.001
  59. Boglou P, Steiropoulos P, Papanas N, Bouros D. Hypoglycaemia due to interaction of glimepiride with isoniazid in a patient with type 2 diabetes mellitus. BMJ Case Rep. 2013;2013:bcr2012008528. Published 2013 Apr 16. doi:10.1136/bcr-2012-008528
  60. Singhal A, Jie L, Kumar P, et al. Metformin as adjunct antituberculosis therapy. Sci Transl Med. 2014; 6(263):263ra159.doi:10.1126/scitranslmed.3009885.
  61. Yu X, Li L, Xia L, et al. Impact of metformin on the risk and treatment outcomes of tuberculosis in diabetics: a systematic review. BMC Infect Dis. 2019; 19(1):859. Published 2019 Oct 17. doi:10.1186/s12879-019-4548-4
  62. Niazi AK, Kalra S. Diabetes and tuberculosis: a review of the role of optimal glycemic control. J Diabetes MetabDisord. 2012; 11(1):28. Published 2012 Dec 20. doi:10.1186/2251-6581-11-28
  63. Hsia DS, Grove O, Cefalu WT. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. CurrOpin Endocrinol Diabetes Obes. 2017; 24(1):73-79. doi:10.1097/MED.0000000000000311
  64. Huaman MA, Henson D, Ticona E, Sterling TR, Garvy BA. Tuberculosis and Cardiovascular Disease: Linking the Epidemics. Trop Dis Travel Med Vaccines. 2015; 1:10. doi:10.1186/s40794-015-0014-5
  65. Liu A, Hu Y, Coates A. Sudden cardiac death and tuberculosis - how much do we know? Tuberculosis (Edinb). 2012; 92(4):307-313. doi:10.1016/j.tube.2012.02.002
  66. CKD TB : Ates G, Yildiz T, Danis R, et al. Incidence of tuberculosis disease and latent tuberculosis infection in patients with end stage renal disease in an endemic region. Ren Fail. 2010; 32(1):91-95. doi:10.3109/08860220903367528
  67. Hussein MM, Mooij JM, Roujouleh H. Tuberculosis and chronic renal disease. Semin Dial. 2003; 16(1):38-44. doi:10.1046/j.1525-139x.2003.03010.x
  68. Levey AS, Atkins R, Coresh J, et al. Chronic kidney disease as a global public health problem: approaches and initiatives - a position statement from Kidney Disease Improving Global Outcomes. Kidney Int. 2007; 72(3):247-259. doi:10.1038/sj.ki.5002343
  69. Gupta S, Shenoy VP, Bairy I, Srinivasa H, Mukhopadhyay C. Diabetes mellitus and HIV as co-morbidities in tuberculosis patients of rural south India. J Infect Public Health. 2011;4(3):140-144. doi:10.1016/j.jiph.2011.03.005
  70. Faurholt-Jepsen D, Range N, Praygod G, et al. Diabetes is a risk factor for pulmonary tuberculosis: a case-control study from Mwanza, Tanzania. PLoS One. 2011;6(8):e24215. doi:10.1371/journal.pone.0024215
  71. Kibirige D, Ssekitoleko R, Mutebi E, Worodria W. Overt diabetes mellitus among newly diagnosed Ugandan tuberculosis patients: a cross sectional study. BMC Infect Dis. 2013; 13:122. Published 2013 Mar 5. doi:10.1186/1471-2334-13-122
  72. Boillat-Blanco N, Ramaiya KL, Mganga M, et al. Transient Hyperglycemia in Patients With Tuberculosis in Tanzania: Implications for Diabetes Screening Algorithms. J Infect Dis. 2016;213(7):1163-1172. doi:10.1093/infdis/jiv568

 

Autoimmune Polyglandular Syndromes

ABSTRACT

The autoimmune polyglandular syndromes (APS) are clusters of endocrine abnormalities that occur in discreet patterns in subjects with immune dysregulation and that permit treatment and anticipation of associated systemic or other hormonal deficiencies. Three major entities are recognized, APS1, APS2 and APS3; the rare X-linked syndrome of immunodysregulation, polyendocrinopathy, and enteropathy due to mutations in the FOXP3 gene also qualifies as an APS. An additional increasingly described category occurs in patients treated with immunoregulatory agents such as checkpoint inhibitors for cancer, so that tumor antigens that have evaded recognition can now be targeted, but at the expense of activating autoimmunity. APS1 is a syndrome characterized by chronic muco-cutaneous candidiasis, hypoparathyroidism, and primary adrenal insufficiency, as well as ectodermal dystrophy and a host of other endocrine and non-endocrine tissue involvement in autoimmune destructive processes. The underlying cause is a homozygous inactivating mutation in the autoimmune regulator gene AIRE which permits the intra-thymic expression of ectopic antigens normally expressed only in specific peripheral tissues (e.g., insulin), so that T-cells as they mature within the thymus and acquire a receptor for the self- antigen are eliminated (negative selection), thereby avoiding autoimmunity. Recent studies demonstrate that in addition to the classical homozygous mutations, single gene dominant mutations in AIRE play an important role in autoimmune regulation and its disorders. APS2 and APS3 are both due to mutations in the HLA DQ/DR regions which regulate antigen presentation to T-cell receptors; however, their genetic profile is more complex. APS2 is characterized by type 1 diabetes mellitus (T1DM), Addison Disease, and Hypothyroidism, whereas APS3 is similar but without Addison disease. In keeping with other autoimmune disorders, these entities are more frequent in females, whereas APS1 has no sexual predominance. The recent emergence of autoimmune endocrinopathies in patients treated with immunoregulatory agents for cancer adds a new dimension to considerations of autoimmune polyendocrinopathy syndromes. Rapid progress in the immunology and genetics of these entities offers the promise of potential amelioration and eventual reversal via genetic manipulation before organ damage is established.

 

AUTOIMMUNE POLYGLANDULAR SYNDROMES

 

The autoimmune polyglandular syndromes are clusters of endocrine abnormalities that occur in discreet patterns in subjects with immune dysregulation and that permit treatment and anticipation of associated systemic or other hormonal deficiencies (1-8). Three major entities are recognized, APS1, APS2, and APS3 as well as the extremely rare X-linked syndrome of immunodysregulation, polyendocrinopathy, and enteropathy. An additional but increasing category occurs in patients treated with immunoregulatory agents for cancer, by which the tumor’s blockade of immune regulatory checkpoints is inhibited, so that tumor antigens that had evaded recognition can now be targeted, but at the expense of activating autoimmunity including against endocrine organs.

 

APS1 results from a failure to eliminate T-cells that have acquired receptors with high affinity to auto-antigens, as these T-cells mature and traverse the thymic epithelium during their development. Normally, such T-cells are prevented from entering the periphery because of the ectopic expression of multiple antigens within the thymus that usually are expressed only in discrete tissues, e.g., insulin in pancreatic β-cells. A developing T-cell that acquires and expresses a high affinity receptor for insulin will be bound to the ectopically expressed insulin antigen within the thymus, undergo apoptosis and be excluded from entering the periphery to initiate auto-immunity (Fig1). This ectopic expression of antigens within the thymus is mediated by the Auto-Immune REgulator gene (AIRE) located on chromosome 21. Discovered in 1997 as the gene whose variable inactivation is responsible for the clinical entity APS1 (1, 2, 4, 6), this gene is now known to be an essential component of the adaptive immune response cascade, and the spectrum of disorders ascribed to mutations in this gene extend beyond the APS1 syndrome (2, 4). Moreover, although APS1 is a rare entity with predilection for particular populations, the increased incidence of autoimmunity in persons with trisomy 21, a relatively common genetic abnormality, may be due to abnormal function of the AIRE gene (9). In addition, it is becoming apparent that mutations in AIRE can have autosomal dominant effects and become manifest as autoimmune disorders later in life in patterns that differ from the classical APS1 (2, 4). Since the incidence of such autosomal mutations may be as high as 1:1000, whereas the incidence of APS1 is much rarer (1:9000 in Iranian Jews; 1:14500 in Sardinia; 1:25000 in Finland), the influence of the AIRE gene on autoimmune processes and diseases may be far wider than hitherto appreciated, especially since Treg cells (regulatory T cells) also are now implicated in the abnormalities induced by AIRE deficiency (2, 4). Both T-cell and B-cell abnormalities are observed in APS1, so that circulating antibodies to various hormonal, connective tissues, and protein antigens such as enzymes in the steroidogenic or thyroid synthesis cascades are evident in the serum of affected patients with APS1, and indeed in all forms of the autoimmune polyglandular syndromes. Figure 1 summarizes these concepts.

 

Several recent case series indicate that the phenotypic variation and age of symptom onset vary greatly, even within the same family (10,11), implying that other genes such as major histocompatibility complex genes, or environmental exposures, influence the phenotype and natural course (1,11). This wide variation in presentation and symptomatology makes the diagnosis of APS-1 challenging.

 

Figure1. Left panel: Modified from Autoimmune Polyglandular Syndromes in Pediatric Endocrinology 4th Edition, Ed. Sperling MA. (with permission of the authors Drs. Michael Haller, William Winter, Desmond Schatz) A developing T-cell migrates from its origins in the bone marrow to the thymus where it acquires its repertoire of receptors. The expression of self-antigens, including ectopic expression of antigens mediated via the AIRE gene, results in apoptosis of the T-cell possessing the complementary receptor, and prevention of the T-cell entering the periphery, a fate of almost all T-cells. A small fraction of T-cells enter the periphery where they remain anergic to self-antigens, but can mount an immune response to non-self-antigens. Right panel: Failure of self-tolerance, due to non-expression of self-antigens as would occur with inactivating mutations of the AIRE gene, results in failure of central tolerance as well as failure of recognition of self-antigens that leads to an auto-immune response.

 

APS2 is characterized by the triad of T1DM, adreno-cortical insufficiency, and hypothyroidism as a result of autoimmunity to components of the pancreatic β-cell, adrenal cortex, and thyroid synthesizing machinery. APS3 is essentially identical to APS2 except that adreno-cortical insufficiency is absent. This similarity has led some investigators to label the former as APS2a and the latter as APS2b. Whereas APS2a is rare, APS2b is relatively common, as approximately 20% of patients with T1DM harbor circulating antibodies to thyroid synthesis components, namely thyroid peroxidase (TPO) and thyroglobulin (TGB), markers associated with Hashimoto thyroiditis. Note however that the presence of autoantibodies is not necessarily predictive of glandular failure and its clinical manifestations. The genes responsible for the disordered immunity in APS2 and APS3 are in the DQ and DR regions of the HLA complex on the short arm of chromosome 6; specific alleles or mutations facilitate the presentation of antigens co-expressed with the particular HLA complex by antigen presenting cells such as dendritic cells and macrophages. This facilitated presentation of self-antigens, along with other regulatory factors such as lower expression of T-regulatory cells, initiate auto- immunity. Consistent with the generally heightened immune responses in females, these forms of autoimmune endocrine disorders are significantly more prevalent in women, whereas in APS1 the sex distribution is equal.

 

AUTOIMMUNE POLYGLANDULAR SYNDROME 1 (APS1-APECED)

 

APS1 is characterized by 3 classical features; muco-cutaneous candidiasis, hypoparathyroidism with hypocalcemia hyperphosphatemia and low PTH concentrations, and Addison disease with cortisol deficiency, occasional aldosterone deficiency, and marked elevations in adrenocorticotropic hormone (ACTH). Clinical manifestations of primary adrenal insufficiency include hyperpigmentation, abdominal pain, vomiting, weight loss, and electrolyte disturbances, as well as hypoglycemia with fasting. Two of the 3 classical features are required to make a diagnosis of APS1. Other manifestations include periodic rash with fever, kerato-conjunctivitis, chronic diarrhea, primary gonadalfailure occurring pre-or post-puberty, Hashimoto thyroiditis with hypothyroidism, Vitamin B12 deficiency, chronic active hepatitis, (T1DM), and ectodermal dystrophy-hence the term APECED (Autoimmune Poly Endocrinopathy, Candidiasis, Ectodermal Dystrophy).The features of ectodermal dystrophy include enamel hypoplasia affecting only the permanent teeth, pitted nail dystrophy unrelated to candidiasis of the nails, and visible alterations in the tympanic membranes characterized by calcium deposits. Iritis, optic atrophy, and skin changes termed keratopathy, as well as alopecia and vitiligo also are reported (1). Most affected patients manifest their problem(s) by 5 years of age; non-endocrine manifestations precede the endocrine manifestations in about 75% of cases, with mucocutaneous, including oral, candidiasis as the first manifestation in about 60% and malabsorption in about 10%, and vitiligo, alopecia, hepatitis and keratopathy in about 5% of affected subjects (see table 1). Mucocutaneous candidiasis, the most common nonendocrine manifestation, occurs due to defective receptor- mediated internalization of Candida by monocytes as well as decreased kinase activation (12). In these subjects, the median interval to an endocrine manifestation is about 4 years with a range from 0.1-33 years.

 

When an endocrine disorder is the first manifestation it is almost invariably hypoparathyroidism; overall about 70%-80% develop hypoparathyroidism, and in those who develop hypoparathyroidism first, about 60% develop Addison disease. If Addison disease is the first manifestation, about a third also develop hypoparathyroidism. The manifestations vary in sequence and age at onset; the description of all known Finnish cases in 2006 by Peerhentupa (1) remains one of the most detailed series description and indicates the remarkable heterogenous pattern with the 3 most common being muco-cutaneous candidiasis (~80%), followed by hypoparathyroidism (~80%), and Addison disease (~70%). Ovarian failure occurs in about 60% of affected females but testicular failure only occurs in about 15% of males; parietal cell atrophy with atrophic gastritis and B12 deficiency, and T1DM occur in only about 12% of patients, with diabetes a late complication in comparison to the early manifestations of parathyroid and adrenal deficiency; although anti-thyroid antibodies are common, hypothyroidism only develops in about 5% of affected patients. Rare manifestations include diabetes insipidus, growth hormone deficiency secondary to hypophysitis, and infertility due to sperm antibodies in males and ovarian failure in females. In most patients with APS1, disease manifestations develop earlier than in APS2, as noted above, and are usually more severe than in APS-2. Typically, a given APS-1 patient develops an average of 4–5 manifestations of the syndrome, but may have as few as one or as many as twenty. Due to chronic mucocutaneous candidiasis, patients are also susceptible to squamous carcinoma of the oral mucosa and esophagus over time. In general, patients with APS-1 have an increased mortality risk, due to cancer, adrenal and hypocalcemic crises, and certain conditions induced by aberrant autoimmune responses, particularly hepatitis, nephritis and pneumonitis (13).

 

Circulating antibodies against components of the parathyroid, adrenal, and thyroid glands as well as those of the pancreatic islets are hallmarks of this disease which affects T-cell as well as B- cell function. Although lymphocytic infiltration of the parathyroid glands is frequent, the protein NALP5 that serves as the antigen for the immune response was not discovered until 2008 (5). Antibodies to NALP5 (NACHT leucine-rich-repeat protein 5) were found tobe highly specific and present only in those with hypoparathyroidism as part of APS1, but absent in other forms of autoimmune syndromes (5) or patients with APS1 but without hypoparathyroidism. Antibodies against adrenal cytochrome P450 enzymes such as Cyp21, Cyp17, and Cyp11A1 are present in many patients but wane with glucocorticoid treatment. Circulating antibodies to GAD 65 and IA2 may be present but are not strong predictors for the development of T1DM. Thyroid peroxidase and anti-thyroglobulin antibodies also are common but not predictive for development of hypothyroidism. Antibodies against liver microsomal proteins, against parietal cells (α & β subunits of H+/K+ ATPase), and against intrinsic factor also are reported. Other less common autoantibodies observed in APS-1 include BPI Fold Containing Family B Member 1 (BPIFB1), the potassium channel regulator KCNRG, expressed in the lung, and transglutaminase-4, expressed solely in the prostate gland (14, 15, 16).  

 

A unique feature is the presence of autoantibodies that neutralize type1 interferon, mostly interferon1α and 1ω; these antibodies appear to be specific for this entity and therefore have clinical diagnostic utility (17). Since over 95% of patients with APS-1 have autoantibodies to type 1 interferons, it has been proposed that evaluating the presence of these interferon antibodies should be part of the diagnostic evaluation of patients suspected of harboring APS1. In addition, patients with AIRE mutations possess high-affinity disease-ameliorating autoantibodies, which may explain the low incidence and late appearance of T1DM in patients with APS1 (18). In contrast to the autoantibodies mentioned above, systemic autoantibodies to certain cytokines are highly prevalent in many, if not most, APS-1 patients. Autoantibodies to the interleukin (IL) 17 family of cytokines, especially IL-22 are also prevalent in APS-1, exceeding 90% in some series (4).

 

The cause of this autoimmunity are inactivating mutations in the autoimmune regulator gene (AIRE) on chromosome 21q22.3, which normally acts to permit ectopic expression in the thymus of numerous tissue restricted hormonal and other peripheral antigens, so that developing T-cells that acquire high affinity receptors for these antigens as the developing T-cell traverses the thymic epithelium are eliminated and do not enter the periphery to cause auto-immunity (2, 4, 6, 7). For the classic case, this is an autosomal recessive inherited disorder; however, point mutations resulting in an autosomal dominant form have been reported, albeit this autosomal dominant form seems less severe than the classic autosomal recessive disease, suggesting that this genetic disorder may be more prevalent in various immune disorders hitherto not considered to be due to AIRE mutations (2, 4, 19,11).

 

The structure of the AIRE gene, the sites of autosomal dominant and autosomal recessive mutations, their influence on the expression and function of the gene and its consequences, are elegantly discussed in recent reviews (2,4,19,11). To date, over 100 different disease-causing mutations have been reported. The most common is the so-called Finnish major mutation p.R257X, located in the SAND-domain (named after a range of proteins in the protein family: Sp100, AIRE-1, NucP41/75, DEAF-1). The Finnish major mutation is especially prevalent in people in Finland, Russia, and Eastern Europe (20). Another common mutation is the so-called 13 base pair deletion (p.C322del13) in the histone protein reading region called plant homeodomain 1 (PHD1), prevalent in persons in Norway, the British Isles, France, and North-America (10, 21). Additionally, patients with unique dominant negative mutations in AIRE with autosomal dominant inheritance have recently been identified. These dominant negative mutations are associated with milder disease, often with accompanying pernicious anemia, vitiligo, autoimmune thyroid disease, and T1DM, and can be confused with the much more common condition, APS-2, which has a complex inheritance. The dominant gene variants are located both in the PHD1 and SAND domain (22, 23). Recent findings indicate that AIRE controls immune tolerance by an additional mechanism—the induction of a unique population of FOXP3-positive T regulatory cells (Tregs) in the thymus that have the ability to suppress autoreactive cells (24, 25, 11). Thus, not only do more autoreactive cells escape deletion, but those Tregs normally in place to limit their activities are either not developed or are dysfunctional.

 

The peri-post pubertal period is a common time for presentation of some manifestations, although initial presentation may occur as early as the first year of life (3). The classic disorder is rare, and altogether it is estimated that there were only several hundred cases worldwide. However, the syndrome is more common in certain populations; 1: 25,000 in Finns, 1:14,500 Sardinians, 1:9000 Iranian Jews, all examples of past “isolated” populations that demonstrate a founder effect. Surprisingly, however, diabetes mellitus is uncommon and generally appears as a latemanifestation in the third and fourth decades of life (1-4). Unusual features include chronic kidney disease, apparent mineralocorticoid excess, asplenia, and oral or esophageal malignancy. The frequency, patterns and long-term outcomes of this syndrome vary in different populations that harbor different mutations; recent reviews of the patterns and outcomes in cohorts from Sardinia (26), Norway (10) and India (27) highlight these unique patterns. The classic features based on the Finnish cohort are summarized in Table1.

 

Table 1. Clinical Features of APS1

Symptom

Percentage of patients

Mucocutaneous candidiasis

80%

Hypoparathyroidism

70-80%

Adrenal Insufficiency

60%

Type 1 Diabetes Mellitus

12%

Hypothyroidism

4%

Ovarian Failure in Affected Females

60%

Testicular Failure in Affected Males

14%

Gastric Parietal Cell Failure

15%

Hepatitis

13%

Ectodermal Dysplasia

33%

Keratopathy

22%

Alopecia

27%

Vitiligo

13%

Based on references (1-4)

 

TREATMENT OF APS1

 

Treatment guidelines for this condition have been proposed (28); they are based on immune suppression and modulation with agents including glucocorticoids such as prednisone, cyclosporin, the calcineurin inhibitors tacrolimus and sirolimus, methotrexate, mycophenolate mofetil, and rituximab, a CD20 inhibitor; these are especially used for auto-immune hepatitis, enteropathy, tubulo-interstitial nephritis, interstitial lung disease and keratoconjunctivitis, andare detailed by Kisand et al (4).

 

In general, management of autoimmune polyendocrine syndromes includes hormonal replacement therapy as needed and treatment of complications.

 

Hormonal and vitamin (Vitamin D, B12) replacement should be implemented for the known hormonal deficiencies, and other deficiencies should be anticipated and screened for periodically, especially in those with circulating antibodies for components of adrenal steroidogenesis (21-hydroxyase, 17-hydroxylase), thyroid (TPO, TG antibodies) and calcium, phosphate, or parathyroid hormone levels as indicated. Periodic assessment of HbA1c, fasting glucose, liver function via ALT and AST should complement careful clinical assessment at 6 month-1year intervals in affected patients. When hypoparathyroidism and chronic mucocutaneous candidiasis are the initial manifestations, screening for primary adrenal insufficiency via an afternoon ACTH concentration is suggested to be performed every 6 months and at least annually. A level of ACTH greater than 80pg/ml is highly suggestive and a level exceeding 100pg/ml is virtually diagnostic. Whereas some recommend performing an ACTH- stimulation test to document adrenal reserve, others recommend starting cortisol replacement therapy and ongoing monitoring of sodium and potassium levels to exclude evolving aldosterone deficiency, as well as checking supine and standing blood pressure. Anti-candida drugs such as ketoconazole when used to treat the candidiasis should alert the treating physicians to exclude the possibility of adrenal insufficiency since these agents are known to interfere with cortisol synthesis and hence may accelerate the appearance of adrenal insufficiency or worsen manifestations of existing adrenal deficiency. Cortisol should initially be given in stress dosage, commonly 2-3 times the daily maintenance of ~10mg/m2/d for several days once initial diagnosis is established; thereafter, normal replacement doses of ~8- 10mg/M2/day may be given in 3 divided oral doses daily. When initial diagnosis is established during an inpatient admission, or at a subsequent hospital stay, consideration should be given to administer the hydrocortisone via parenteral means, intravenously or via intramuscular injection. This precaution is advised as oral medication may be less absorbed due to the concomitant presence of candidiasis of the esophagus and lower GI tract which might impair absorption. Patients should also be advised to wear a Medic-Alert bracelet, necklace or key chain, so that cortisol treatment is not delayed should a patient be involved in a motor vehicle accident or be in coma due to adrenal crisis or hypoglycemia. There is evidence that the predilection for autoimmunity in persons with trisomy 21 (Downs syndrome) may also be due to abnormality in the AIRE gene (9). Absent the classic triad of hypoparathyroidism, chronic mucocutaneous candidiasis, and primary adrenal insufficiency, or 2 of these three manifestations, it is likely that many cases are missed; the wide spectrum of potential presentations suggest that genetic testing via AIRE mutational analysis be considered in patients with hepatitis, chronic diarrhea, and periodic rash with fever (1). Recent reviews also raise the possibility of genetic manipulation of certain mutations to restore thymic surveillance at some future date (2). Patients with APS-1 are best followed by a multi-disciplinary team led by a pediatric or adult endocrinologist at an academic medical center. Patients should have a minimum of two follow-up visits per year due to the complexity of the entity, and asymptomatic mutations carriers should be followed at least annually. It is mandatory to check all siblings of APS-1 patients even if they are adults and seemingly well. Screening for 21-hydroxylase and NALP5 antibodies is useful in assessing the risk of development of adrenal insufficiency and hypoparathyroidism, respectively.

 

AUTOIMMUNE POLYGLANDULAR SYNDROME 2 (APS2a; SCHMIDT SYNDROME)

 

APS2 is characterized by the triad of T1DM, Addison disease, and thyroid autoimmunity with hypothyroidism, hyperthyroidism, or Hashimoto thyroiditis; T1DM and Addison disease are obligatory components, but thyroid autoimmunity is not and a host of other autoimmune entities can also be associated. These entities include celiac disease, vitiligo, alopecia, myasthenia gravis, pernicious anemia, IgA deficiency, hepatitis, and hypogonadism. Peak prevalence is in the range of 20-40 years of age. In keeping with an autoimmune basis, the syndrome is more prevalent in females and associated with specific HLA DR3 and DR4 haplotypes and with the class II HLA alleles DQ2 and DQ8, also strongly linked to celiac disease. Autoantibodies to islet cell components (GAD65, IA2, ZnT8), thyroid (anti thyroglobulin TG, anti-thyroid peroxidase TPO), adrenal leading to Addison disease (anti-21-hydroxylase or anti 17-hydroxylase), and celiac disease (tissue transglutaminase and gliadin) are commonly present and should be periodically sought in those with one or more autoimmune endocrinopathies such as Addison disease and T1DM. Specific treatment for each entity should be continued in the hospital, with cortisol dosage adjusted for stress (8). A mechanism by which viral disease may trigger autoimmunity in the gut leading to celiac disease has recently been proposed and may have relevance to the other auto-immune diseases that form this entity (29).

 

The onset of APS-2 typically appears later than APS-1, mostly in young adulthood. Currently, there are no unique tests to detect patients with APS-2, but testing for autoantibodies may be helpful in assessing disease risk, since the relevant autoantibodies are frequently detectable years before disease onset. Despite the major advancement in identification of disease genes, the heritability of APS-2 is complex. Some authors propose splitting this syndrome into further subtypes, but there is little evidence for distinct etiologies in such subcategories, so the broader term APS-2 for all of these patients seems appropriate (11).

 

Figure 2. Illustrative Case

 

A 16-year-old girl was admitted to the hospital in coma and found to have profound hypoglycemia. Her physical findings were striking for pigmented patches on her tongue, gums, and lips and her skin was deeply suntanned (see figure 2). The mother related that her daughter has T1DM since age 12 and had been experiencing numerous hypoglycemic episodes unrelated to food intake or exercise. Accordingly, the dose of insulin had been reduced to about 50% of what it had been 3 months previously. She responded to glucose infusion and recovered full consciousness. Laboratory tests documented a marked elevation of ACTH, low morning cortisol, elevated antibodies to 21OH, and markedly elevated TSH with a low T4. Thus, this patient fulfills all the criteria for APS2. The hypoglycemia was due to a combination of deficient hormonal counter-regulation (cortisol deficiency) as well as the delayed clearance of insulin as a result of hypothyroidism.

 

AUTOIMMUNE POLYGLANDULAR SYNDROME 3 (APS2b)

 

APS-3 also known as APS2b, is sometimes referred to as Carpenter’s syndrome, and has the same array of endocrine tissue autoimmune abnormalities as APS2, but without Addison disease. Almost 20% of patients with T1DM have thyroglobulin (TG) and thyroid peroxidase (TPO) antibodies, but only a minority progress to clinical or biochemical hypothyroidism, so APS3 (APS2b) could be considered as a relatively common disorder (9). The clinical features of APS 2 and 3 are shown in table 2.

 

Table 2. Clinical Features of APS2 and APS3

APS2

APS3

Type 1 Diabetes Mellitus

Type 1 Diabetes Mellitus

Thyroid autoimmunity

Thyroid autoimmunity

Adrenal Insufficiency

 

 

TREATMENT OF APS2 and APS3

 

Treatment of APS-2 should focus on replacement of missing hormones according to current guidelines for treating the main components of APS-2 and 3. Physicians should be particularly aware that a patient with APS-2 or 3 has an increased risk of developing another organ-specific autoimmune disease. Massive family accumulation of autoimmune diseases, especially with early debut could indicate a monogenic disease, possibly a “non-classical” APS-1 especially if vitiligo and pernicious anemia is prevalent (2).

 

ADRENAL INSUFFICIENCY

 

Since adrenal insufficiency is a hallmark feature of APS1 and 2a syndromes, and since it is the most life threatening, we briefly review the crucial role of the adrenal in metabolic homeostasis. During stress, cortisol produced by the zona fasciculata of the adrenal gland is required to maintain normoglycemia and hemodynamic stability. Cortisol regulates carbohydrate metabolism to maintain normoglycemia, decreases capillary permeability to maintain a normal blood pressure, and is required for activating enzymatic activity to convert norepinephrine to epinephrine. Cortisol production is under the regulation of the hypothalamus and pituitary. The hypothalamic-pituitary-adrenal (HPA) axis is mediated through the circulating level of plasma cortisol by negative feedback of cortisol on corticotropin releasing factor (CRF) and ACTH secretion. Aldosterone produced by the zona glomerulosa is predominantly regulated by the renin-angiotensin system. Aldosterone stimulates the kidneys to reabsorb sodium and water and excrete potassium. At high concentrations, cortisol can also act on the mineralocorticoid receptor to increase sodium and water retention as the activity of 11β-hydroxysteroid-dehydrogenase 2 which inactivates cortisol to cortisone is overwhelmed.

 

Presentation of adrenal insufficiency is often chronic, presenting with fatigue, anorexia, and weight loss; hyperpigmentation of the buccal mucosa and skin creases or generalized tanning of the skin occur with primary adrenal insufficiency from the excess of melanocyte stimulating hormone produced as a byproduct in the formation of excess ACTH.

 

Adrenal insufficiency can be caused by primary adrenal disease or dysfunction of the HPA axis (secondary adrenal insufficiency). The most common etiologies of primary adrenal disease in children, adolescents and young adults include autoimmune disease, retroperitoneal trauma, and rare genetic syndromes involving the formation of the adrenal gland, the biosynthetic formation of cortisol and the ability of the adrenal gland to respond to ACTH. Severe defects may present in the neonatal period or may be unmasked later in life by the requirement for higher secretion during a physiological stress situation such as sepsis or trauma. Secondary adrenal insufficiency is most commonly caused by damage to the hypothalamus or pituitary gland by trauma or neurological surgery or impingement on these structures by a tumor or mass; congenital defects of isolated ACTH formation or action also may occur. Morecommonly, suppression of the HPA axis can occur in patients chronically treated with potent glucocorticoid steroids.

 

Patients with adrenal insufficiency can present acutely in a severe life-threatening event termed adrenal crisis, particularly if there is an inciting event such as a septic illness, surgical procedure, anesthesia, or trauma. These patients have symptoms of nausea, vomiting, abdominal pain, dehydration, altered mental status, hypotension, hypoglycemia, or shock (32). Hypotension may be unresponsive to fluid resuscitation alone due to deficiency of cortisol required to activate β-adrenergic receptors and vascular tone. Salt wasting (hyponatremia, hyperkalemia) results from aldosterone deficiency. A cardinal feature of primary adrenal insufficiency is hyperpigmentation owing to concurrent rise in melanocyte stimulating hormone (MSH) associated with elevated ACTH production. Darkening of the skin is most prominent at the axillae, palmer creases, areolae, genitalia, and pigmentary lines of the gums (see Fig 2 above). This hyperpigmentation does not occur in secondary adrenal insufficiency as there is no increase in ACTH. Secondary causes of adrenal insufficiency, and certain forms of primary adrenal insufficiency that do not affect aldosterone production, do not present with salt-wasting and Addisonian crisis.

 

Because of the circadian rhythm and diurnal variation in ACTH and cortisol production, early morning serum cortisol and ACTH concentrations provide the best assessment of endogenous adrenal function. An early morning serum cortisol of <10 mcg/dl is suspicious for adrenal insufficiency. The corresponding ACTH concentration is elevated in primary adrenal insufficiency; a low ACTH concentration is suspicious for secondary adrenal insufficiency. However, a randomly timed cortisol measurement of <15 mcg/dl, in the setting of an acute illness has been proposed as concerning for adrenal insufficiency in adults (30). In the absence of clinical clues suggesting primary adrenal insufficiency, such as hyperpigmentation, stimulation with ACTH is the best diagnostic test for identifying those with adrenal insufficiency. At baseline, ACTH and cortisol blood levels are obtained and 250 mcg of synthetic ACTH (cosyntropin) is administered either via the intravenous (IV) or intramuscular (IM) route. The test is considered diagnostic of adrenal insufficiency if the peak cortisol level is less than 18 mcg/dl, 60 minutes following cosyntropin administration (31). Such a supra- physiologic dose of ACTH may overcome a defect in the hypothalamic-pituitary-adrenal axis to produce a rise in serum cortisol. If there is a high suspicion for secondary adrenal insufficiency in the face of a normal cortisol response to high dose ACTH, early morning serum cortisol and ACTH concentrations may bemore informative. The causes of adrenal insufficiency as well as that of thyroid dysfunction and their management are described elsewhere in Endotext (32, 33).

 

AUTO-IMMUNITY ASSOCIATED WITH CANCER IMMUNOTHERAPY

 

An increasingly important and frequent cause of endocrine-autoimmune syndromes is their appearance in association with the increasing use of immunotherapy as a front line or back-up therapy in various cancers. Indeed, the variety and severity of endocrine autoimmune syndromes associated with the use of inhibitors of CTLA4 (cytotoxic T-lymphocyte-associated protein 4) such as ipilimumab, and immune checkpoint blockade of programmed death 1(PD-1) and its ligands PDL1 and PDL2, has recently been termed “the Achilles heel of cancer immunotherapy” (34). The range of autoimmune endocrine manifestations includes hypophysitis with disturbances in anterior pituitary hormones, hypo-and hyperthyroidism, adrenal insufficiency, and T1DM (35-37). Key checkpoints by which autoimmunity is regulated in normal individuals are also exploited by tumors to evade recognition and elimination via the immune system; employing immuno-regulatory agents that block these checkpoints facilitates the recognition of tumor antigensas foreign and activates their destruction, but at the same time stimulates autoimmune responses to self-antigens. Clinicians should be aware of these autoimmune manifestations and screen for involvement of endocrine tissues or their clinical manifestations. Notably, some of these endocrine autoimmune manifestations may appear months to years after initiation of immune therapy for cancer (36).

 

Thyroid disorders, typically associated with anti-PD-1 antibodies and hypophysitis commonly related to anti-CTLA-4 therapy, are the two most frequent endocrine organ pathologies (37). Notably, in a large cohort, it was shown that the incidence of any-grade immune-related adverse event (irAE) is higher with CTLA-4 (53.8%) than with PD-1 (26.5%) and PD-L1 (17.1%). Moreover, the incidence of any-grade irAE was highest in patients receiving CTLA-4 plus PD-1/PD-L1 combinations (61.1%) (38). The incidence of endocrine adverse events reported with the use of immune checkpoint inhibitors (ICI) ranges from 5% to 20% (39, 40), with a recent systematic review and meta-analysis reporting an overall incidence of clinically significant endocrinopathies of approximately 10% (41). Hypophysitis appears most often in men older than 60 years and is 2–5 times more frequent than in women. The incidence reported is 4%–20% with ipilimumab, 8% with the combination ipilimumab plus nivolumab, 0.6% with nivolumab, and 0.7% with pembrolizumab (40). The incidence of hypothyroidism ranges from 6% to 13% and for thyrotoxicosis from 3% to 16%. However, when subclinical hypothyroidism or hyperthyroidism is included, the incidence can reach 28% and 22%, respectively (42). These risks were reported to be dose dependent; in the case of anti-CTLA-4 treatment, the risk was observed only above a treatment threshold of 10 mg/kg.

 

Endocrinopathies less often reported include T1DM, primary adrenal insufficiency, central diabetes insipidus, and hypoparathyroidism. In the case of primary adrenal insufficiency, an incidence of less than 1% with monotherapy and 4%–8% with combined immunotherapy has been reported (41). For T1DM an overall incidence of 0.4% was reported in patients treated with anti-PD-1/PD-L1 but not those treated with anti-CTLA-4 (43, 44). However, a recent study reported a prevalence of 0.9% among 2,960 patients treated by ICI (45). In clinical practice, significant irAEs (grade 2 or higher) are managed with systemic immunosuppression mostly in the form of corticosteroids with methylprednisolone 0.5–1 mg/kg for grade 2 and 1–2 mg/kg for grade 2–3; for grade 4 irAEs, resuming treatment with the drug is contraindicated. More rarely, anti-tumor necrosis factor-α agents have been used if steroids are not effective or contraindicated (37).

 

X-LINKED IMMUNODYSREGULATION, POLYENDOCRINOPATHY, AND ENTEROPATHY (IPEX)

 

X-linked Immunodysregulation, Polyendocrinopathy and Enteropathy - or IPEX- is an extremely rare inherited syndrome characterized by early onset T1DM (46), autoimmune enteropathy with intractable diarrhea and malabsorption, and dermatitis that may be eczematiform, ichthyosiform or psoriasiform. Eosinophilia and elevated IgE-levels are frequently present in IPEX. Some patients develop kidney disease, most often membranous glomerulonephritis or interstitial nephritis. Later manifestations may include autoimmune thyroiditis, alopecia, various autoimmune cytopenias, hepatitis, and exocrine pancreatitis (47). Several of these features overlap with APS-1, but in IPEX they usually develop much earlier in life. The disorder is frequently fatal in the first few years of life, unless patients are diagnosed and promptly treated with immunosuppressive agents or, if possible, receive an allogenic bone marrow transplant, which can be curative (47).

 

The defective gene was mapped to mutations in the FOXP3 (human) gene (48). To date, about 70 different mutations have been reported in patients. FOXP3 is currently recognized as a master transcription factor that is highly expressed in Tregs in association with other key Treg elements such as CD4, cytotoxic T Lymphocyte-associated protein 4 (CTLA4), and CD25, the high affinity IL-2 receptor (11, 49, 50).

 

Patients with IPEX, like those with APS-1, develop circulating autoantibodies that can be helpful in making the diagnosis. Despite the rarity of IPEX, studies of affected patients have revealed a key pathway for self-tolerance that has aided in the understanding of Tregs and has led to research aimed at the development of methods to enhance Treg function in transplantation and as a treatment for autoimmune disorders (51).

 

NEW DIRECTIONS

 

Over the past decade better diagnostic tools including genetic tests and autoantibody analyses have been developed for the detection and management of APS-related diseases. Early diagnosis in association with personalized genomics might possibly enable physicians to apply early immunomodulatory therapy to ameliorate the autoimmune process before irreversible organ damage has occurred. Restoration of thymic epithelium with intact immune regulatory function via stem cell engineering to reverse the defective immune system remains a long -term goal, but is being pursued (11).

 

REFERENCES

 

  1. Perheentupa, J. Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J Clin Endocrinol Metab, 2006; 91(8): 2843-50.
  2. Anderson, M.S. and M.A. Su. AIRE expands: new roles in immune tolerance and beyond. Nat Rev Immunol, 2016;16(4): 247-58.
  3. Fierabracci, A. Type 1 Diabetes in Autoimmune Polyendocrinopathy-Candidiasis- Ectodermal Dystrophy Syndrome (APECED): A "Rare" Manifestation in a "Rare" Disease. Int J Mol Sci, 2016;17(7).
  4. Kisand, K. and P. Peterson. Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy. J Clin Immunol, 2015;35(5): 463-78.
  5. Alimohammadi, M., et al. Autoimmune polyendocrine syndrome type 1 and NALP5, a parathyroid autoantigen. N Engl J Med, 2008; 358(10):1018-28.
  6. Finnish-German, A.C. An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat Genet, 1997; 17(4): 399-403.
  7. Anderson, M.S., et al. Projection of an immunological self shadow within the thymus by the aire protein. Science, 2002; 298(5597):1395-401.
  8. Kakleas, K., et al. Associated autoimmune diseases in children and adolescents with type 1 diabetes mellitus (T1DM). Autoimmun Rev, 2015; 14(9): 781-97.
  9. Gimenez-Barcons, M., 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.
  10. Bruserud Ø, Oftedal BE, Landegren N, Erichsen MM, Bratland E, Lima K, Jørgensen AP, Myhre AG, Svartberg J, Fougner KJ, Bakke Å, Nedrebø BG, Mella B, Breivik L, Viken MK, Knappskog PM, Marthinussen MC, Løvås K, Kämpe O, Wolff AB, Husebye ES. A Longitudinal Follow-up of Autoimmune Polyendocrine Syndrome Type 1. J Clin Endocrinol Metab. 2016;101(8):2975-83
  11. Husebye ES, Anderson MS, Kämpe O. Autoimmune Polyendocrine Syndromes. N Engl J Med. 2018; 22;378(12):1132-1141
  12. Brannstrom, J., et al. Defect internalization and tyrosine kinase activation in Aire deficient antigen presenting cells exposed to Candida albicans antigens. Clin Immunol, 2006; 121(3): 265-73.
  13. Bensing S, Brandt L, Tabaroj F, Sjöberg O, Nilsson B, Ekbom A, Blomqvist P, Kämpe O. Increased death risk and altered cancer incidence pattern in patients with isolated or combined autoimmune primary adrenocortical insufficiency. Clin Endocrinol (Oxf). 2008;69:697-704
  14. Shum AK, Alimohammadi M, Tan CL, et al. BPIFB1 is a lung-specific autoantigen associated with interstitial lung disease. Science translational medicine. 2013; 5:206ra139.
  15. Alimohammadi M, Dubois N, Skoldberg F, et al. Pulmonary autoimmunity as a feature of autoimmune polyendocrine syndrome type 1 and identification of KCNRG as a bronchial autoantigen. Proc Natl Acad Sci U S A. 2009;106:4396–401.
  16. Landegren N, Sharon D, Shum AK, et al. Transglutaminase 4 as a prostate autoantigen in male subfertility. Science translational medicine. 2015; 7:292.
  17. Meager, A., et al. Anti-interferon autoantibodies in autoimmune polyendocrinopathy syndrome type 1. PLoS Med, 2006;3(7):289.
  18. Meyer, S., et al., AIRE-Deficient Patients Harbor Unique High-Affinity Disease-Ameliorating Autoantibodies. Cell, 2016;166(3): 582-595.
  19. Kisand K, Bøe Wolff AS, Podkrajsek KT, Tserel L, Link M, Kisand KV, Ersvaer E, Perheentupa J, Erichsen MM, Bratanic N, Meloni A, Cetani F, Perniola R, Ergun-Longmire B, Maclaren N, Krohn KJ, Pura M, Schalke B, Ströbel P, Leite MI, Battelino T, Husebye ES, Peterson P, Willcox N, Meager A. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with
  20. Orlova EM, Sozaeva LS, Kareva MA, Oftedal BE, Wolff ASB, Breivik L, Zakharova EY, Ivanova ON, Kämpe O, Dedov II, Knappskog PM, Peterkova VA, Husebye ES. Expanding the Phenotypic and Genotypic Landscape of Autoimmune Polyendocrine Syndrome Type 1. J Clin Endocrinol Metab. 2017;102(9):3546-3556.
  21. Proust-Lemoine E, Saugier-Véber P, Lefranc D, Dubucquoi S, Ryndak A, Buob D, Lalau JD, Desailloud R, Weill J, Prin L, Lefebvre H, Wémeau JL. Autoimmune polyendocrine syndrome type 1 in north-western France: AIRE gene mutation specificities and severe forms needing immunosuppressive therapies. Horm Res Paediatr. 2010;74(4):275-84.
  22. Cetani F, Barbesino G, Borsari S, Pardi E, Cianferotti L, Pinchera A, Marcocci C. A novel mutation of the autoimmune regulator gene in an Italian kindred with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, acting in a dominant fashion and strongly cosegregating with hypothyroid autoimmune thyroiditis. J Clin Endocrinol Metab. 2001;86(10):4747-52.
  23. Abbott JK, Huoh YS, Reynolds PR, et al. Dominant-negative loss of function arises from a second, more frequent variant within the SAND domain of autoimmune regulator (AIRE). J Autoimmun. 2017
  24. Malchow S, Leventhal DS, Nishi S, Fischer BI, Shen L, Paner GP, Amit AS, Kang C, Geddes JE, Allison JP, Socci ND, Savage PA. Aire-dependent thymic development of tumor-associated regulatory T cells. Science. 2013;339(6124):1219-24.
  25. Leonard JD, Gilmore DC, Dileepan T, Nawrocka WI, Chao JL, Schoenbach MH, Jenkins MK, Adams EJ, Savage PA. Identification of Natural Regulatory T Cell Epitopes Reveals Convergence on a Dominant Autoantigen. Immunity. 2017;47(1):107-117
  26. Meloni, A., et al. Autoimmune polyendocrine syndrome type 1: an extensive longitudinal study in Sardinian patients. J Clin Endocrinol Metab, 2012;97(4):1114-24.
  27. Zaidi, G., et al. Autoimmune polyendocrine syndrome type 1 in an Indian cohort: a longitudinal study. Endocr Connect, 2017;6(5):289-296.
  28. Husebye, E.S., et al. Clinical manifestations and management of patients with autoimmune polyendocrine syndrome type I. J Intern Med, 2009; 265(5): 514-29.
  29. Verdu, E.F. and A. Caminero. How infection can incite sensitivity to food. Science, 2017;

         356(6333):29-30.

  1. Cooper, M.S. and P.M. Stewart. Corticosteroid insufficiency in acutely ill patients. N Engl J Med, 2003; 348(8):727-34.
  2. Bornstein, S.R., et al. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab, 2016; 101(2): 364-89.
  3. Weetman, A. and L.J. DeGroot. Autoimmunity to the Thyroid Gland, in Endotext, L.J. De Groot, et al., Editors. 2000: South Dartmouth (MA).
  4. Nicolaides, N.C., Chrousos, and E. Charmandari. Adrenal Insufficiency, in Endotext, L.J. De Groot, et al., Editors. 2000: South Dartmouth (MA).
  5. June, C.H., J.T. Warshauer, and J.A. Bluestone. Is autoimmunity the Achilles' heel of cancer immunotherapy? Nat Med, 2017; 23(5): 540-547.
  6. Ryder, M., et al. 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.
  7. Pedoeem, A., et al. Programmed death-1 pathway in cancer and autoimmunity. Clin Immunol, 2014;153(1): 145-52.
  8. Del Rivero J, Cordes LM, Klubo-Gwiezdzinska J, Madan RA, Nieman LK, Gulley JL. Endocrine-Related Adverse Events Related to Immune Checkpoint Inhibitors: Proposed Algorithms for Management. Oncologist, 2020;25(4):290-300.
  9. Faje AT, Sullivan R, Lawrence D et al. Ipilimumab-induced hypophysitis: A detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J Clin Endocrinol Metab, 2014;99:4078–4085.
  10. Corsello SM, Barnabei A, Marchetti P et al. Endocrine side effects induced by immune checkpoint inhibitors. J Clin Endocrinol Metab, 2013;98:1361–1375.
  11. Torino F, Barnabei A, Paragliola RM et al. Endocrine side-effects of anti-cancer drugs: mAbs and pituitary dysfunction: Clinical evidence and pathogenic hypotheses. Eur J Endocrinol, 2013;169:153–164
  12. Barroso-Sousa R, Barry WT, Garrido-Castro AC et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: A systematic review and meta-analysis. JAMA Oncol, 2018;4:173–182
  13. Illouz F, Drui D, Caron P et al. Expert opinion on thyroid complications in immunotherapy. Ann Endocrinol (Paris) 2018;79:555–561.
  14. Byun DJ, Wolchok JD, Rosenberg LM et al. Cancer immunotherapy - Immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol, 2017;13:195–207
  15. Sznol M, Postow MA, Davies MJ et al. Endocrine- related adverse events associated with immune checkpoint blockade and expert insights on their management. Cancer Treat Rev, 2017;58:70–76
  16. Stamatouli AM, Quandt Z, Perdigoto AL et al. Collateral damage: Insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes, 2018;67:1471–1480.
  17. Wildin RS, Smyk-Pearson S, Filipovich AH. Clinical and molecular features of the immunodysregulation, polyendocrinopathy, enteropathy, X linked (IPEX) syndrome. J Med Genet, 2002;39(8):537-45.
  18. Barzaghi F, Amaya Hernandez LC, Neven B, et al. Long-term follow up of IPEX syndrome patients after different therapeutic strategies: an international multicenter retrospective study. The Journal of allergy and clinical immunology, 2017
  19. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, Kelly TE, Saulsbury FT, Chance PF, Ochs HD. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet, 2001;27(1):20-1.
  20. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell, 2008;133(5):775-87.
  21. Caudy AA, Reddy ST, Chatila T, Atkinson JP, Verbsky JW. CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like syndrome, and defective IL-10 expression from CD4 lymphocytes. J Allergy Clin Immunol, 2007;119(2):482-7.
  22. Bluestone JA, Buckner JH, Fitch M, et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Science translational medicine. 2015; 7:315ra189

Physiology of the Hypothalamic-Pituitary-Thyroid Axis

ABSTRACT

 

The activity of the thyroid gland is predominantly regulated by the concentration of the pituitary glycoprotein hormone, thyroid-stimulating hormone (TSH). In the absence of the pituitary or of thyrotroph function, hypothyroidism ensues. Thus, regulation of thyroid function in normal individuals is to a large extent determined by the factors which regulate the synthesis and secretion of TSH. Those factors are reviewed in this chapter and consist principally of thyrotropin-releasing hormone (TRH) and the feedback effects of circulating thyroid hormones at the hypothalamic and pituitary levels. The consequence of the dynamic interplay of these two dominant influences on TSH secretion, the positive effect of TRH on the one hand and the negative effects of thyroid hormones on the other, results in a remarkably stable morning concentration of TSH in the circulation and consequently little alteration in the level of circulating thyroid hormones from day to day and year to year. This regulation is so carefully maintained that an abnormal serum TSH in most patients is believed to indicate the presence of a disorder of thyroid gland function. The utility of TSH measurements has been recognized and its use has remarkably increased due to the development of immunometric methodologies for its accurate quantitation in serum, although the criteria to define a “normal range” still remain a matter of controversy. This chapter is organized into two general sections. The first portion reviews basic studies of TSH synthesis, post-translational modification, and release. The second deals with physiological studies in humans which serve as the background for the diagnostic use of TSH measurements and reviews the results of TSH assays in pathophysiological disorders.

 

The Regulation of Thyroid-Stimulating Hormone synthesis and

secretion: Molecular Biology and Biochemistry

The TSH Molecule

 

TSH is a heterodimer consisting of an alpha and a beta subunit that are tightly, but non-covalently, bound (1,2). While the molecular weight of the deduced amino-acid sequence of the mature alpha and beta subunits in combination is approximately 28,000 Da, additional carbohydrate (15% by weight) results in a significantly higher molecular weight estimate based on sizing by polyacrylamide gel electrophoresis. The alpha subunit (glycoprotein hormones, alpha polypeptide) is common to TSH, follicle-stimulating hormone (FSH), luteinizing hormone (LH), and chorionic gonadotropin (CG). The beta subunit confers specificity to the molecule since it interacts with the TSH receptor (TSH-R) expressed on the basolateral membrane of thyroid follicular cells, and is rate-limiting in the formation of the mature heterodimeric protein. However, the free beta subunit is inactive and requires noncovalent combination with the alpha subunit to express hormonal bioactivity. The linear sequence of the human alpha subunit consists of 92 amino acids including 10 cystine residues that form a total 5 bonds through disulfide linkage. The human TSH beta (hTSH beta) subunit contains 118 amino-acids, as predicted by complementary DNA sequences, but hTSH beta isolated from the pituitary gland has an apoprotein core of 112 amino-acids, due to carboxyl-terminal truncation during purification.

 

The production rate (PR) of human TSH is normally between 50 and 200 mU/day and increases markedly (up to >4000 mU/day) in primary hypothyroidism; the metabolic clearance rate (MCR) of the hormone is about 25 ml/min/m2 in euthyroidism, while significantly higher in hyperthyroidism and lower in hypothyroidism (3). The PR of free alpha subunit is about 100 µg/day, increases increase approximately two-fold in primary hypothyroidism and in post-menopausal women, and decreases (about to one half) in hyperthyroidism (4). The PR of free TSH beta subunit is too low to be calculated in all hyperthyroid and in most euthyroid subjects, while is 25-30 ug/day in primary hypothyroidism (4). The MCR of the free subunits is 2-3 times faster than that of TSH, being about 68 ml/min/m2 for alpha and 48 ml/min/m2 for the beta subunit (4). The half-life of circulating TSH ranges from 50 to 80 minutes (4).

 

The gene coding for the alpha subunit (CGA) is located on chromosome 6 and the thyroid stimulating hormone subunit beta (TSHB)  gene on chromosome 1 (5). The structure of CGA gene has been determined in several animal species (6,7). The genes of each species are approximately of the same size and similarly organized in four exons and three introns. The human gene is 9.4 kilobases (kb) in length, with three introns measuring 6.4 kb, 1.7 kb and 0.4 kb, respectively. The TSHB gene has been isolated in mouse (7), rat (8), and humans (9,10), among other species. In contrast to the CGA gene, the organization of the TSHB gene is somewhat variable between the different species. The rat and the human genes are organized in three exons, while the mouse gene contains two additional 5'-untranslated exons. The first exon is untranslated, the leader peptide and the first 34 amino-acids are encoded by the second exon, while the third exon represents the remaining coding region and 3'-untranslated sequences. A single transcription start has been identified in the hTSHB gene, while the rat and the mouse genes contain two starting sites separated by approximately 40 base pairs (bp); transcription begins predominantly from the downstream site, which corresponds to the location of the human transcriptional start. A schematic representation of the TSHB gene is shown in Figure 1.

Figure 1. Thyrotropin β (TSHB) gene structure. Some mutations of the gene found in patients with congenital central hypothyroidism are also depicted (modified from McDermott et al. (11) and Baquedano et al. (12)).

 

The pre-translational regulation of TSH synthesis and secretion is a complex process, detailed in the next paragraphs. The formation of mature TSH involves several post-translational steps including the excision of signal peptides from both subunits and co-translational glycosylation with high mannose oligosaccharides (13,14). As the glycoproteins are successively transferred from the rough endoplasmic reticulum to the Golgi apparatus, the trimming of mannose and further addition of fucose, galactose and sialic acid occurs (15). The alpha subunit has two and the beta subunit has one asparagine (N)-linked oligosaccharide(s) showing a typical biantennary structure fully sulfated in bovine and half-sulfated in human TSH (2). The primary intracellular role of these glycosylation events may be to allow proper folding of the alpha and beta subunits permitting their heterodimerization and also preventing intracellular degradation (16,15). On the basis of crystallographic studies on hCG and other glycoprotein hormones, a homology model of the tridimensional structure of TSH has been proposed (17). This model (Fig. 2) predicts for both the alpha and the beta subunit the presence of two beta-hairpin loops (L1 and L3) on one side of a central "cystine knot" (pair of cysteine molecules) formed by three disulfide bonds, and a long loop (L2) on the other side. Both alpha and beta chains have functionally important domains involved in TSH-R binding and activation (Fig. 2) (18). Of particular relevance is the so-called “seat belt” region of the beta chain comprised between the 10th (C86) and the 12th (C105) cysteine residue (Fig. 2 and Fig. 3). The name “seat belt” derives from the conformational structure of the beta chain determined by the disulfide bridge (C39/C125) toward the C-terminal tail of the beta subunit that wraps the alpha subunit like a “seat belt” (Fig. 3), and stabilizes the heterodimerization of TSH (2,18).

Figure 2. Schematic drawing of human TSH, based on a molecular homology model built on the template of a hCG model (17). The α-subunit is shown as checkered, and the β-subunit as a solid line. The two hairpin loops in each subunit are marked L1, L3; each subunit has also a long loop (L2), which extends from the opposite site of the central cystine knot. The functionally important α-subunit domains are boxed. Important domains of the β-subunit are marked directly within the line drawing (crossed line, beaded line and dashed line). Reproduced from Grossmann et al. (2) with permission, where further details can be found.

Figure 3. Structural model of TSH based on the FSH x-ray structure, which is the best available structural template for TSH. The boxed residue numbers represent cysteines residues, which form stabilizing disulfide bridges (yellow): 5 in α-subunit (red orange), and 6 in the β-subunit (magenta). The disulfide bridge (C39/C125) toward the C-terminal tail of the α-subunit of TSH that wraps around the β-subunit like a “seat belt” stabilizes the heterodimerization of TSH as well as that of FSH, LH, and HCG. (Reproduced from Kleinau et al. (18) with permission)

 

Proper TSH glycosylation is also necessary to attain normal bioactivity (19), a process which requires the interaction of the neuropeptide thyrotropin-releasing hormone (TRH) (Fig. 4), with its receptor on thyrotroph cells (20-22). The requirement for TRH in this process is illustrated by the fact that in patients with central hypothyroidism due to hypothalamic-pituitary dysfunction, normal or even slightly elevated levels of TSH are detected by radioimmunoassay, but biologically subpotent forms are found in the circulation together with reduced levels of free T4 (23-25). Chronic TRH administration to such patients normalizes the glycosylation process enhancing both its TSH-R binding affinity as well as its capacity to activate adenyl cyclase. This, in turn, can normalize thyroid function in such patients (26). On the other hand, enhanced TSH bioactivity is invariably found in sera from patients with thyroid hormone resistance (27). Moreover, variations of TSH bioactivity (mostly related to different TSH glycosylation) have been observed in normal subjects during the nocturnal TSH surge, in normal fetuses during the last trimester of pregnancy, in primary hypothyroidism, in patients with TSH-secreting pituitary adenomas, and in non-thyroidal illnesses (27,28). Glycosylation of the molecule can also influence the rapidity of clearance of TSH from the circulation. Taken together, these findings have led to a new concept of a qualitative regulation of TSH secretion, mainly achieved through both the transcriptional and post-transcriptional mechanisms involving not only TSH glycosylation (29), but also thyrocyte physiology and thyroid disorders (30).

Figure 4. Structure of TRH

 

Specific amino-acid sequences in the common alpha and beta subunits are critical for the heterodimerization, secretion and bioactivity of mature TSH. These sequences include highly conserved segments which are essential for TSH-R binding and biological activity (see Refs (2,18) for an extensive review). The peptide sequence 27CAGYC31 (cysteine-alanine-glycine-tyrosine-cysteine) is highly conserved in the beta subunit of TSH, LH, hCG, as well as FSH, and is thought to be important in heterodimerization with the alpha subunit (31,32). Several inherited TSHB gene mutations are responsible for isolated familial central hypothyroidism and are listed in Table 1 and depicted in Fig. 1. The most frequent mutation is a homozygous single-base deletion in codon 105 that results in a substitution of cysteine 105 by valine and an additional 8 amino acid nonhomologous peptide extension on the mutant protein (C105V, 114X). The mutation destroys a disulfide bond essential for normal protein conformation and bioactivity and leads to an unstable heterodimer (33-38,11,39,40,12,41,42).

 

Table 1. Mutations in the Beta Subunit Gene Responsible for Congenital Isolated Central Hypothyroidism

Mutation of TSHB

Consequence of mutation on TSH heterodimer formation

G29R (31)

Prevents dimer formation modifying the CAGYC region

E12X (33)

Truncated TSH beta subunit unable to associate with alpha chain

C105V, 114X (41)

Destruction of a disulfide bond, non-homologous carboxyterminus. Change of amino acid sequence in the “seat belt” region leads to unstable heterodimer

Q49X (37)

Truncated TSH beta subunit forming a bio-inactive heterodimer with the alpha chain

IVS2+5A (39)

Base substitution at intron 2 (position +5) with shift of the translational start point to an out of frame position of exon 3 resulting in a truncated transcript

C85R (43)

T to C transition at codon 85 of exon 3 resulting in a change of cysteine to arginine, preventing the formation of a functional heterodimer with the alpha subunit

C162GA (12)

G to A change at the 5’ donor splice site of exon/intron 2 transition causing a (CGACGG) polymorphism, which although per se silent, disrupts the 5’ consensus sequence critical for splicing and causes complete skipping of exon 2

C88Y (12)

323G>A transition resulting in a C88Y change. This cysteine residue is conserved among all pituitary and placental glycoprotein hormone-beta subunits and the loss alters the conformation and intracellular degradation

 

The understanding of the relationship between molecular structure and biological activity of TSH recently allowed the synthesis of TSH variants designed by site-directed mutagenesis with either antagonist (43) or superagonist (44) activity that potentially offer novel therapeutic alternatives. More recently, newly chemically modified compounds with low molecular-weight and able to antagonize the TSH receptor have been reported (45,46). These drugs may possess agonist or antagonist properties. Indeed, a non peptidic antagonist, therefore devoid of intrinsic immunogenicity, might be very useful in the treatment of Graves’ disease and other forms of hyperthyroidism, such as TSH-secreting pituitary adenomas, Graves’ orbitopathy, and activating mutations of the TSH receptor (47,48).

 

Other Thyrotropic Hormones

 

A second thyrotropic hormone formed by a heterodimer of two distinct glycoprotein subunits (glycoprotein hormone alpha 2-subunit - GPA2 and glycoprotein hormone beta 5-subunit - GPB5) has been identified in the human pituitary and called thyrostimulin (49-53). Thyrostimulin has a sequence similarity of 29% with the alpha and 43% with the beta subunit and is able to activate the TSH-R (54,18). Although it has been hypothesized that it could account for the residual stimulation of thyroid gland observed in patients with central hypothyroidism (55), its physiological role is still unknown. The GPA2/GPB5 heterodimer is localized in extrapituitary tissues such as the eye, testis, bone, and ovary (54,56,57), while the anterior pituitary expresses almost exclusively GPA2 (54). In the rat ovary, thyrostimulin activates the TSH-R expressed in granulosa cells suggesting a potential paracrine activity (56).

 

Regulation of TSH Synthesis and Secretion        

 

The major regulators of TSH production are represented by the inhibitory effects of thyroid hormone (58) and by the stimulatory action of TRH. As shown in Fig. 5, T3 acts via binding to the nuclear thyroid hormone receptor β2 isoform present in thyrotrophs, and T4 mainly acts via its intra-pituitary or intra-hypothalamic conversion to T3, although a direct negative effect of T4 independent from local T3 generation on TSHB gene expression has been documented (59). Both thyroid hormones directly regulate the synthesis and release of TSH at the pituitary level and indirectly affect TSH synthesis via their effects on TRH and other neuropeptides. TRH is the major positive regulator of hTSHB gene expression and mainly acts by activating the phosphatidylinositol-protein kinase C pathway. Other hormones/factors are also implicated in the complex regulation of TSHB gene expression, as detailed below.

Figure 5. Basic elements in the regulation of thyroid function. TRH is a necessary tonic stimulus to TSH synthesis and release. TRH synthesis is regulated directly by thyroid hormones. T4 is the predominant secretory product of the thyroid gland, with peripheral deiodination of T4 to T3 in the liver and kidney supplying roughly 80% of the circulating T3. Both circulating T3 and T4 directly inhibit TSH synthesis and release independently, T4 after conversion to T3.  SRIH, somatostatin

 

EFFECTS OF THYROID HORMONE ON TSH SYNTHESIS

 

In animal models, thyroid hormone administration is followed by a marked decrease of both alpha and TSHB subunit mRNA expression (60,61), but TSHB is suppressed more rapidly and more completely than the alpha subunit. In humans with primary hypothyroidism a paradoxical increase of serum TSH concentration has been observed shortly after beginning thyroid hormone replacement therapy, followed later by TSH suppression (62). The precise mechanism for this phenomenon has not been fully elucidated: it could be due to a generalized defect in protein synthesis as a consequence of hypothyroidism, or to the presence of a still unrecognized stimulatory thyroid hormone cis-acting element (see below). Thyroid hormone regulation of TSHB subunit transcription is complex and, at least in the rat and mouse, involves control of gene transcription at both start sites of the gene (Fig. 6) (62-69). Studies of the human, rat and mouse TSHB genes have demonstrated that they contain DNA hexamer half sites with strong similarity to the T3 response elements (TREs) found in genes which are positively regulated by thyroid hormone (70-72). The sequences in the TSHB gene are shown in Fig. 6 and their similarity to the typical hexamer binding sites in positively regulated genes and in the rat CGA gene is demonstrated by comparison to the TRE sequences from positively regulated genes (73). In keeping with this concept, T3 exerts similar negative activity on rat GH3 cells transfected with plasmids constructs containing the putative negative TRE of the rat TSHB gene or containing a half-site motif of the consensus positive TRE (74,75,73,69,76). The conserved TRE-like sequences are the best candidate sites in the TSHB gene to which the T3 receptor (TR) binds. The subsequent binding of T3 to TR-DNA complexes suppresses transcription of both the CGA and TSHB genes (77,66,73,69). The inhibitory effect of thyroid hormone is observed with all alpha and beta isoforms of TR, but TR-beta2 (a TR isoform with pituitary and central nervous system-restricted expression) is affected most substantially (78). This in vitro observation is in keeping with a series of in vivo data obtained in transgenic and knockout mice with generalized or pituitary-selective expression of mutated TR isoform genes. Knockout mice for TR-alpha 1 develop only minor abnormalities in circulating T4 and TSH concentration (79), while mice lacking both beta1 and beta2 isoforms (beta-null) develop increased serum T4 and TSH level, but retain partial TSH suppression by T3 administration (80,81). Mice selectively lacking the TR-beta2 isoform develop hormonal abnormalities similar to TR-beta-null animals, indicating a key role of TR-beta2 as a mediator of T3-dependent negative regulation (82). On the other hand, the residual T3-dependent TSH suppression observed in mice lacking TR-beta isoforms suggests that TR-alpha 1 may partially substitute for TR-beta in mediating T3 suppression: accordingly, mice lacking all (alpha and beta) TR isoforms develop dramatic increases in circulating T4 and TSH concentration, indicating that a complete expression of all TR isoforms is required for normal regulation of the hypothalamic-pituitary thyroid axis (83-85). Further studies have been carried out with models of mice expressing selectively at the pituitary (83,86) and hypothalamic (87) level different combinations of double homozygous or combined heterozygous deletions of both TR-alpha and TR-beta genes. These studies confirmed the key role of TR-beta integrity both at the pituitary and hypothalamic level for the inhibition of TSHB and TRH gene expression. TR-alpha however, may partially substitute for TR-beta in mediating a partial thyroid hormone dependent TSH suppression.

 

Figure 6. DNA sequences of the putative TREs in the rat, mouse, and human TSHB gene promoters. A comparison of the proximal promoter regions of the rat, mouse, and human TSHB genes is shown. The straight arrows denote TRE consensus half-sites identified by functional and TR binding assays. The first exons (relative to the downstream promoter for the rat and mouse genes) are shaded, and the bent arrows denote the sites of transcription initiation.  Note a nine-nucleotide deletion in the human gene relative to the rodent genes indicated by the triangle just 5' of the transcriptional start site. (Reproduced from Chin et al. (69) with permission.)

 

The negative transcription conferred by TSH beta TRE sequences is retained even if they are transferred to a different gene or placed in a different position within a heterologous gene (88,89,73,90). This suggests that the negative transcriptional response to thyroid hormone is intrinsic to this TRE structure. In contrast with positive TREs, little is known about the mechanism of T3-dependent negative regulation of genes like TSHB. The data discussed above clearly show the crucial role of the TR-beta in the negative regulation of TSH synthesis. Like for positive TREs, it has been recently established that TR binding to DNA is required for negative gene regulation (91). Early experiments suggested that unliganded TR homodimers stimulate the expression of TSH beta (an effect that is a mirror image of the silencing effect on positive TREs), but the methodology employed was not adequate to study the low level of basal TSHB transcriptional activity. The use of CV1 cell lines containing the TSH beta CAT (chloramphenicol acetyltransferase) reporter allowed a more accurate study of the molecular mechanisms involved in the liganded TR suppression (92). In this experimental system, TSHB gene suppression was dependent on the amounts of T3 and TR, but unliganded TR did not stimulate TSH beta activity, suggesting that TR itself is not an activator. Moreover, recruiting of co-activators and co-repressors were shown to be not necessarily essential, but are required for full suppression of the TSHBa gene (92).

 

In contrast to the potentiating activity exerted on stimulatory TREs, retinoid X receptors (RXR) either unliganded or in combination with retinoic acid (RA) block thyroid hormone-mediated inhibition of the TSHB gene, possibly through competition with the TR-T3 complex binding to DNA (93,76,94,92). However, RA is also able to suppress TSHB gene expression when bound to RAR and RXR interacting with response elements separate from negative TREs (95,96). Taken together, these findings imply that distinct mechanisms are involved in thyroid hormone dependent inhibition and stimulation of TSH synthesis (97,98). Indirect support for this concept derives from the identification of patients with selective pituitary thyroid hormone resistance carrying TR mutations associated with normal or enhanced function on stimulatory TREs in peripheral tissues, but defective function on inhibitory TREs of the TSHB and TRH genes (99).  

 

Another peculiar feature of the negative TSH beta TRE is that its 5' portion (Fig. 6) displays high homology with the consensus sequence of binding sites for c-Jun and c-Fos, which heterodimerize to form the transcription factor called AP-1. This makes the negative TSH beta TRE a "composite element" able to bind both thyroid hormone receptors and AP-1 (100,101,90,99). Since AP-1 antagonizes the inhibition exerted by thyroid hormone in vitro, it may act as a modulator of TRH-dependent regulation of the TSHB gene in vivo (90). The role of other important TSHB gene activity modulators (such as Pit-1 and its splice variants) will be discussed later. Other abnormalities of the mechanisms involved in the negative feed-back on TSH by thyroid hormones could be involved in rare pathological conditions of difficult identification and diagnosis.

 

Since unliganded TR does not behave as an activator of the TSHB gene, other mechanisms are involved in the increase in TSH production observed in hypothyroidism. In the hypothyroid rat TSH production is increased 15 to 20-fold over that in the euthyroid state. This can be attributed to the stimulatory effects of TRH (see below) unopposed by the negative effects of T3; moreover, besides the transcription rate per cell, there is a 3 to 4 fold increase in the absolute number of thyrotrophs in the hypothyroid pituitary (102). Electron microscopic studies have shown near total depletion of secretory granules in the thyrotrophs of hypothyroid animals, a change that is reversed soon after administration of thyroid hormone (103).

 

THYROID HORMONE EFFECTS ON RELEASE OF TSH

 

The acute administration of T3 to the hypothyroid rat causes a rapid and marked decrease in the level of serum TSH (58,104) (Fig. 7). This decrease occurs prior to the decrease in pituitary alpha and beta-TSH mRNAs (104,61,105). During the period that circulating TSH is falling, pituitary TSH content remains unchanged or increases slightly (106). The suppression of TSH release is rapid, beginning within 15 minutes of intravenous T3 injection, but is preceded by the appearance of T3 in pituitary nuclei (106). In the experimental setting in the rat, as the bolus of injected T3 is cleared and the plasma T3 level falls, nuclear T3 decreases followed shortly by a rapid increase in plasma TSH. Both the chronological and quantitative relationships between receptor bound T3 and TSH release are preserved over this time (106).

 

Figure 7. Time course of specific pituitary nuclear T3 binding and changes in plasma TSH in hypothyroid rats after a single intravenous injection of 70 ng T3 per 100 g of body weight.  Since the maximal capacity of thyroid hormone binding in pituitary nuclear proteins is about 1 ng T3/mg DNA, the peak nuclear T3 content of 0.44 ng T3/mg corresponds to 44% saturation.  The plasma level falls to about 55% of its initial basal level by 90 minutes after T3 injection demonstrating that there is both a chronological and a quantitative correlation between nuclear T3 receptor saturation and suppression of TSH release. (From Silva and Larsen (107) with permission).

 

The mechanism for this effect of T3 is unknown. As discussed before, suppression of basal TSH release is difficult to study in vitro. Accordingly, the T3 induced blockade of TRH-induced TSH release has been used as a model for this event. This T3 effect is inhibited by blockers of either protein or mRNA synthesis (108,109). The effect is not specific for TRH since T3 will also block the TSH release induced by calcium ionophores, phorbol ester, or potassium (110,111). Furthermore, T3 will also block the TRH-induced increase in intracellular calcium which precedes TSH release (112). Thus, T3 inhibits TSH secretion regardless of what agent is used to initiate that process.

 

T4 can cause an equally rapid suppression of TSH via its intrapituitary conversion to T3 (104) (Fig. 5). This T4 to T3 conversion process is catalyzed by the deiodinase type 2.  An effect of T4 per se can be demonstrated if its conversion to T3 is blocked by a general deiodinase inhibitor such as iopanoic acid (113,104). In this case, the T4 in the cell rises to concentrations sufficient to occupy a significant number of receptor sites even though its intrinsic binding affinity for the receptor is only 1/10 compared to T3. A similar effect can be achieved by rapid displacement of T4 from its binding proteins by flavonoids (114). It seems likely, however, that under physiological circumstances the feedback effects of T4 on TSH secretion and synthesis can be accounted for by its intracellular conversion to T3.

 

The effect of suppressive doses of T3, T4 and triiodothyroacetic acid on serum TSH has been evaluated in humans by ultrasensitive TSH assays (115). TSH suppression was shown to be a complex, biphasic, nonlinear process, with three temporally distinct phases: phase 1, a rapid TSH suppression, starting after 1 h and lasting for 10-20 h; phase 2, slower suppression, starting between 10 and 20 h and lasting for 6-8 weeks; and phase 3, with stable low TSH level (<0.01 mU/L). This pattern of thyroid hormone suppression of TSH is reproducible and independent of the basal thyroid status or the thyroid hormone analog used.

 

Based on the analyses of the sources of nuclear T3 in the rat pituitary, one would predict that approximately half of the feedback suppression of TSH release in the euthyroid state can be attributed to the T3 derived directly from plasma; the remainder accounted for by the nuclear receptor bound T3 derived from intrapituitary T4 to T3 conversion (104). Various physiological studies in both rats and humans confirm this concept in that a decrease in either T4 or T3 leads to an increase in TSH. The effect of T4 is best illustrated in the iodine deficient rat model (Fig. 8). 

Figure 8. Serum T3, T4, and TSH concentrations (mean ± SD) in rats receiving a low iodine diet (LID), with or without potassium iodide (KI) supplementation in the drinking water. (From Riesco et al. (116) with permission)

 

In this paradigm, rats are placed on a low iodine diet and serum T3, T4, and TSH quantitated at frequent times thereafter (116). Even though serum T3 concentrations remain constant, there is a marked increase in TSH as the serum T4 falls. In humans, severe iodine deficiency produces similar effects (117). The most familiar example of the independent role of circulating T4 in suppression of TSH is found in patients in the early phases of primary hypothyroidism in whom serum T4 is slightly reduced, serum T3 is normal or even increased into the high normal range, but serum TSH is elevated (118,119) (Table 2).

 

Table 2 . Serum concentration of total thyroid hormones and TSH in patients with primary hypothyroidism of increasing severity

 

 

 

TSH (mU/L)

Group*

T4 ug/dl

T3 ng/dl

Basal

After 200 ug TRH

Control

7.1±0.9

115±31

1.3±0.5

11±4.6

1

6-9

119±40

5.3±2.3

39±15

2

4-6

103±20

13±10

92±50

3

2-4

101±35

63±56

196±120

4

<2

43±28

149±144

343±326

Results are mean ± SD.  *Patients were categorized according to the severity of thyroid disease based on serum total T4 concentrations. (Adapted from Bigos et al. (118) with permission)

 

THE ROLE OF THYROTROPIN RELEASING HORMONE (TRH) IN TSH SECRETION

 

TRH is critical for the synthesis and secretion of TSH either in the presence or absence of thyroid hormones. Destruction of the parvo-cellular region of the rat hypothalamus, which synthesizes the TRH relevant for TSH regulation, causes hypothyroidism (120,121). Hypothalamic TRH synthesis is in turn regulated by thyroid hormones and thus TRH synthesis and release are an integral part of the feedback loop regulating thyroid status (see Fig. 5). TRH also interacts with thyroid hormone at the thyrotroph raising the set-point for thyroid hormone inhibition of TSH release (120). The data supporting these general concepts are reviewed in subsequent sections.

 

Control of Thyrotroph-Specific TRH Synthesis  

 

TRH is synthesized as a large pre-pro-TRH protein in the hypothalamus and in several tissues, such as the brain, the beta cells of the pancreas, the C cells of the thyroid gland, the myocardium, reproductive organs including the prostate and testis, in the spinal cord, and in the anterior pituitary (122,123,120,124-127). Recent investigations employing sophisticated techniques such as fast atom bombardment mass spectrometry and gas phase sequence analysis showed that most TRH immunoreactivity found in extrahypothalamic tissues is actually accounted by TRH-immunoreactive peptides displaying different substitutions of the amino-acid histidine of authentic TRH, which could be active in autocrine/paracrine networks involving also extrapituitary TSH secretion (127). On the other hand, pituitary TSH production is dependent only on TRH synthesized in specific areas of the paraventricular nucleus (PVN) (Fig. 9), located at the dorsal limits of the third ventricle (128). In particular, TRH neurons are almost exclusively found in the parvicellular part of the PVN and, while TRH-synthesizing neurons are found in all parvicellular subdivisions of the PVN, hypophysiotropic TRH neurons are located exclusively in the periventricular and medial subdivisions (Fig. 9).

Figure 9. Distribution of TRH-synthesizing neurons in the PVN. Low power micrographs (A–C) illustrate the TRH neurons at three rostrocaudal levels of the PVN. Schematic drawings (D–F) illustrate the subdivisions of the PVN where hypophysiotropic TRH neurons are localized (gray). AP, anterior parvocellular subdivision; DP, dorsal parvocellular subdivision; LP, lateral parvocellular subdivision; MN, magnocellular part of PVN; MP, medial parvocellular subdivision, PV, periventricular parvocellular subdivision; III, third ventricle. (From Fekete & Lechan (128) with permission)

 

Hypophysiotropic TRH neurons project their axons to the median eminence, where TRH is released and drained to the anterior pituitary through the long portal veins (128). Although paracrine and autocrine activity has been recently described for TRH secreted in the anterior pituitary (129), the physiological relevance of pituitary TRH is unknown. The human pre-pro-TRH molecule is a protein of 29 kDa containing 6 progenitor sequences for TRH (130-132). These six peptides consist of a Gln-His-Pro-Gly peptide preceded and followed by Lys-Arg or Arg-Arg di-peptides. The basic di-peptides are the cleavage sites for release of the tetra-peptide progenitor sequence. The glycine residue is the source of the terminal amide for the proline residue of TRH (Fig. 4). In addition to the pro-TRH peptides which are released from the pre-pro TRH molecule, intervening non-TRH peptides which have potential physiological function are co-released (133). In particular, the prepro-TRH fragment 160-169, also known as hST10, TRH-enhancing peptide, and Ps4 (134,135) is able to stimulate TSHB gene expression and to enhance the TRH-induced release of TSH and prolactin (PRL) from the pituitary (136,137,134,138Ps4). Ps4 high affinity receptors have been shown within several extrapituitary neural tissues and other endocrine systems (mainly in the pancreas and the male reproductive system), and targeted pre-pro TRH gene disruption results in hyperglycemia besides the expected hypothyroidism (134). Another pre-proTRH peptide (fragment 178-199) (139,140). appears to be a modulator of ACTH secretion, although the physiological relevance of this phenomenon is unknown. The prepro-TRH processing is mostly mediated by the prohormone convertases PC1 and PC2, and takes place during axonal transport after removal of the signal peptide (138). Subsequent cleavages occur as the peptides move down the axon toward the nerve terminal, from which TRH is released into the hypothalamic-pituitary portal plexus (120,121).

 

Thyroid hormones exert strong negative regulation on TRH synthesis at the hypothalamic level (141-145). Increases in TRH mRNA levels occur during primary or central hypothyroidism and implantation of a small crystal of T3 adjacent to the PVN results in a decrease in TRH mRNA (143). This regulation is observed in vivo exclusively in the parvo-cellular division of the PVN (142,143) (whose neurons contain the functional TR isoforms alpha1, beta2 and beta1 (146)), while in tissues outside the central nervous system expressing the TRH gene, negative regulation by thyroid hormone is absent (147). TR beta2 is the key isoform responsible for T3-mediated feedback regulation by hypophysiotropic TRH neurons (148). Targeted disruption of TR beta2 expression results in increased TRH mRNA expression in the PVN, similar to that found in hypothyroidism. In contrast to the anterior pituitary, where ablation of TR beta2 or the entire TR beta allele produces only partial TH resistance (80,81), the lack of TR beta is associated with a complete resistance of the modulation of TRH synthesis exerted by severe hypo- or hyperthyroidism (148).

 

The physiological source of the T3 causing downregulation of TRH mRNA in the hypothalamus is the subject of ongoing investigations. Somewhat surprisingly, the PVN does not contain the type 2 5' iodothyronine deiodinase (D2) which is thought to be the source of at least 80% of the intracellular T3 in the central nervous system (104,149). However, studies with T3 containing mini-pumps implanted into thyroidectomized rats indicate that, for normalization of circulating TSH and hypothalamic pre-pro-TRH mRNA, T3 concentrations about twice normal have to be maintained in rat plasma (144). Thus, for both systems (TRH and TSH), feedback regulation requires a source of T3 in addition to that provided by the ambient levels of this hormone. While this T3 seems likely to be produced locally from T4, the main anatomical location of such a process has been identified only more recently in the specialized ependymal cells called tanycytes lining the floor and the infralateral wall of the third ventricle between the rostral and caudal poles of the median eminence and the infundibular recess (150,128). Tanycytes are one of the major sources of D2, with D2 mRNA expressed in the cell bodies, in the processes, and in their end feet (128). Originally believed to only serve as part of the blood-brain barrier, tanycytes have complex functions including an active role in endocrine regulation. In particular, T3 locally produced by tanycytes from circulating T4 represents the primary source of T3 involved in the feed-back regulation of hypophysiotropic neurons, unable to express D2 (128). The anatomical location of tanycytes places them in a strategic position to extract T4 from the bloodstream or from cerebrospinal fluid after T4 has traversed the choroid plexus (Fig. 10). Despite their lipophilic nature, the transport of thyroid hormone into the cells require an active processes involving a long list of transporters (151). Two transporter families have been shown to be important in the transport of thyroid hormones in the brain: the monocarboxylate transporter (MCT8)(152) and the organic anion transporting polypeptide (OATP1C1)(153). Several lines of evidence support an important role of MCT8, a member of the MTC family in central nervous system thyroid hormone transport expressed primarily in neurons and in tanycytes. Data from both MCT8 KO mice and from humans with MCT8 mutations indicate that lack of functional MCT8 result in hypothyroid TRH neurons, in spite of high circulating T3 concentration, suggesting that MCT8 is necessary for physiological feed-back regulation (128).

 

Figure 10. Schematic illustration of the feedback system regulating the hypothalamic-pituitary-thyroid axis. Thyroid hormones exert negative feedback effect at the level of hypothalamic TRH neurons and of pituitary gland. The central feedback effect of thyroid hormones depends on the circulating T4 levels. In the hypothalamus, T4 is converted to T3 by D2 in tanycytes. By volume transmission, T3 secreted from tanycytes reaches the hypophysiotropic TRH neurons, where T3 inhibits the proTRH gene expression via TR-β2 receptors. The set point of the feedback regulation can be altered by two mechanisms: (i) regulation of D2 activity in tanycytes may alter the hypothalamic T3 availability independently from the peripheral T4 concentration. (ii) Neuronal afferents can alter the PCREB concentration in the hypophysiotropic TRH neurons that can change the set point of feedback regulation through competition of PCREB and thyroid hormone receptors for the multifunctional binding site (Site 4) of the TRH promoter. ARC, hypothalamic arcuate nucleus; C1-3, C1-3 adrenergic area of the brainstem; CSF, cerebrospinal fluid; DMN, hypothalamic dorsomedial nucleus; ME, median eminence; NTS, nucleus tractus solitarius; PVN, hypothalamic paraventricular nucleus; py, pyramidal tract; sp5, spinal trigeminal tract. (From Fekete & Lechan (128) with permission)

 

The synthesis of TRH is under complex transcriptional control sharing several mechanisms, besides the negative regulation by thyroid hormone, with the TSHB gene. The human TRH gene (Fig. 11) is located on chromosome 3 (3q13.3q21) (154); the 5' flanking sequence of the TRH gene has potential glucocorticoid and cyclic AMP response elements (GRE and CRE) (130). There are also potential negative TREs located in this portion of the gene which offer regulatory sites for thyroid hormone control of TRH gene transcription. The thyroid hormone negative regulatory elements of the TRH gene are localized in its 5' flanking element (-242 to +54 bp). Four sequences within this region exhibit a high degree of homology with the consensus sequences for TRE half-sites (AGGTCA) and two of them also show homology with elements implicated in negative regulation by thyroid hormone of the TSHB gene (147). In the absence of thyroid hormone, proTRH gene expression as well as prohormone convertase enzymes (PC1/3 and PC2) are increased in the PVN, while the content of TRH in the median eminence is decreased due to increased secretion of the mature hormone in the portal circulation (128). In contrast, hyperthyroidism is associated with decreased proTRH-mRNA in the PVN (128). The negative feed-back of thyroid hormones is exerted directly on hypophysiotropic TRH neurons of the PVN which express all thyroid hormone receptor isoforms. The recent availability of transgenic mice lacking either TRH, TR-beta isoforms, or both provided evidence for a pivotal role of TRH in the physiological TH feed-back on the hypothalamic pituitary-thyroid (HPT) axis (155). Double TSH and TR-beta knockout mice had reduced TH and TSH levels associated with low TSH content in pituitary thyrotrophs and both serum TSH and pituitary TSH content was increased by chronic exogenous TRH administration (156). Thus, the TRH neuron appears to be required for both TSH and TH synthesis and is the predominant locus of control of the HPT axis (155).  However, studies carried out with different animal models of congenital hypothyroidism show that the thyrotrophs exhibit hyperplasia and hypertrophy along with increased TSH mRNA expression not only in the athyreotic Pax8-/- mice, but also in TRHR1-/- Pax8-/- double-knockout mice, which miss a functional thyroid gland and the TRH receptor at the pituitary level, suggesting that the stimulation of thyrotroph proliferation and TSH synthesis is rather a direct consequence of the continue here athyroidism of the animals (157). Further studies are therefore required to determine the relative contributions of TRH and TH for bioactive pituitary TSH release.

 

As shown in Fig. 11, the TRH gene promoter contains potential binding sites for cAMP response element (CRE) binding protein (CREB), and both human and rat TRH genes are positively regulated by cAMP (147). One of the potential CREs of TRH promoter is a sequence that has overlapping TRE/CRE bases –53 to –60 bp (TGACCTCA) (147). There is evidence for competitive interactions of TR beta1 and CREB at the overlapping TRE/CRE in the TRH promoter (147). Constructs of the TRH promoter with mutations in this overlapping site prevent both the inhibition by the TR-T3 complex and the basal activation in the presence of unliganded TR, underlining the relative importance of the TRE/CRE site in relation to the other TREs in the TRH promoter (147).

Figure 11. Genomic and promoter structure of the TRH gene. The murine, rat and human TRH genes are composed of three exons and two introns (A). The coding sequence for the precursor protein is present on exons 2 and 3. As depicted, the TRH promoter region precedes the transcription start site in exon 1. The proximal 250-bp sequences of the human, mouse and rat promoters are similar and share the indicated transcription factor binding sites. The location of the CREB binding site (Site 4) and sequences in human (H), mouse (M) and rat (R) are shown. (B, C) Hypothesized schematic representation of the interaction between PCREB and the thyroid hormone receptor at Site 4. (B) Illustrates that in the presence of abundant PCREB, there may be less availability for binding of the thyroid hormone receptor/T3 complex, hence, an increase in TRH gene transcription. When PCREB concentrations fall as shown in (C), increased binding of the thyroid hormone receptor/T3 complex reduces TRH gene transcription (From Fekete & Lechan (128) with permission)

 

A glucocorticoid-responsive element (GRE) is also present in the TRH gene promoter (130). and the glucocorticoid receptor has been identified on TRH neurons of the PVN (158). The role of corticosteroids in TRH gene expression is unclear, since both inhibitory and stimulatory effects have been reported (159,160). The direct effect of glucocorticoids on TRH gene expression is generally stimulatory in vitro, but in vivo this activity may be overridden by the complex neuroendocrine reactions following glucocorticoid excess or deficiency (159).

 

TRH INTERACTION WITH PITUITARY THYROTROPHS AND WITH THYROID HORMONE

 

Although TRH (either maternal or embryonic) is not required for the normal development of fetal pituitary thyrotrophs, and TRH-deficient mice are not hypothyroid at birth, TRH is required later for the postnatal maintenance of the normal thyrotroph function (161). TRH exerts its activity binding to a specific receptor in the plasma membrane of the thyrotroph to induce the release of TSH and to stimulate TSH synthesis. The TRH receptor of several animal species (including humans) has been cloned and has been identified as a G-protein-coupled receptor with seven highly conserved transmembrane domains (162-165). Biallelic inactivating mutations in the 5’-part of the TRH receptor gene are one of the molecular causes for central congenital hypothyroidism (166-169). TRH-receptor number and mRNA are increased by glucocorticoids and decreased by thyroid hormone, as well as by TRH itself (170,171). The second messenger for induction of the thyrotroph response to TRH is intracellular Ca2+ ([Ca2+]i) (172-174). TRH was previously believed to act also through stimulation of the adenyl cyclase-cAMP pathway (120), but this mechanism has not been confirmed by studies carried out with recombinant TRH-receptor transfected in different cell systems (175). TRH activates a complex [Ca2+]i response pattern dependent upon both agonist concentration and cell context. The first phase of the TRH response is an acute increase of [Ca2+]i within the thyrotrophs via release from internal stores. This is the consequence of increased inositol triphosphate concentrations from hydrolysis of phosphatidyl inositol (PI) in the cell membrane (176,173,177,178). The hydrolysis of PI is mediated by G protein activation of phospholipase C and also generates diacylglycerol, which in turn activates intracellular protein kinase C (PKC). Stimulation of extracellular calcium influx through verapamil-sensitive channels is also observed after TRH stimulation (172,179). Both TRH and increased [Ca2+]i stimulate intracellular calcium efflux, which helps in terminating the agonist activity (177,179,180). In transfection systems in which the TSHB gene promoter has been linked to a reporter gene, both the calcium ionophore ionomycin and phorbol esters (a protein kinase C activator) stimulate TSH gene transcription, confirming the key role of these second messengers in mediating TRH activity (66). Both increased [Ca2+]i and PKC appear to be independently operative in normal thyrotrophs (181).

 

The molecular mechanism(s) underlying the stimulation of TSHB gene expression by TRH have been partially elucidated. In GH3 cells transfected with hTSHB promoter constructs, two distinct regions of the human TSHB gene responding positively to stimulation by TRH were identified between -130 and +37 bp of the gene (182-184) (Fig. 12) The 3'-region corresponds to eight bp of the first exon; the 5'-region ranged between -128 to -60 bp of the 5'-flanking region (182,183).

Figure 12. The 5’ flanking sequence of the human preproTRH gene between –192 and +58 bp. Four potential thyroid response element (TRE, boxed) and two potential CREB binding elements (CRE, underlined) are shown. One sequence (from –60 to –53 bp) consists of overlapping TRE/CRE sites (bold). (Modified from Wilber & Xu (147))

 

INACTIVATION OF TRH

 

TRH is rapidly inactivated within the central nervous system by a cell-surface peptidase called TRH-degrading ectoenzyme (TRH-DE) (185). TRH-DE is very specific, since there is no other ectopeptidase known capable of degrading TRH and TRH is the only known substrate of this unique enzyme (185). TRH-DE has been purified to homogeneity and cDNA encoding rat TRH-DE has been cloned. In rodents, pituitary TRH-DE mRNA and enzymatic activity are stringently positively regulated by thyroid hormones, and reduced by estrogens (185). This suggests that TRH-DE may act as a regulatory element modulating pituitary TSH secretion. The expression of TRH-DE in the brain is high and displays a distinct distribution pattern, but it is not influenced by peripheral hormones, supporting the concept that brain TRH-DE may act as a terminator of TRH signals (185).

OTHER FACTORS INVOLVED IN THE REGULATION OF TSH/TRH SYNTHESIS AND SECRETION

A number of other substances, including ubiquitous and pituitary or thyrotroph-specific transcription factors, hormones, neuropeptides and cytokines influence TSH synthesis and secretion of TRH (Table 3, Fig. 11&13).

 

Table 3. Predominant Effects of Various Agents on TSH Secretion

STIMULATORY

INHIBITORY

Thyrotropin-releasing hormone (TRH)

Thyroid hormones and analogues

Prostaglandins (?)

Dopamine

Alpha-adrenergic agonists (? Via TRH)

Somatostatin

Opioids (humans)

Gastrin

Arginine-vasopressin (AVP)

Opioids (rat)

Glucagon-like peptide 1 (GLP-1)

Glucocorticoids (in vivo)

Galanin

Serotonin

Leptin

Cholecystokinin (CCK)

Glucocorticoids (in vitro)

Gastrin-releasing peptide (GRP)

 

Vasopressin (AVP)

 

Neuropeptide Y (NPY)

 

Interleukin 1 beta and 6

 

Tumor necrosis factor alpha

 

Role of Pit-1 and its Splicing Variants in the Regulation of TSHB Gene Expression

Sequence analysis of the hTSHB promoter reveals three areas with high (75-80%) homology to the consensus sequence for the pituitary-specific transcription factor Pit-1 (182,183,186,184). These areas are localized between -128 and -58 bp of the 5'-flanking region. Selective mutation analysis revealed that the integrity of these areas was needed for the stimulatory effect of either TRH or forskolin (187). Expression of an inactive mutant of Pit-1 decreases TRH stimulation of hTSHB (183) and transfection of Pit-1 in cell lines lacking this factor restores cAMP induction of the hTSHB gene (186). Taken together, these results strongly support an important role of Pit-1 in the regulation of hTSHB gene expression. Phosphorylation markedly increases the stimulatory activity of Pit-1 in TSHB gene expression (187), and TRH stimulates transient phosphorylation of Pit-1 in GH3 pituitary cells (188).

 

Further support for a role of Pit-1 in the regulation of TSHB gene expression derives from animal models (dwarf mice) and from clinical syndromes of combined pituitary hormone deficiency (CPHD) (189,167). Snell and Jackson dwarf mice lack a functioning Pit-1 protein due to a point mutation and a gross structural rearrangement in the Pit-1 gene, respectively (190). Both species show low serum concentration of GH, prolactin and TSH associated with the loss of somato-, lacto- and thyrotropic pituitary cells. Several Pit-1 point mutations and a deletion of the entire coding sequence have been described in patients with CPHD: the effects on TSH secretion differ with the localization of the mutation, but generally result in central hypothyroidism (191-194,94,189,195,196). Finally, the important role of Pit-1 in the control of TSH synthesis and secretion has been documented by the finding that circulating Pit-1 antibodies are associated with combined GH, prolactin, and TSH deficiency, the so called “anti-PIT-1 antibody syndrome” (197-200).

 

Although important, the role of Pit-1 for cell-specific expression of TSHB is not as clear as with the GH and PRL genes (201,184). Attention has been focused on thyrotropin-specific transcription factors, including Pit-1 splicing variants. Of those, a variant called Pit-1T (containing a 14 amino-acid insertion in the transactivation domain) is found only in thyrotropic cells expressing TSHB and it increases TSHB promoter activity when transfected in non-thyrotropic cells expressing wild type Pit-1 (202,203). These results suggest that the combination of both Pit-1 and Pit-1T may have a synergistic stimulatory effect on TSHB promoter activity (204).

Other Transcription Factors Involved in TSHB Gene Expression

 

As stated above, the transcription factor AP-1 may be involved in modulating regulation of TSHB gene expression mediated by thyroid hormone (Fig. 13). Accordingly, a potential AP-1 binding site is present between -1 to +6 bp of the TSHB gene (184), and the integrity of this site is required for maximal stimulation of  the hTSHB gene (205). Haugen et al. (206) described a new 50 kd thyrotroph-specific protein whose binding together with Pit-1 is needed for optimal basal expression of the mouse TSHB gene; this factor was subsequently identified as the transcription factor GATA-2 (207). GATA-2 stimulates the mouse TSHB promoter synergistically with Pit-1 and is needed for optimal TSHB gene basal activity. Another pituitary-specific protein (P-Lim), which binds and activates the common glycoprotein hormone alpha subunit promoter, also synergizes with Pit-1 in the transcriptional activation of the TSHB gene in mice (208). Moreover, characterization of the dwarfed Ames (df) mouse led to the cloning of the paired-like homeodomain factor Prop-1 (Prophet of Pit-1) (209). PROP-1 is necessary for Pit1 expression. Biallelic mutations in the human PROP-1 gene have been identified as a further cause of CPHD phenotype affecting somatotropes, lactotropes, and thyrotropes (210,189,167,211).

 

Figure 13. The regulatory region of human TSHB gene (see text for details)

 

cAMP

 

An increase in intracellular cAMP stimulates expression of both the common CGA and TSHB subunit genes (182). In contrast to the TRH gene, this action of cAMP is probably not mediated through direct binding of CREB to a CRE sequence, but by promoting Pit-1 phosphorylation with subsequent activation of the TSHB promoter  (183,186).

Steroid Hormones

 

Steroid hormones including corticosteroids, estrogen and testosterone modulate TSHB gene expression. Dexamethasone in pharmacological doses decreases serum TSH concentrations in normal subjects (212), in patients (213), and rats (214) with TSH-secreting pituitary adenomas, but does not significantly change TSH subunit mRNA levels (214). This suggests that glucocorticoids may act on TSH biosynthesis at a translational or post-translational level. Furthermore, as discussed before for the TRH gene, several other neuroendocrine mechanisms may participate in vivo in the modulation of TSH synthesis and secretion by glucocorticoids. In keeping with this concept, it has been shown in humans that enhanced hypothalamic somatostatinergic and dopaminergic inhibitory activities are involved in the glucocorticoid-dependent blunting of the TSH response to TRH (215).

 

Estrogens and testosterone have limited direct effects on TSH synthesis and secretion in humans. Estrogens mildy reduce mRNA levels coding for the alpha and beta TSH subunits in hypothyroid rats (216), perhaps interacting with the same response elements involved in thyroid hormone regulation. Testosterone has similar effects, at least in part explained by its peripheral conversion to estrogen (217).

Other Hormones, Neuropeptides and Cytokines

 

Somatostatin, the major physiological inhibitor of GH secretion, is also an inhibitor of TSH secretion in rats and humans (218-220). The physiological relevance of this inhibition is suggested by studies carried out with antibodies to somatostatin whose administration in rats increases serum TSH in basal conditions and after TRH or cold-exposure (212). Indirect evidence for a physiological role of somatostatin in the regulation of TSH secretion has been obtained in humans by the demonstration that stimulation of the endogenous somatostatin tone by oral glucose inhibits TSH response to TRH (221). The TSH-inhibiting activity of somatostatin is an acute phenomenon, while long-term treatment with somatostatin analogues does not cause hypothyroidism in man (222,223), presumably because the effects of the initial decrease in serum thyroid hormone concentration overrides the inhibitory effects of somatostatin. Somatostatin binds to five distinct types of receptors expressed in the anterior pituitary and brain and differing in binding specificities, molecular weight, and linkage to adenylyl cyclase (224). Binding of somatostatin to its receptor causes activation of Gi proteins which in turn inhibit adenylyl cyclase. Somatostatin also induces cellular hyperpolarization via modulation of voltage-dependent potassium channels (225). This mechanism is cAMP-independent and leads to a fall of [Ca2+]i by reducing extracellular calcium influx (226).

 

In animal models, TSH secretion is affected by other hypothalamic hormones: in particular, corticotropin-releasing hormone (CRH) stimulates TSH secretion in chickens (227) through an interaction with CRH-receptor-2 (228), and melanin-concentrating hormone (MCH) suppresses in vivo and in vitro TSH release in rats (229).

 

Neurotransmitters are important direct and indirect modulators in TSH synthesis and secretion. A complex network of neurotransmitter neurons terminates on cells bodies of hypophysiotropic neurons and several neurotransmitters (such as dopamine) are directly released into hypophysial portal blood exerting direct effects on anterior pituitary cells. Furthermore, many dopaminergic, serotoninergic, histaminergic, catecolaminergic, opioidergic, and GABAergic systems project from other hypothalamic/brain regions to the hypophysiotropic neurons involved in TSH regulation. These projections are important for a normal TSH circadian rhythm, response to stress, and cold exposure, while basal TSH secretion is mainly regulated by intrinsic hypothalamic activity (230-232). Despite the difficulty to precisely identify the relative contributions of different neurotransmitter systems in the regulation of TSH secretion, the role of some of them (particularly dopamine and catecholamines) has been rather well defined.

 

Dopamine, acting via the DA2 class of dopamine receptors, inhibits TSH synthesis and release; similarly, to somatostatin, this activity is exerted through a decrease in adenylate cyclase (233-235). Dopamine also inhibits mRNA coding for alpha and TSHB subunits and gene transcription in cultured rat anterior pituitary cells (77). In contrast, with its inhibitory activity at the thyrotroph level, dopamine at the hypothalamic levels stimulates both TRH and somatostatin release (236,237), with an opposite effect on TSH secretion.

 

In contrast to dopamine, adrenergic activation positively regulates TSH secretion. Central stimulation of alpha-adrenergic pathways increases TSH release in rats, presumably through stimulation of TRH secretion. Furthermore, alpha1 adrenergic agonists also enhance TSH release from pituitary cells in vitro by mechanisms which are independent of those activated by TRH (238,239,236,237). It is thought that alpha-adrenergic activity on thyrotrophs is linked to adenylate cyclase activation since agents increasing intracellular cyclic AMP in these cells can increase TSH release (240-242).

 

Opioids inhibit TSH secretion in rats and this action is blocked by the antagonist naloxone (243), while in humans they appear to exert a stimulatory effect, especially on the nocturnal TSH surge (244,232).  Several other neuropeptides may affect TSH secretion in vivo or in vitro. Cholecystokinin (CCK) (245), gastrin-releasing peptide (GRP) (246), and neuropeptide Y (NPY) (247) exert inhibitory effects, while arginine-vasopressin (AVP) (248), glucagon-like peptide-1 (GLP-1) (249), galanin (250), and leptin (251,252) stimulate TSH secretion. Although the precise physiological role of these peptides remains to be clarified, it has been recently suggested that they may be important in connecting nutrition status and thyroid function (253), as discussed in more detail later.

 

Cytokines have recently been demonstrated to have important effects on TRH or TSH release. Both interleukin 1 beta (IL-1 beta) and tumor necrosis factor alpha (cachectin) inhibit TSH basal release (254-257), while no inhibition is observed on TSH response to TRH (258), and this effect is independent of thyroid hormone uptake or receptor occupancy. At the same time, IL-1 beta stimulates the release of corticotropin-releasing hormone and activates the hypothalamic-pituitary-adrenal axis (259). Interleukin-1 beta is produced in rat thyrotrophs, and this production is markedly increased by bacterial lipopolysaccharide (260,261). It could thus reduce TSH secretion by either autocrine or paracrine mechanisms. The IL-1 beta-dependent cytokine interleukin 6 (IL-6) exerts similar inhibitory effects on TSH secretion. Both IL-1 beta and IL-6 acutely inhibit TSH release from the thyrotrophs, while IL-1 beta (but not IL-6) also decreases hypothalamic TRH mRNA and gene expression (262,146,263). Both IL-1 beta and IL-6 stimulate 5’-deiodinase activity in cultured pituitary cells (264), suggesting that increased intrapituitary T4T3 conversion may be involved in the inhibitory activity on TSH production. IL-6 is produced by the folliculo-stellate cells of the anterior pituitary (265,266), and, like IL-1 beta may regulate TSH release in a paracrine fashion (263,259). As discussed later, increased concentrations of circulating pro-inflammatory cytokines are involved in the alterations of hypothalamic-pituitary-thyroid axis observed in non-thyroidal illnesses.

 

SIRT1, a NAD-dependent deacetylase, has been proven to be important for TSH secretion by thyrotrophic cells by the SITR1-phosphatidylinositol-4-phosphate 5-kinase-gamma pathway (267).

 

In summary, an intricate set of relationships within and outside the central nervous system controls the TRH-producing neurons in the medial basal hypothalamus. Alterations in any of these mechanisms can influence TRH and consequently TSH release (Fig 13 and 14). The relative importance in human physiology of these neural pathways, which have been directly studied only in animal models, is unknown.

Figure 14. Schematic representation of the main factors interacting in the regulation of TSH synthesis and secretion (DA: dopamine; SS: somatostatin; α-AD: α adrenergic pathways). Red arrows: stimulation; blue blunted arrows: inhibition

 

SHORT AND ULTRA SHORT-LOOP FEEDBACK CONTROL OF TSH SECRETION

 

In additional to the classic negative feed-back of thyroid hormone on TSH and TRH secretion detailed in the above paragraphs, evidence is accumulating that pituitary TSH is able to inhibit TRH secretion at the hypothalamic level (short feedback) and TSH secretion at the pituitary level (ultra-short feedback) (268). Early observations of inhibition of TSH secretion by injection of pituitary extracts have been recently corroborated by the demonstration of TSH receptor expression (together with other pituitary hormone receptors) in the hypothalamus (269,270) and in the folliculo-stellate cells of the adenohypophysis (271). The precise physiological role of short and ultra-short feedback in controlling TRH/TSH secretion remains to be elucidated. It may be speculated that they concur in the fine tuning of the homeostatic control and in the generation of the pulsatility of TSH secretion. The possibility that thyroid-stimulating autoantibodies present in Graves’ disease recognize hypothalamic and pituitary TSH receptors has also been suggested to explain suppressed serum TSH levels in some euthyroid Graves’ patients (268).

Summary of the Main Steps Involved in the Hypothalamic-Pituitary-Thyroid (HPT) Axis

 

An attempt to summarize the main steps involved in the feedback regulation of the HPT axis is illustrated in Fig 14 (128). Thyroid hormones inhibit the effects of TRH on TSH release without interfering with TRH binding to its receptors, but exerting complex negative transcriptional and post-transcriptional activities on TSH synthesis and secretion discussed above. Several factors other than thyroid hormones are involved in the fine regulation of HPT axis as depicted in Fig. 13 and described in more detail in the following paragraphs.

 

PHYSIOLOGICAL REGULATION OF TSH SECRETION IN HUMANS

 

A number of experimental paradigms have been used to mimic clinical situations that affect the hypothalamic-pituitary thyroid axis in man. However, with the exception of the studies of thyroid status and iodine deficiency, such perturbations have limited application to humans due to differences in the more subtle aspects of TSH regulation between species. For example, starvation is a severe stress and markedly reduces TSH secretion in rats, but only marginally in humans. Cold stress increases TSH release in adult rats by alpha-adrenergic stimulation, while this phenomenon is usually not observed in the adult human. Thus, it is more relevant to evaluate the consequences of various pathophysiological influences on TSH concentrations in humans rather than to extrapolate from results in experimental animals. This approach has the disadvantage that, in many cases, the precise mechanism responsible for the alteration in TSH secretion cannot be identified. This deficit is offset by the enhanced relevance of the human studies for understanding clinical pathophysiology.

 

Table 4.  Common Polymorphisms Related to Serum Thyroid Hormones and TSH Variation (270)

Gene

Polymorphism

Effect on serum

 

 

TSH

T4

T4/T3

T3

rT3

T3/rT3

TSHR

rs10149689 A/G*

­

=

=

=

=

=

 

rs12050077 AG

­

=

=

=

=

=

DIO1

D1a-C/T

=

 

 

¯

­

¯

 

D1b-A/G

=

 

 

­

¯

 

 

rs2235544 C/A

=

 

 

­

¯

­

DIO2

D2-ORFa-Asp3

=

­1

=

=

=

=

 

Thr92Ala

=

=

=

=

=

=2

 

rs225014 C/T

=

=

=

=

=

=3

THRB

TRHB-in9 A/G

(­)

=

=

=

=

=

PDE8B

rs4704397 A/G

­

=

=

=

=

=

* Alleles associated with the specified trait are reported in bold; 1 Only in young subjects;

2 Influence L-T4 dose needed to normalize serum TSH in hypothyroid patients; 3 Influence psychological well-being of hypothyroid patients on L-T4 therapy

 

Normal Physiology

 

The concentration of TSH can now be measured with exquisite sensitivity using immunometric techniques (see below). In euthyroid humans, this concentration ranges from 0.4-0.5 to 4.0-5.0 mU/L. This normal range is to some extent method-dependent in that the various assays use reference preparations of slightly varying biological potency. The glycosylation of circulating TSH is different from that of standard TSH, thus preventing the calculation of a precise molar equivalent for TSH concentrations (272,273). Recently, a narrower range (0.5-2.5 mU/L) has been proposed in order to exclude subjects with minimal thyroid dysfunction, particularly subclinical hypothyroidism (274), but the issue is still controversial (275). Moreover, data form large epidemiological studies mostly carried out in iodine sufficient countries like the USA, suggest that age together with racial/ethnic factors may significantly affect the respective “normal” TSH range, with higher levels for older Caucasian subjects (276,277). These data differ from the findings previously reported in selected small series of healthy elderly subjects (278) suggesting an age-associated trend to lower serum TSH concentrations (see below). The reason(s) for such discrepancies are still not understood. Independently from the “true” normal range of serum TSH, there is substantial evidence that this is genetically controlled, the heritability being estimated between 40-65% (279).  As reported in Table 4, polymorphisms of several genes encoding potentially involved in the control of HPT axis show a significant association with serum TSH concentrations (280) and PDE8B, a gene encoding a high-affinity phosphodiesterase catalyzing the hydrolysis and inactivation of cAMP, has been shown by genome-wide association study to be one of the most important (281).

 

The free alpha subunit is also detectable in serum with a normal range of 1 to 5 µg/L, but free TSHB is not detectable (4,282). Both the intact TSH molecule and the alpha subunit increase in response to TRH. The alpha subunit is also increased in post-menopausal women; thus, the level of gonadal steroid production needs to be taken into account in evaluating alpha subunit concentrations in women. In most patients with hyperthyroidism due to TSH-producing thyrotroph tumors, there is an elevation in the ratio of the alpha subunit to total TSH (4,16,283,182,184). In the presence of normal gonadotropins, this ratio is calculated by assuming a molecular weight for TSH of 28,000 and of 13,600 Da for the alpha subunit. The approximate specific activity of TSH is 0.2 mU/mg. To calculate the molar ratio of alpha subunit to TSH, the concentration of the alpha subunit (in ug/L) is divided by the TSH concentration (in mU/L) and this result multiplied by 10. The normal ratio is <1.0 and it is usually elevated in patients with TSH-producing pituitary tumors but it is normal in patients with thyroid hormone resistance unless they are post-menopausal (284).

 

The volume of distribution of TSH in humans is slightly larger than the plasma volume, the half-life is about 1 hour, and the daily TSH turnover between 40 and 150 mU/day (283). Patients with primary hypothyroidism have serum TSH concentrations greater than 5 and up to several hundred mU/L (118). In patients with hyperthyroidism due to Graves' disease or autonomous thyroid nodules, TSH is suppressed with levels which are inversely proportional to the severity and duration of the hyperthyroidism, down to levels as low as <0.004 mU/L (285-287).

 

TSH secretion in humans is pulsatile (288-290). The pulse frequency is slightly less than 2 hours and the amplitude approximately 0.6 mU/L. The TSH pulse is significantly synchronized with PRL pulsatility: this phenomenon is independent of TRH and suggests the existence of unidentified underlying pulse generator(s) for both hormones (291). The frequency and amplitude of pulsations increases during the evening reaching a peak at sleep onset, thus accounting for the circadian variation in basal serum TSH levels (292,293). The maximal serum TSH is reached between 21:00 and 02:00 hours and the difference between the afternoon nadir and peak TSH concentrations is 1 to 3 mU/L. Sleep prevents the further rise in TSH as reflected in the presence of increases in TSH to 5-10 mU/ml during sleep deprivation (294,295). The circadian variation of TSH secretion is probably the consequence of a varying dopaminergic tone modulating the pulsatile TSH stimulation by TRH (296). Interestingly, TSH molecules secreted during the night are less bioactive and differently glycosylated than those circulating in the same individual during the day, thus explaining why thyroid hormone levels do not rise after the nocturnal TSH surge (296). There is convincing evidence seasonal change in basal TSH (297), but there are no gender-related differences in either the amplitude or frequency of the TSH pulses (290). The diurnal rhythmicity of serum TSH concentration is maintained in mild hyper- and hypothyroidism, but it is abolished in severe short-term primary hypothyroidism, suggesting that the complete lack of negative feedback to the hypothalamus or pituitary or both may override the central influences on TSH secretion (298).

 

TSH in Pathophysiological States

 

NUTRITION

 

In the rat, starvation causes a marked decrease in serum TSH and thyroid hormones.  While there is an impairment of T4 to T3 conversion in the rat liver due to a decrease in both thiol co-factor and later in the Type 1 deiodinase (302-304), the decrease in serum T3 in the fasted rat is primarily due to the decrease in T4 secretion consequent to TSH deficiency (304,305). In humans, starvation and moderate to severe illness are also associated with a decrease in basal serum TSH, pulse amplitude and nocturnal peak (306-310). In the acutely-fasted man, serum TSH falls only slightly and TRH responsiveness is maintained, although blunted (311,312). This suggests that the thyrotroph remains responsive during short-term fasting and that the decrease in TSH is likely due to changes secondary to decreased TRH release. There is evidence to support this in animal studies, showing reduced TRH gene expression in fasted rats (313,314). Administration of anti-somatostatin antibodies prevents the starvation induced serum TSH falls in rats, suggesting a role for hypothalamic somatostatinergic pathways (315). However, fasting-induced changes in dopaminergic tone do not seem to be sufficient to explain the TSH changes (315,309).

 

Recent studies provide compelling evidence that the starvation-induced fall in leptin levels (Fig. 15) plays a major role in the decreased TSH and TSH secretion of fasted animals and, possibly, humans (251,316,317). This concept stems from the observation that administration of leptin prevents the starvation-induced fall of hypothalamic TRH (318). The mechanisms involved in this phenomenon include decreased direct stimulation by leptin of TRH production by neurons of the PVN (251,319), as well as indirect effects on distinct leptin-responsive neuroendocrine circuits communicating with TRH neurons (318,320). The direct stimulatory effects of leptin on TRH production are mediated by binding to leptin receptors, followed by STAT3 activation and subsequent binding to the TRH promoter (321,322). One of the latter circuits has been identified in the melanocortin pathway, a major target of leptin action. This pathway involves 2 ligands expressed in distinct populations of arcuate nucleus neurons in the hypothalamus [the alpha-MSH and the Agouti receptor protein (AgRP)] and the melanocortin 4 receptor (MC4R) on which these ligands converge, but exert antagonistic effects (stimulation by alpha-MSH; inhibition by AgRP). Leptin activates MC4R by increasing the agonist alpha-MSH and by decreasing the antagonist AgRP and this activation is crucial for the anorexic effect of leptin. The specific involvement of the melanocortin pathway in TRH secretion is suggested by the presence of alpha-MSH in nerve terminals innervating hypothalamic TRH neurons in rat (128) and human (323) brains and by the ability of alpha-MSH to stimulate and of AgRP to inhibit hypothalamus-pituitary thyroid axis both in vitro and in vivo (319). The activities of alpha-MSH and AgRP on the thyroid axis are fully mediated by MCR4, as shown by experiments carried out in MCR4 knock out mice (324). Fasting may inhibit the hypothalamic-pituitary-thyroid axis also via the orexigenic peptide NPY, which inhibits TRH synthesis by activation of Y1 and Y5 receptors in hypophysiotropic neurons of the hypothalamic paraventricular nucleus (325). At least two distinct populations of NPY neurons innervate hypophysiotropic TRH neurons (326), suggesting that NPY is indeed an important regulator of the hypothalamic-pituitary-thyroid axis.

 

A further contributing cause to the decreased TSH release in fasting may be an abrupt increase in the free fraction of T4 due to the inhibition of hormone binding by free fatty acids (327). This would cause an increase in pituitary T4 and, hence, in pituitary nuclear T3. Fasting causes a decrease in the amplitude of TSH pulses, not in their frequency (328).

 

Ingestion of food results in an acute decline of the serum TSH concentration: this is the consequence of meal composition, rather than stomach distension (329). Long-term overfeeding is associated to a transient increase of serum T3 concentration and a sustained increased response of TSH to TRH (330).

 

Taken together, the above data provide compelling evidence that the hypothalamic-pituitary-thyroid axis is tightly related to the mechanisms involved in weight control. In keeping with this concept, several epidemiological studies suggest that small differences in thyroid function may be important for the body mass index and the occurrence of obesity in the general population (331-334).

 

ILLNESS

 

The changes in circulating TSH which occur during fasting are more exaggerated during illness. In moderately ill patients, serum TSH may be slightly reduced but the serum free T4 does not fall and is often mildly increased (327,335-337). However, if the illness is severe and/or prolonged, serum TSH will decrease and both serum T4 (and of course T3) decrease during the course of the illness. This may be due to a decreased pulse amplitude and nocturnal TSH secretion (338-341). Since such changes are short-lived, they do not usually cause symptomatic hypothyroidism. They are often associated with an impaired TSH release after TRH (306). However, the illness-induced reductions in serum T4 and T3 will often be followed by a rebound increase in serum TSH as the patient improves. This may lead to a transient serum TSH elevation in association with the still subnormal levels of circulating thyroid hormones and thus be mistaken for primary hypothyroidism (342). On occasion, a transient TSH elevation occurs while the patient is still ill. The pathophysiology of this apparent resistance of the thyroid gland to TSH is not clear (343), although this phenomenon could be the consequence of reduced TSH bioactivity, possibly a consequence of abnormal sialylation (344). The transient nature of these changes is reflected in normalization of the pituitary-thyroid axis after complete recovery. It is currently not clearly established whether the above abnormalities in hypothalamic-pituitary-thyroid axis during critical illness reflect an adaptation of the organism to illness or instead a potentially harmful condition leading to hypothyroidism at the tissue level (345,346).

 

NEUROPSYCHIATRIC DISORDERS

 

Certain neuropsychiatric disorders may also be associated with alterations in TSH secretion.  In patients with anorexia nervosa or depressive illness, serum TSH may be reduced and/or TRH-induced TSH release blunted (347). Such patients often have decreases in the nocturnal rise in TSH secretion (293). The etiology of these changes is not known although it has been speculated that they are a consequence of abnormal TRH secretion (348,349). The latter is supported by observations that TRH concentrations in cerebrospinal fluid of some depressed patients are elevated (350,351). There may be a parallel in such patients between increases in TRH and ACTH secretion (352). The increased serum T4 and TSH levels sometimes found at the time of admission to psychiatric units is in agreement with this concept (353,349).

 

MECHANISMS INVOLVED IN THE HYPOTHALAMIC-PITUITARY-THYROID AXIS SUPPRESSION IN NON-THYROIDAL ILLNESSES    

 

The precise mechanism(s) underlying the suppression of the hypothalamic-pituitary-thyroid axis in severe illnesses are only partially known. Evidence for a direct involvement of TRH-producing neurons in humans has been recently provided by the demonstration of low levels of TRH mRNA in the PVN of patients who died of non-thyroidal disease (354). Alterations in neuroendocrine pathways including opioidergic, dopaminergic and somatostatinergic activity have been suggested, but in acutely ill patients the major role appears to be played by glucocorticoids (355) (See below for a more detailed discussion). Activation of pro-inflammatory cytokine pathways is another mechanism potentially involved in the suppression of TSH secretion in nonthyroidal illness. As discussed earlier, IL-1 beta, TNF-alpha and IL-6 exert in vivo and in vitro a marked inhibitory activity on TRH-TSH synthesis/secretion. High levels of pro-inflammatory cytokines (particularly IL-6 and TNF-alpha) have been described in sera of patients with non-thyroidal illnesses (356,357,262,358,359). Serum cytokine concentration is directly correlated with the severity of the underlying disease and to the extent of TSH and thyroid hormone abnormalities observed in these patients. Furthermore, cytokines also affect thyroid hormone secretion, transport and metabolism providing all the characteristics to be considered important mediators of thyroid hormone abnormalities observed in non-thyroidal illness (360-362).

 

EFFECTS OF HORMONES AND NEUROPEPTIDES

Dopamine and Dopamine Agonists  

 

Dopamine and dopamine agonists inhibit TSH release by mechanisms discussed earlier.  Dopamine infusion can overcome the effects of thyroid hormone deficiency in the severely ill patient, suppressing the normally elevated TSH of the patient with primary hypothyroidism nearly into the normal range (235,363). Dopamine causes a reduction of the amplitude of TSH pulsatile release, but not in its frequency (328). However, chronic administration of dopamine agonists, for example in the treatment of prolactinomas, does not lead to central hypothyroidism despite the fact that there is marked decrease in the size of the pituitary tumor and inhibition of prolactin secretion.

 

Glucocorticoids  

 

The acute administration of pharmacological quantities of glucocorticoids will transiently suppress TSH (364-366). The mechanisms responsible for this effect may act both at the hypothalamic and pituitary level, as discussed above. Direct evidence of suppressed TRH synthesis was provided by an autopsy study showing reduced hypothalamic TRH mRNA expression in subjects treated with corticosteroids before death (367). TSH secretion recovers and T4 production rates are generally not impaired. In Cushing's syndrome, TSH may be normal or suppressed and, in general, there is a decrease in serum T3 concentrations relative to those of T4 (366). High levels of glucocorticoids inhibit basal TSH secretion slightly and may influence the circadian variation in serum TSH (222). Perhaps as a reflection of this, a modest serum TSH elevation may be present in patients with Addison's disease (368,369). TSH normalizes with glucocorticoid therapy alone if primary hypothyroidism is not also present. Similar to patients treated with long-acting somatostatin analogs, patients receiving long-term glucocorticoid therapy do not have a sustained reduction of serum TSH nor does hypothyroidism develop, because of the predominant effect of reduced thyroid hormone secretion in stimulating TSH secretion (370).

Gonadal Steroids

 

Aside from the well described effects of estrogen on the concentration of thyroxine-binding globulin (TBG), estrogen and testosterone have only minor influences on thyroid economy. In contrast with the mild inhibitory activity on alpha and beta TSH  subunits expression described in rats(216), in humans TSH release after TRH is enhanced by estradiol treatment perhaps because estrogens increase TRH receptor number (371,372). Treatment with the testosterone analog, fluoxymesterone, causes a significant decrease in the TSH response to TRH in hypogonadal men (373), possibly due to an increase in T4 to T3 conversion by androgen (374). This and the small estrogen effect may account for the lower TSH response to TRH in men than in women although there is no difference in basal TSH levels between the sexes. This is one of the few instances where there is not a close correlation between basal TSH levels and the response to TRH (see below).

 

Growth Hormone (GH) 

 

The possibility that central hypothyroidism could be induced by GH replacement in GH-deficient children was raised in early studies (375,376). However, these patients received human pituitary GH which in some cases was contaminated with TSH, perhaps inducing TSH antibodies. Nonetheless, in a cohort of children treated with recombinant hGH (rhGH) and affected with either idiopathic isolated GHD or MPHD, it was demonstrated that in the former the decrease in serum FT4 levels was not of clinical relevance, while in the latter a clear state of central hypothyroidism was seen in more than a half of the children (377). Concerning adults with GHD treated with rhGH, contradictory results have been reported. One study showed no significant changes in TSH concentrations during rhGH therapy of adults with GH deficiency (378). Later on, in two studies, thyroid function was evaluated in a large cohort of patients with adult or childhood onset of severe GHD. In 47% and 36% of euthyroid subjects, independently from rhGH dose, serum FT4 clearly fell into the hypothyroid range and some of these patients reported symptoms of hypothyroidism (375,376). Such results underline that, in adults as well as in children with organic GHD, rhGH therapy unmasks a state of central hypothyroidism, hidden by the condition of GHD itself.

 

In conclusion, GH does cause an increase in serum free T3, a decrease in free T4, and an increase in the T3 to T4 ratio in both T4-treated and T4 untreated patients. This suggests that the GH-induced increase in IGF-I stimulates T4 to T3 conversion. In keeping with this concept, IGF-I administration in healthy subjects is followed by a fall in serum TSH concentration (379).

 

Catecholamines  

 

Different from the rat, there is scanty evidence of an adrenergic control of TSH secretion in humans. Acute infusions of alpha or beta adrenergic blocking agents or agonists for short periods of time do not affect basal TSH (380,381), although a small stimulatory activity for endogenous adrenergic pathways is suggested by other studies (382,383). Furthermore, there is no effect of chronic propranolol administration on TSH secretion even though there may be modest inhibition of peripheral T4 to T3 conversion if amounts in excess of 160 mg/day are given (384). Evidence of a tonic inhibition of TSH secretion mediated by endogenous catecholamines has been obtained in women during the early follicular phase of the menstrual cycle (385).

 

The Response of TSH to TRH in Humans and the Role of Immunometric TSH Assays

 

More than 4 decades ago, application of ultrasensitive TSH measurements to the evaluation of patients with thyroid disease has undergone a revolutionary change.  This is due to the widespread application of the immunometric TSH assay. This assay uses monoclonal antibodies which bind one epitope of TSH and do not interfere with the binding of a second monoclonal or polyclonal antibody to a second epitope. The principle of the test is that TSH serves as the link between an immobilized antibody binding TSH at one epitope and a labelled (radioactive, chemiluminescent or other tag) monoclonal directed against a second portion of the molecule. This approach has improved both sensitivity and specificity by several orders of magnitude. Technical modifications have led to successive "generations" of TSH assays with progressively greater sensitivities (218,316). The first generation TSH assay was the standard radioimmunoassay which generally has lower detection limits of 1-2 mU/L. The "second" generation (first generation immunometric) assay improved the sensitivity to 0.1-0.2 mU/L and “third" generation assays further improved the sensitivity to approximately 0.005 mU/L.  From a technical point-of-view, the American Thyroid Association recommendations are that third generation assays should be able to quantitate TSH in the 0.010 to 0.020 mU/L range on an interassay basis with a coefficient of variation of 20% or less (386). As assay sensitivity has improved, the reference range has not changed, remaining between approximately 0.5 and 5.0 mU/L in most laboratories.  However, the TSH concentrations in the sera of patients with severe thyrotoxicosis secondary to Graves' disease have been lower with each successive improvement in the TSH assays: using a fourth-generation assay, the serum TSH is <0.004 mU/L in patients with severe hyperthyroidism (287,387).

 

The primary consequence of the availability of (ultra)sensitive TSH assays is to allow the substitution of a basal TSH measurement for the TRH test in patients suspected of thyrotoxicosis (388,285,389,286,287). Nonetheless, it is appropriate to review the results of TRH tests from the point-of-view of understanding thyroid pathophysiology, particularly in patients with hyperthyroidism or autonomous thyroid function. In healthy individuals, bolus i.v. injection of TRH is promptly followed by a rise of serum TSH concentration peaking after 20 to 30 minutes. The magnitude of the TSH peak is proportional to the logarithm of TRH doses between 6.25 up to ³400 ug, is significantly higher in women than in men, and declines with age (390,391). The individual TSH response to TRH is very variable and declines after repeated TRH administrations at short time intervals (391). In the presence of normal TSH bioactivity and adequate thyroid functional reserve, serum T3 and T4 also increase 120-180 minutes after TRH injection (391). There is a tight correlation between the basal TSH and the magnitude of the TRH-induced peak TSH (Fig. 12) Using a normal basal TSH range of 0.5 to 5 mU/L, the TRH response 15 to 20 minutes after 500 ug TRH (intravenously) ranges between 2 and 30 mU/L. The lower responses are found in patients with lower (but still normal) basal TSH levels (287). These results are quite consistent with older studies using radioimmunoassays (392). When the TSH response to TRH of all patients (hypo-, hyper- and euthyroid) is analyzed in terms of a "fold" response, the highest response (approximately 20-fold) occurs at a basal TSH of 0.5 mU/L and falls to less than 5 at either markedly subnormal or markedly elevated basal serum TSH concentrations (Fig. 16) (287). Thus, a low response can have two explanations.  The low response in patients with hyperthyroidism and a reduced basal TSH is due to refractoriness to TRH or depletion of pituitary TSH as a consequence of chronic thyroid hormone excess. In patients with primary hypothyroidism, the low fold-response reflects only the lack of sufficient pituitary TSH to achieve the necessary increment over the elevated basal TSH.

Figure 16. Relationship between basal and absolute (TRH stimulated-basal TSH) TRH-stimulated TSH response in 1061 ambulatory patients with an intact hypothalamic-pituitary (H-P) axis compared with that in untreated and T4-treated patients with central hypothyroidism. (From Spencer et al. (287) with permission)

 

Although, as stated before, the clinical relevance of the TRH test is presently limited, there are still some conditions in which the test may still be useful. These include subclinical primary hypothyroidism, central hypothyroidism (25), the syndromes of inappropriate TSH secretion (393) and non-thyroidal illnesses.

 

In patients with normal serum thyroid hormone concentrations and borderline TSH, an exaggerated TSH response to TRH not followed by an adequate increase in serum thyroid hormone levels may confirm the presence of subtle primary hypothyroidism (391).

 

An abnormal relationship between the basal TSH and the TRH-response is found in patients with central hypothyroidism. Here the fold TSH response to TRH is lower than normal (371,23,287). Again, however, TRH testing does not add substantially to the evaluation of such patients in that the diagnosis of central hypothyroidism is established by finding a normal or slightly elevated basal TSH in the presence of a significantly reduced free T4 concentration. While statistically (287) lower and sometimes delayed increments in TSH release after TRH infusion are found in patients with pituitary as opposed to hypothalamic hypothyroidism, the overlap in the TSH increments found in patients with these two conditions is sufficiently large (371,23,24,394), so that other diagnostic technologies, such as MRI, must be used to provide definitive localization of the lesion in patients with central hypothyroidism. It should be recalled that the TRH test may be useful in the diagnosis and follow-up of several pituitary disorders, but the discussion of this point is beyond the purpose of this chapter.

 

The TRH test still provides fundamental information in the differential diagnosis of hyperthyroidism due to TSH-secreting adenomas from syndromes with non-neoplastic TSH hypersecretion due to pituitary selective or generalized thyroid hormone resistance. In all the above conditions, increased or “inappropriately normal” serum TSH concentrations are observed in the presence of elevated circulating thyroid hormone levels. However, in most (>90%) of TSH-secreting adenomas serum TSH does not increase after TRH, while TRH responsiveness is observed in >95% of patients with nontumoral inappropriate TSH secretion (283,213,391).

 

Perhaps of most interest pathophysiologically is the response to TRH in patients with non-thyroidal illness and either normal or low free T4 indices (Fig. 12). Results from these patients fit within the normal distribution in terms of the relationship between basal TSH (whether suppressed or elevated) and the fold-response to TRH.  Thus the information provided by a TRH infusion test adds little to that obtained from an accurate basal TSH measurement (395). With respect to the evaluation of sick patients, while basal TSH values are on average higher than in patients with thyrotoxicosis, there is still some overlap between these groups (396,337,287,397). This indicates that even with second or third generation TSH assays, it may not be possible to establish that thyrotoxicosis is present based on a serum TSH measurement in a population which includes severely ill patients.

 

CLINICAL APPLICATION OF TSH MEASUREMENTS AND SUMMARY

 

Table 5 lists conditions in which basal TSH values may be altered as practical examples of the pathophysiology of the hypothalamic-pituitary thyroid axis.

 

Table 5. Conditions which May be Associated with Abnormal Serum TSH Concentrations

 

Expected TSH (mU/L)

Thyroid

Status

FT4

TSH reduced

 

 

 

1. Hyperthyroidism

<0.1

­

­, T3

2. “Euthyroid” Graves’ disease

0.2-0.5

N (­)

N(T3­)

3. Autonomous nodules

0.2-0.5

N (­)

N(T3­)

4. Excess thyroid hormone treatment

0.1-0.5

N,­

N,­

5.   Other forms of subclinical hyperthyroidism (including thyroiditis variants)

0.1-0.5

N,­

N,­

6. Illness with or without dopamine

0.1-5.0

N

­, N,¯

7. First trimester pregnancy

0.2-0.5

N (­)

N (­)

8. Hyperemesis gravidarum

0.2-0.5

N (­)

­(N)

9. Hydatidiform mole

0.1-0.4

­

­

10. Acute psychosis or depression (rare)

0.4-10

N

N (­)

11. Elderly (small fraction)

0.2-0.5

N

N

12. Cushing’s syndrome and glucocorticoids excess (inconsistent)

0.1-0.5

N

N

13. Retinoid X receptor-selective ligands

0.01-0.2

¯

¯

14. Various forms of central hypothyroidism

<0.1-0.4

¯

¯

15.  15. Congenital TSH deficiency

    a) Pit-1 mutations

    b) PROP1 mutations

    c) Mutations of TSHB gene in CAGYC region

    d) Skipping of TSHB gene exon 2

    e) Inactivating mutation of TRH receptor gene

 

0

0

0

 

0

1-2   ¯  ¯

 

 

 

¯

¯

¯

 

¯

 

¯

¯

¯

 

¯

 

TSH Elevated

 

 

 

1. Primary hypothyroidism

6-500

¯

¯

2. Resistance to TSH

 

6->100

 

N,¯

 

N,¯

3. Recovery from severe illness

5-30

N

N,¯

4. Iodine deficiency

6-150

N,¯

¯

5. Thyroid hormone resistance

1-15

N (¯,­)

­

6. Thyrotroph tumor

3-30

­

­

7. Central (“tertiary”) hypothyroidism

1-19

¯

¯

8. Psychiatric illness (especially bipolar disorders)

0.4-10

N

N

9. Test artifacts (endogenous anti-mouse gamma-globulin antibodies as well as “macroTSH”)

10-500

N

N

10.  10. Addison’s disease

 

5-30

 

N

 

N

 

 

Clinical Situations Associated with Subnormal TSH Values

 

The most common cause of a reduced TSH in a non-hospitalized patient is thyroid hormone excess. This may be due to endogenous hyperthyroidism or excess exogenous thyroid hormone. The degree of suppression of basal TSH is in proportion to the degree and duration of the thyroid hormone excess. The reduced TSH is the pathophysiological manifestation of the activation of the negative feedback loop.

 

While a low TSH in the presence of elevated thyroid hormones is logical, it results from multiple causes. Prolonged excessive thyroid hormone levels cause physiological "atrophy" of the thyroid stimulatory limb of the hypothalamic-pituitary thyroid axis. Thus, TRH synthesis is reduced, TRH mRNA in the PVN is absent, TRH receptors in the thyrotroph may be reduced, and the concentration of TSH beta and alpha subunits and both mRNAs in the thyrotroph are virtually undetectable. Therefore, it is not surprising that several months are usually required for the re-establishment of TSH secretion after the relief of thyrotoxicosis. This is especially observed in patients with Graves' disease after surgery or radioactive iodine, in whom TSH remains suppressed despite a rapid return to a euthyroid or even hypothyroid functional status (398,399).  Since TRH infusion will not increase TSH release in this situation, it is clear that the thyrotroph is transiently dysfunctional (400). A similar phenomenon occurs after excess thyroid hormone treatment is terminated, and after the transient hyperthyroidism associated with subacute or some variants of autoimmune thyroiditis, though the period of suppression is shorter under the latter circumstances (401). This cause of reduced circulating thyroid hormones and reduced or normal TSH should be distinguishable from central hypothyroidism by the history.

 

Severe illness is a common cause of TSH suppression although it is not often confused with thyrotoxicosis. Quantitation of thyroid hormones will generally resolve the issue (327). Patients receiving high-dose glucocorticoids acutely may also have suppressed TSH values although chronic glucocorticoid therapy does not cause sufficient TSH suppression to produce hypothalamic-pituitary hypothyroidism (see above).

 

Exogenous dopamine suppresses TSH release. Infusion of 5-7.5 mg/kg/min to normal volunteers causes an approximately 50% reduction in the concentrations of TSH and consequent small decreases in serum T4 and T3 concentrations (363). In critically ill patients, this effect of dopamine can be superimposed on the suppressive effects of acute illness on thyroid function, reducing T4 production to even lower levels (357). Dopamine is sufficiently potent to suppress TSH to normal levels in sick patients with primary hypothyroidism (363). This needs to be kept in mind when evaluating severely ill patients for this condition. Dopamine antagonists such as metoclopramide or domperidone cause a small increase in TSH in humans. However, somewhat surprisingly, patients receiving the dopamine agonist bromergocryptine do not become hypothyroid. Although L-dopa causes a statistically significant reduction in the TSH response to TRH, patients receiving this drug also remain euthyroid (370).

 

Studies in animals have suggested that pharmacological amounts of retinoids may decrease serum TSH concentration (see also paragraph “Effect of Thyroid Hormone on TSH Secretion”) (402,96). Severe central hypothyroidism associated with very low serum TSH concentration has been reported in patients with cutaneous T-cell lymphoma treated with high-dose bexarotene, a retinoid X receptor-selective ligand able to suppress TSH secretion (403).

 

hCG may function as a thyroid stimulator. During pregnancy, hCG stimulates the thyroid gland of the mother resulting in the typical transient decrease of the TSH levels during the first trimester (0.2 - 0.4 mU/L). Pathologic hCG secretion can result in frank, often mild, hyperthyroidism in patients with choriocarcinomas or molar pregnancies (404).

 

Patients with acute psychosis or depression and those with agitated psychoses may have high thyroid hormone levels and suppressed or elevated TSH values.  The etiology of the alterations in TSH are not known. Those receiving lithium for bipolar illness may also have elevated TSH values due to impairment of thyroid hormone release. Patients with underlying autoimmune thyroid disease or multi-nodular goiter are especially susceptible (405). A small fraction of elderly patients, particularly males, have subnormal TSH levels with normal serum thyroid hormone concentrations. It is likely that this reflects mild thyrotoxicosis if it is found to be reduced on repeated determinations.

 

Congenital central hypothyroidism with low serum TSH may result from mutations affecting TSHB gene or the Pit-1 gene (see paragraphs “The Thyroid-Stimulating Hormone Molecule”,  “Role of Pit-1 and its splicing variants in the regulation of TSHB gene expression” and “Other Transcription Factors Involved in TSHB Gene Expression”.

 

 

Causes of an Elevated TSH

 

Primary hypothyroidism is the most common cause of an elevated serum TSH. The serum free T4 is low normal or reduced in such patients but the serum free T3 values remain normal until the level of thyroid function has markedly deteriorated (118). Another common cause of an elevated TSH in an iodine-sufficient environment is the transient elevation which occurs during the recovery phase after severe illness (342,343). In such patients a "reawakening" of the hypothalamic-pituitary-thyroid axis occurs pari passu with the improvement in their clinical state. In general, such patients do not have underlying thyroid dysfunction. Iodine deficiency is not a cause of elevated TSH in Central and North America but may be in certain areas of Western Europe, South America, Africa and Asia.

 

The remainder of the conditions associated with an elevated TSH are extremely rare. Inherited (autosomal recessive) forms of partial (euthyroid hyperthyrotropinemia) or complete (congenital hypothyroidism) TSH resistance have been described associated with inactivating biallelic point mutations of the TSH receptor gene (406,407). Interestingly, inherited dominant forms of partial TSH resistance have also been described in the absence of TSH receptor gene mutations (408,409). The underlying molecular defect(s) remain(s) to be elucidated in such cases. More frequently, in a patient who has an elevated serum FT4, the presence of TSH at normal or increased levels should lead to a search for either resistance to thyroid hormone or a thyrotroph tumor. Hypothalamic-pituitary dysfunction may be associated with normal or even modest increases in TSH and are explained by the lack of normal TSH glycosylation in the TRH-deficient patient. The diagnosis is generally made by finding a serum free T4 index which is reduced to a greater extent than expected from the coincident serum TSH. Psychiatric illness may be associated with either elevated or suppressed TSH levels, but the abnormal values are not usually in the range normally associated with symptomatic thyroid dysfunction. The effect of glucocorticoids to suppress TSH secretion has already been mentioned. This is of relevance in patients with Addison's disease in whom TSH may be slightly elevated in the absence of primary thyroid disease.

 

Lastly, while most of the artifacts have been eliminated from the immunometric TSH assays, there remains the theoretical possibility of an elevated value due to the presence of endogenous anti-mouse gamma globulin antibodies (410,411). These heterophilic antibodies, like TSH, can complex the two TSH antibodies resulting in artificially elevated serum TSH assay results in euthyroid patients. Such artifacts can usually be identified by finding non-linear results upon assay of serial dilutions of the suspect serum with that from patients with a suppressed TSH. Moreover, the possible presence of “macro TSH” should be investigated in patients with high levels of TSH, normal circulating free thyroid hormones and absence of clinical signs and symptoms of hypothyroidism (410,411). Macro TSH is a large molecular-sized TSH that is mostly a complex of TSH and IgG. Precipitation of the serum with PEG and measurement of TSH in the supernatant is mandatory to confirm the presence of macro TSH, a procedure that is similar to that documenting the presence of macro PRL (412-415).

 

REFERENCES

 

  1. Shupnik MA, Ridgway EC, Chin WW. Molecular biology of thyrotropin. Endocr Rev 1989;10:459-475
  2. Grossmann M, Weintraub BD, Szkudlinski MW. Novel insights into the molecular mechanisms of human thyrotropin action: structural, physiological, and therapeutic implications for the glycoprotein hormone family. Endocr Rev 1997;18:476-501
  3. Ridgway EC, Weintraub BD, Maloof F. Metabolic clearance and production rates of human thyrotropin. J Clin Invest 1974;53:895-903
  4. Kourides IA, Re RN, Weintraub BD, Ridgway EC, Maloof F. Metabolic clearance and secretion rates of subunits of human thyrotropin. J Clin Invest 1977;59:508-516
  5. Naylor SL, Chin WW, Goodman HM, Lalley PA, Grzeschik KH, Sakaguchi AY. Chromosome assignment of genes encoding the alpha and beta subunits of glycoprotein hormones in man and mouse. Somatic Cell Genet 1983;9:757-770
  6. Burnside J, Buckland PR, Chin WW. Isolation and characterization of the gene encoding the alpha-subunit of the rat pituitary glycoprotein hormones. Gene 1988;70:67-74
  7. Gordon DF, Wood WM, Ridgway EC. Organization and nucleotide sequence of the gene encoding the beta-subunit of murine thyrotropin. DNA 1988;7:17-26
  8. Carr FE, Need LR, Chin WW. Isolation and characterization of the rat thyrotropin beta-subunit gene. Differential regulation of two transcriptional start sites by thyroid hormone. J Biol Chem 1987;262:981-987
  9. Tatsumi K, Hayashizaki Y, Hiraoka Y, Miyai K, Matsubara K. The structure of the human thyrotropin beta-subunit gene. Gene 1988;73:489-497
  10. Wondisford FE, Radovick S, Moates JM, Usala SJ, Weintraub BD. Isolation and characterization of the human thyrotropin beta-subunit gene. Differences in gene structure and promoter function from murine species. J Biol Chem 1988;263:12538-12542
  11. McDermott MT, Haugen BR, Black JN, Wood WM, Gordon DF, Ridgway EC. Congenital isolated central hypothyroidism caused by a "hot spot" mutation in the thyrotropin-beta gene. Thyroid 2002;12:1141-1146.
  12. Baquedano MS, Ciaccio M, Dujovne N, Herzovich V, Longueira Y, Warman DM, Rivarola MA, Belgorosky A. Two novel mutations of the TSH-beta subunit gene underlying congenital central hypothyroidism undetectable in neonatal TSH screening. J Clin Endocrinol Metab 2010;95:E98-103
  13. Magner JA. Thyroid-stimulating hormone: biosynthesis, cell biology, and bioactivity. Endocr Rev 1990;11:354-385
  14. Magner JA. Biosynthesis, cell biology, and bioactivity of thyroid-stimulating hormone: update 1994. In: Braverman L, Refetoff S, eds. Endocrine Reviews Monograph. Vol 3. Rockville: The Endocrine Society; 1994:55.
  15. Weintraub BD, Stannard BS, Magner JA, Ronin C, Taylor T, Joshi L, Constant RB, Menezes-Ferreira MM, Petrick P, Gesundheit N. Glycosylation and posttranslational processing of thyroid-stimulating hormone: clinical implications. Recent Prog Horm Res 1985;41:577-606
  16. Weintraub BD, Stannard BS, Linnekin D, Marshall M. Relationship of glycosylation to de novo thyroid-stimulating hormone biosynthesis and secretion by mouse pituitary tumor cells. J Biol Chem 1980;255:5715-5723
  17. Lapthorn AJ, Harris DC, Littlejohn A, Lustbader JW, Canfield RE, Machin KJ, Morgan FJ, Isaacs NW. Crystal structure of human chorionic gonadotropin. Nature 1994;369:455-461
  18. Kleinau G, Krause G. Thyrotropin and homologous glycoprotein hormone receptors: structural and functional aspects of extracellular signaling mechanisms. Endocr Rev 2009;30:133-151
  19. Amir SM, Kubota K, Tramontano D, Ingbar SH, Keutmann HT. The carbohydrate moiety of bovine thyrotropin is essential for full bioactivity but not for receptor recognition. Endocrinology 1987;120:345-352
  20. Menezes-Ferreira MM, Petrick PA, Weintraub BD. Regulation of thyrotropin (TSH) bioactivity by TSH-releasing hormone and thyroid hormone. Endocrinology 1986;118:2125-2130
  21. Gesundheit N, Fink DL, Silverman LA, Weintraub BD. Effect of thyrotropin-releasing hormone on the carbohydrate structure of secreted mouse thyrotropin. Analysis by lectin affinity chromatography. J Biol Chem 1987;262:5197-5203
  22. Taylor T, Weintraub BD. Altered thyrotropin (TSH) carbohydrate structures in hypothalamic hypothyroidism created by paraventricular nuclear lesions are corrected by in vivo TSH-releasing hormone administration. Endocrinology 1989;125:2198-2203
  23. Petersen VB, McGregor AM, Belchetz PE, Elkeles RS, Hall R. The secretion of thyrotrophin with impaired biological activity in patients with hypothalamic-pituitary disease. Clin Endocrinol (Oxf) 1978;8:397-402
  24. Faglia G, Bitensky L, Pinchera A, Ferrari C, Paracchi A, Beck-Peccoz P, Ambrosi B, Spada A. Thyrotropin secretion in patients with central hypothyroidism: evidence for reduced biological activity of immunoreactive thyrotropin. J Clin Endocrinol Metab 1979;48:989-998
  25. Beck-Peccoz P, Rodari G, Giavoli C, Lania A. Central hypothyroidism - a neglected thyroid disorder. Nat Rev Endocrinol 2017;13:588-598
  26. Beck-Peccoz P, Amr S, Menezes-Ferreira MM, Faglia G, Weintraub BD. Decreased receptor binding of biologically inactive thyrotropin in central hypothyroidism. Effect of treatment with thyrotropin-releasing hormone. N Engl J Med 1985;312:1085-1090
  27. Beck-Peccoz P, Persani L. Variable biological activity of thyroid-stimulating hormone. Eur J Endocrinol 1994;131:331-340
  28. Persani L, Terzolo M, Asteria C, Orlandi F, Angeli A, Beck-Peccoz P. Circadian variations of thyrotropin bioactivity in normal subjects and patients with primary hypothyroidism. J Clin Endocrinol Metab 1995;80:2722-2728
  29. Persani L. Hypothalamic thyrotropin-releasing hormone and thyrotropin biological activity. Thyroid 1998;8:941-946
  30. Zabczynska M, Kozlowska K, Pochec E. Glycosylation in the Thyroid Gland: Vital Aspects of Glycoprotein Function in Thyrocyte Physiology and Thyroid Disorders. Int J Mol Sci 2018;19
  31. Hayashizaki Y, Hiraoka Y, Endo Y, Miyai K, Matsubara K. Thyroid-stimulating hormone (TSH) deficiency caused by a single base substitution in the CAGYC region of the beta-subunit. Embo J 1989;8:2291-2296
  32. Hayashizaki Y, Hiraoka Y, Tatsumi K, Hashimoto T, Furuyama J, Miyai K, Nishijo K, Matsuura M, Kohno H, Labbe A, et al. Deoxyribonucleic acid analyses of five families with familial inherited thyroid stimulating hormone deficiency. J Clin Endocrinol Metab 1990;71:792-796
  33. Dacou-Voutetakis C, Feltquate DM, Drakopoulou M, Kourides IA, Dracopoli NC. Familial hypothyroidism caused by a nonsense mutation in the thyroid-stimulating hormone beta-subunit gene. Am J Hum Genet 1990;46:988-993
  34. Medeiros-Neto G, Herodotou DT, Rajan S, Kommareddi S, de Lacerda L, Sandrini R, Boguszewski MC, Hollenberg AN, Radovick S, Wondisford FE. A circulating, biologically inactive thyrotropin caused by a mutation in the beta subunit gene. J Clin Invest 1996;97:1250-1256
  35. Doeker BM, Pfaffle RW, Pohlenz J, Andler W. Congenital central hypothyroidism due to a homozygous mutation in the thyrotropin beta-subunit gene follows an autosomal recessive inheritance. J Clin Endocrinol Metab 1998;83:1762-1765.
  36. Heinrichs C, Parma J, Scherberg NH, Delange F, Van Vliet G, Duprez L, Bourdoux P, Bergmann P, Vassart G, Refetoff S. Congenital central isolated hypothyroidism caused by a homozygous mutation in the TSH-beta subunit gene. Thyroid 2000;10:387-391.
  37. Bonomi M, Proverbio MC, Weber G, Chiumello G, Beck-Peccoz P, Persani L. Hyperplastic pituitary gland, high serum glycoprotein hormone alpha- subunit, and variable circulating thyrotropin (TSH) levels as hallmark of central hypothyroidism due to mutations of the TSH beta gene. J Clin Endocrinol Metab 2001;86:1600-1604.
  38. Vallette-Kasic S, Barlier A, Teinturier C, Diaz A, Manavela M, Berthezene F, Bouchard P, Chaussain JL, Brauner R, Pellegrini-Bouiller I, Jaquet P, Enjalbert A, Brue T. PROP1 gene screening in patients with multiple pituitary hormone deficiency reveals two sites of hypermutability and a high incidence of corticotroph deficiency. J Clin Endocrinol Metab 2001;86:4529-4535.
  39. Pohlenz J, Dumitrescu A, Aumann U, Koch G, Melchior R, Prawitt D, Refetoff S. Congenital secondary hypothyroidism caused by exon skipping due to a homozygous donor splice site mutation in the TSHbeta-subunit gene. J Clin Endocrinol Metab 2002;87:336-339.
  40. Deladoey J, Vuissoz JM, Domene HM, Malik N, Gruneiro-Papendieck L, Chiesa A, Heinrich JJ, Mullis PE. Congenital secondary hypothyroidism due to a mutation C105Vfs114X thyrotropin-beta mutation: genetic study of five unrelated families from Switzerland and Argentina. Thyroid 2003;13:553-559.
  41. Kleinau G, Kalveram L, Kohrle J, Szkudlinski M, Schomburg L, Biebermann H, Gruters-Kieslich A. Minireview: Insights Into the Structural and Molecular Consequences of the TSH-beta Mutation C105Vfs114X. Mol Endocrinol 2016;30:954-964
  42. Kalveram L, Kleinau G, Szymanska K, Scheerer P, Rivero-Muller A, Gruters-Kieslich A, Biebermann H. The Pathogenic TSH beta-subunit Variant C105Vfs114X Causes a Modified Signaling Profile at TSHR. Int J Mol Sci 2019;20
  43. Azzam N, Bar-Shalom R, Kraiem Z, Fares F. Human thyrotropin (TSH) variants designed by site-directed mutagenesis block TSH activity in vitro and in vivo. Endocrinology 2005;146:2845-2850
  44. Mueller S, Kleinau G, Szkudlinski MW, Jaeschke H, Krause G, Paschke R. The superagonistic activity of bovine thyroid-stimulating hormone (TSH) and the human TR1401 TSH analog is determined by specific amino acids in the hinge region of the human TSH receptor. J Biol Chem 2009;284:16317-16324
  45. Neumann S, Huang W, Titus S, Krause G, Kleinau G, Alberobello AT, Zheng W, Southall NT, Inglese J, Austin CP, Celi FS, Gavrilova O, Thomas CJ, Raaka BM, Gershengorn MC. Small-molecule agonists for the thyrotropin receptor stimulate thyroid function in human thyrocytes and mice. Proc Natl Acad Sci U S A 2009;106:12471-12476
  46. Neumann S, Eliseeva E, McCoy JG, Napolitano G, Giuliani C, Monaco F, Huang W, Gershengorn MC. A new small-molecule antagonist inhibits Graves' disease antibody activation of the TSH receptor. J Clin Endocrinol Metab 2011;96:548-554
  47. Beck-Peccoz P. Antithyroid drugs are 65 years old: time for retirement? Endocrinology 2008;149:5943-5944
  48. Gershengorn MC, Neumann S. Update in TSH receptor agonists and antagonists. J Clin Endocrinol Metab 2012;97:4287-4292
  49. Nakabayashi K, Matsumi H, Bhalla A, Bae J, Mosselman S, Hsu SY, Hsueh AJ. Thyrostimulin, a heterodimer of two new human glycoprotein hormone subunits, activates the thyroid-stimulating hormone receptor. J Clin Invest 2002;109:1445-1452.
  50. Karponis D, Ananth S. The role of thyrostimulin and its potential clinical significance. Endocr Regul 2017;51:117-128
  51. Hausken KN, Tizon B, Shpilman M, Barton S, Decatur W, Plachetzki D, Kavanaugh S, Ul-Hasan S, Levavi-Sivan B, Sower SA. Cloning and characterization of a second lamprey pituitary glycoprotein hormone, thyrostimulin (GpA2/GpB5). Gen Comp Endocrinol 2018;264:16-27
  52. Wang P, Liu S, Yang Q, Liu Z, Zhang S. Functional Characterization of Thyrostimulin in Amphioxus Suggests an Ancestral Origin of the TH Signaling Pathway. Endocrinology 2018;159:3536-3548
  53. Hausken K, Levavi-Sivan B. Synteny and phylogenetic analysis of paralogous thyrostimulin beta subunits (GpB5) in vertebrates. PLoS One 2019;14:e0222808
  54. Nagasaki H, Wang Z, Jackson VR, Lin S, Nothacker HP, Civelli O. Differential expression of the thyrostimulin subunits, glycoprotein alpha2 and beta5 in the rat pituitary. J Mol Endocrinol 2006;37:39-50
  55. Wondisford FE. The thyroid axis just got more complicated. J Clin Invest 2002;109:1401-1402.
  56. Sun SC, Hsu PJ, Wu FJ, Li SH, Lu CH, Luo CW. Thyrostimulin, but not thyroid-stimulating hormone (TSH), acts as a paracrine regulator to activate the TSH receptor in mammalian ovary. J Biol Chem 2010;285:3758-3765
  57. Bassett JH, van der Spek A, Logan JG, Gogakos A, Bagchi-Chakraborty J, Williams AJ, Murphy E, van Zeijl C, Down J, Croucher PI, Boyde A, Boelen A, Williams GR. Thyrostimulin Regulates Osteoblastic Bone Formation During Early Skeletal Development. Endocrinology 2015;156:3098-3113
  58. Reichlin S, Utiger RD. Regulation of the pituitary-thyroid axis in man: relationship of TSH concentration to concentration of free and total thyroxine in plasma. J Clin Endocrinol Metab 1967;27:251-255
  59. Bogazzi F, Bartalena L, Brogioni S, Burelli A, Grasso L, Dell'Unto E, Manetti L, Martino E. L-thyroxine directly affects expression of thyroid hormone-sensitive genes: regulatory effect of RXRbeta. Mol Cell Endocrinol 1997;134:23-31
  60. Gurr JA, Kourides IA. Regulation of thyrotropin biosynthesis. Discordant effect of thyroid hormone on alpha and beta subunit mRNA levels. J Biol Chem 1983;258:10208-10211
  61. Shupnik MA, Chin WW, Habener JF, Ridgway EC. Transcriptional regulation of the thyrotropin subunit genes by thyroid hormone. J Biol Chem 1985;260:2900-2903
  62. Ridgway EC, Kourides IA, Chin WW, Cooper DS, Maloof F. Augmentation of pituitary thyrotrophin response to thyrotrophin releasing hormone during subphysiological tri-iodothyroinine therapy in hypothyroidism. Clin Endocrinol (Oxf) 1979;10:343-353
  63. Kourides IA, Gurr JA, Wolf O. The regulation and organization of thyroid stimulating hormone genes. Recent Prog Horm Res 1984;40:79-120
  64. Carr FE, Ridgway EC, Chin WW. Rapid simultaneous measurement of rat alpha- and thyrotropin (TSH) beta-subunit messenger ribonucleic acids (mRNAs) by solution hybridization: regulation of TSH subunit mRNAs by thyroid hormones. Endocrinology 1985;117:1272-1278
  65. Shupnik MA, Ridgway EC. Thyroid hormone control of thyrotropin gene expression in rat anterior pituitary cells. Endocrinology 1987;121:619-624
  66. Carr FE, Shupnik MA, Burnside J, Chin WW. Thyrotropin-releasing hormone stimulates the activity of the rat thyrotropin beta-subunit gene promoter transfected into pituitary cells. Mol Endocrinol 1989;3:717-724
  67. Chatterjee VK, Lee JK, Rentoumis A, Jameson JL. Negative regulation of the thyroid-stimulating hormone alpha gene by thyroid hormone: receptor interaction adjacent to the TATA box. Proc Natl Acad Sci U S A 1989;86:9114-9118
  68. Gurr JA, Januszeski MM, Tidikis IM, Norcross JJ, Kourides IA. Thyroid hormone regulates expression of the thyrotropin beta-subunit gene from both transcription start sites in the mouse and rat. Mol Cell Endocrinol 1990;71:185-193
  69. Chin WW, Carr FE, Burnside J, Darling DS. Thyroid hormone regulation of thyrotropin gene expression. Recent Prog Horm Res 1993;48:393-414
  70. Darling DS, Burnside J, Chin WW. Binding of thyroid hormone receptors to the rat thyrotropin-beta gene. Mol Endocrinol 1989;3:1359-1368
  71. Wondisford FE, Farr EA, Radovick S, Steinfelder HJ, Moates JM, McClaskey JH, Weintraub BD. Thyroid hormone inhibition of human thyrotropin beta-subunit gene expression is mediated by a cis-acting element located in the first exon. J Biol Chem 1989;264:14601-14604
  72. Bodenner DL, Mroczynski MA, Weintraub BD, Radovick S, Wondisford FE. A detailed functional and structural analysis of a major thyroid hormone inhibitory element in the human thyrotropin beta-subunit gene. J Biol Chem 1991;266:21666-21673
  73. Brent GA, Williams GR, Harney JW, Forman BM, Samuels HH, Moore DD, Larsen PR. Effects of varying the position of thyroid hormone response elements within the rat growth hormone promoter: implications for positive and negative regulation by 3,5,3'-triiodothyronine. Mol Endocrinol 1991;5:542-548
  74. Burnside J, Darling DS, Carr FE, Chin WW. Thyroid hormone regulation of the rat glycoprotein hormone alpha-subunit gene promoter activity. J Biol Chem 1989;264:6886-6891
  75. Carr FE, Burnside J, Chin WW. Thyroid hormones regulate rat thyrotropin beta gene promoter activity expressed in GH3 cells. Mol Endocrinol 1989;3:709-716
  76. Carr FE, Wong NC. Characteristics of a negative thyroid hormone response element. J Biol Chem 1994;269:4175-4179
  77. Shupnik MA, Greenspan SL, Ridgway EC. Transcriptional regulation of thyrotropin subunit genes by thyrotropin-releasing hormone and dopamine in pituitary cell culture. J Biol Chem 1986;261:12675-12679
  78. Langlois MF, Zanger K, Monden T, Safer JD, Hollenberg AN, Wondisford FE. A unique role of the beta-2 thyroid hormone receptor isoform in negative regulation by thyroid hormone. Mapping of a novel amino-terminal domain important for ligand-independent activation. J Biol Chem 1997;272:24927-24933
  79. Wikstrom L, Johansson C, Salto C, Barlow C, Campos Barros A, Baas F, Forrest D, Thoren P, Vennstrom B. Abnormal heart rate and body temperature in mice lacking thyroid hormone receptor alpha 1. Embo J 1998;17:455-461.
  80. Forrest D, Hanebuth E, Smeyne RJ, Everds N, Stewart CL, Wehner JM, Curran T. Recessive resistance to thyroid hormone in mice lacking thyroid hormone receptor beta: evidence for tissue-specific modulation of receptor function. Embo J 1996;15:3006-3015.
  81. Weiss RE, Forrest D, Pohlenz J, Cua K, Curran T, Refetoff S. Thyrotropin regulation by thyroid hormone in thyroid hormone receptor beta-deficient mice. Endocrinology 1997;138:3624-3629.
  82. Abel ED, Boers ME, Pazos-Moura C, Moura E, Kaulbach H, Zakaria M, Lowell B, Radovick S, Liberman MC, Wondisford F. Divergent roles for thyroid hormone receptor beta isoforms in the endocrine axis and auditory system. J Clin Invest 1999;104:291-300.
  83. Hayashi Y, Xie J, Weiss RE, Pohlenz J, Refetoff S. Selective pituitary resistance to thyroid hormone produced by expression of a mutant thyroid hormone receptor beta gene in the pituitary gland of transgenic mice. Biochem Biophys Res Commun 1998;245:204-210
  84. Gauthier K, Chassande O, Plateroti M, Roux JP, Legrand C, Pain B, Rousset B, Weiss R, Trouillas J, Samarut J. Different functions for the thyroid hormone receptors TRalpha and TRbeta in the control of thyroid hormone production and post-natal development. Embo J 1999;18:623-631.
  85. Gothe S, Wang Z, Ng L, Kindblom JM, Barros AC, Ohlsson C, Vennstrom B, Forrest D. Mice devoid of all known thyroid hormone receptors are viable but exhibit disorders of the pituitary-thyroid axis, growth, and bone maturation. Genes Dev 1999;13:1329-1341.
  86. Weiss RE, Chassande O, Koo EK, Macchia PE, Cua K, Samarut J, Refetoff S. Thyroid function and effect of aging in combined hetero/homozygous mice deficient in thyroid hormone receptors alpha and beta genes. J Endocrinol 2002;172:177-185.
  87. Abel ED, Moura EG, Ahima RS, Campos-Barros A, Pazos-Moura CC, Boers ME, Kaulbach HC, Forrest D, Wondisford FE. Dominant inhibition of thyroid hormone action selectively in the pituitary of thyroid hormone receptor-beta null mice abolishes the regulation of thyrotropin by thyroid hormone. Mol Endocrinol 2003;17:1767-1776
  88. Morita S, Fernandez-Mejia C, Melmed S. Retinoic acid selectively stimulates growth hormone secretion and messenger ribonucleic acid levels in rat pituitary cells. Endocrinology 1989;124:2052-2056
  89. Zenke M, Munoz A, Sap J, Vennstrom B, Beug H. v-erbA oncogene activation entails the loss of hormone-dependent regulator activity of c-erbA. Cell 1990;61:1035-1049
  90. Wondisford FE, Steinfelder HJ, Nations M, Radovick S. AP-1 antagonizes thyroid hormone receptor action on the thyrotropin beta-subunit gene. J Biol Chem 1993;268:2749-2754.
  91. Shibusawa N, Hollenberg AN, Wondisford FE. Thyroid hormone receptor DNA binding is required for both positive and negative gene regulation. J Biol Chem 2003;278:732-738
  92. Nakano K, Matsushita A, Sasaki S, Misawa H, Nishiyama K, Kashiwabara Y, Nakamura H. Thyroid-hormone-dependent negative regulation of thyrotropin beta gene by thyroid hormone receptors: study with a new experimental system using CV1 cells. Biochem J 2004;378:549-557
  93. Hallenbeck PL, Phyillaier M, Nikodem VM. Divergent effects of 9-cis-retinoic acid receptor on positive and negative thyroid hormone receptor-dependent gene expression. J Biol Chem 1993;268:3825-3828
  94. Cohen O, Flynn TR, Wondisford FE. Ligand-dependent antagonism by retinoid X receptors of inhibitory thyroid hormone response elements. J Biol Chem 1995;270:13899-13905
  95. Breen JJ, Hickok NJ, Gurr JA. The rat TSHbeta gene contains distinct response elements for regulation by retinoids and thyroid hormone. Mol Cell Endocrinol 1997;131:137-146.
  96. Haugen BR, Brown NS, Wood WM, Gordon DF, Ridgway EC. The thyrotrope-restricted isoform of the retinoid-X receptor-gamma1 mediates 9-cis-retinoic acid suppression of thyrotropin-beta promoter activity. Mol Endocrinol 1997;11:481-489
  97. Chiamolera MI, Sidhaye AR, Matsumoto S, He Q, Hashimoto K, Ortiga-Carvalho TM, Wondisford FE. Fundamentally distinct roles of thyroid hormone receptor isoforms in a thyrotroph cell line are due to differential DNA binding. Mol Endocrinol 2012;26:926-939
  98. Ramadoss P, Abraham BJ, Tsai L, Zhou Y, Costa-e-Sousa RH, Ye F, Bilban M, Zhao K, Hollenberg AN. Novel mechanism of positive versus negative regulation by thyroid hormone receptor beta1 (TRbeta1) identified by genome-wide profiling of binding sites in mouse liver. J Biol Chem 2014;289:1313-1328
  99. Cheng SY, Leonard JL, Davis PJ. Molecular aspects of thyroid hormone actions. Endocr Rev 2010;31:139-170
  100. Kim MK, McClaskey JH, Bodenner DL, Weintraub BD. A c-fos/c-jun-like factor and the pituitary-specific factor Pit-1 are both necessary to mediate induction of the human thyrotropin beta promoter. Trans Assoc Am Physicians 1993;106:62-68
  101. Pfahl M. Nuclear receptor/AP-1 interaction. Endocr Rev 1993;14:651-658
  102. Koenig RJ, Watson AY. Enrichment of rat anterior pituitary cell types by metrizamide density gradient centrifugation. Endocrinology 1984;115:317-323
  103. Farquhar R, Rinehart J. Cytological alterations in the anterior pituitary gland following thyroidectomy. Endocrinology 1954;55:857
  104. Larsen PR, Silva JE, Kaplan MM. Relationships between circulating and intracellular thyroid hormones: physiological and clinical implications. Endocr Rev 1981;2:87-102
  105. Franklyn JA, Wood DF, Balfour NJ, Ramsden DB, Docherty K, Chin WW, Sheppard MC. Effect of hypothyroidism and thyroid hormone replacement in vivo on pituitary cytoplasmic concentrations of thyrotropin-beta and alpha-subunit messenger ribonucleic acids. Endocrinology 1987;120:2279-2288
  106. Silva JE, Larsen PR. Pituitary nuclear 3,5,3'-triiodothyronine and thyrotropin secretion: an explanation for the effect of thyroxine. Science 1977;198:617-620.
  107. Silva JE, Larsen PR. Peripheral metabolism of homologous thyrotropin in euthyroid and hypothyroid rats: acute effects of thyrotropin-releasing hormone, triiodothyronine, and thyroxine. Endocrinology 1978;102:1783-1796
  108. Bowers CY, Lee KL, Schally AV. A study on the interaction of the thyrotropin-releasing factor and L-triiodothyronine: effects of puromycin and cycloheximide. Endocrinology 1968;82:75-82
  109. Vale W, Burgus R, Guillemin R. On the mechanism of action of TRF: effects of cycloheximide and actinomycin on the release of TSH stimulated in vitro by TRF and its inhibition by thyroxine. Neuroendocrinology 1968;3:34
  110. Gard TG, Bernstein B, Larsen PR. Studies on the mechanism of 3,5,3'-triiodothyronine-induced suppression of secretagogue-induced thyrotropin release in vitro. Endocrinology 1981;108:2046-2053
  111. Koenig RJ, Senator D, Larsen PR. Phorbol esters as probes of the regulation of thyrotropin secretion. Biochem Biophys Res Commun 1984;125:353-359
  112. Schrey MP, Larsen PR. Evidence for a possible role for Ca++ in the 3,5,3'-triiodothyronine inhibition of thyrotropin-releasing hormone-induced secretion of thyrotropin by rat anterior pituitary in vitro. Endocrinology 1981;108:1690-1696
  113. Obregon MJ, Pascual A, Mallol J, Morreale de Escobar G, Escobar del Rey F. Evidence against a major role of L-thyroxine at the pituitary level: studies in rats treated with iopanoic acid (telepaque). Endocrinology 1980;106:1827-1836
  114. Lueprasitsakul W, Alex S, Fang SL, Pino S, Irmscher K, Kohrle J, Braverman LE. Flavonoid administration immediately displaces thyroxine (T4) from serum transthyretin, increases serum free T4, and decreases serum thyrotropin in the rat. Endocrinology 1990;126:2890-2895
  115. Spencer CA, Takeuchi M, Kazarosyan M, MacKenzie F, Beckett GJ, Wilkinson E. Interlaboratory/intermethod differences in functional sensitivity of immunometric assays of thyrotropin (TSH) and impact on reliability of measurement of subnormal concentrations of TSH. Clin Chem 1995;41:367-374.
  116. Riesco G, Taurog A, Larsen R, Krulich L. Acute and chronic responses to iodine deficiency in rats. Endocrinology 1977;100:303-313
  117. Patel YC, Pharoah PO, Hornabrook RW, Hetzel BS. Serum triiodothyronine, thyroxine and thyroid-stimulating hormone in endemic goiter: a comparison of goitrous and nongoitrous subjects in New Guinea. J Clin Endocrinol Metab 1973;37:783-789
  118. Bigos ST, Ridgway EC, Kourides IA, Maloof F. Spectrum of pituitary alterations with mild and severe thyroid impairment. J Clin Endocrinol Metab 1978;46:317-325
  119. Wennlund A. Variation in serum levels of T3, T4, FT4 and TSH during thyroxine replacement therapy. Acta Endocrinol (Copenh) 1986;113:47-49
  120. Jackson IM. Thyrotropin-releasing hormone. N Engl J Med 1982;306:145-155
  121. Guillemin R. Hypothalamic hormones a.k.a. hypothalamic releasing factors. J Endocrinol 2005;184:11-28.
  122. Jeffcoate SL, White N, Hokfelt T, Fuxe K, Johansson O. Proceedings: Localization of thyrotrophin releasing hormone in the spinal cord of the rat by immunohisto-chemistry and radioimmunoassay. J Endocrinol 1976;69:9P-10P
  123. Martino E, Lernmark A, Seo H, Steiner DF, Refetoff S. High concentration of thyrotropin-releasing hormone in pancreatic islets. Proc Natl Acad Sci U S A 1978;75:4265-4267
  124. Gkonos PJ, Tavianini MA, Liu CC, Roos BA. Thyrotropin-releasing hormone gene expression in normal thyroid parafollicular cells. Mol Endocrinol 1989;3:2101-2109
  125. Bruhn TO, Rondeel JM, Bolduc TG, Jackson IM. Thyrotropin-releasing hormone (TRH) gene expression in the anterior pituitary. I. Presence of pro-TRH messenger ribonucleic acid and pro-TRH-derived peptide in a subpopulation of somatotrophs. Endocrinology 1994;134:815-820
  126. Bruhn TO, Rondeel JM, Jackson IM. Thyrotropin-releasing hormone gene expression in the anterior pituitary. IV. Evidence for paracrine and autocrine regulation. Endocrinology 1998;139:3416-3422
  127. Bilek R. TRH-like peptides in prostate gland and other tissues. Physiol Res 2000;49:S19-26.
  128. Fekete C, Lechan RM. Negative feedback regulation of hypophysiotropic thyrotropin-releasing hormone (TRH) synthesizing neurons: role of neuronal afferents and type 2 deiodinase. Front Neuroendocrinol 2007;28:97-114
  129. Bruhn TO, Huang SS, Vaslet C, Nillni EA. Glucocorticoids modulate the biosynthesis and processing of prothyrotropin releasing-hormone (proTRH). Endocrine 1998;9:143-152
  130. Lee SL, Stewart K, Goodman RH. Structure of the gene encoding rat thyrotropin releasing hormone. J Biol Chem 1988;263:16604-16609
  131. Yamada M, Radovick S, Wondisford FE, Nakayama Y, Weintraub BD, Wilber JF. Cloning and structure of human genomic DNA and hypothalamic cDNA encoding human prepro thyrotropin-releasing hormone. Mol Endocrinol 1990;4:551-556
  132. Yamada M, Wondisford FE, Radovick S, Nakayama Y, Weintraub BD, Wilber JF. Assignment of human preprothyrotropin-releasing hormone (TRH) gene to chromosome 3. Somat Cell Mol Genet 1991;17:97-100
  133. Bulant M, Roussel JP, Astier H, Nicolas P, Vaudry H. Processing of thyrotropin-releasing hormone prohormone (pro-TRH) generates a biologically active peptide, prepro-TRH-(160-169), which regulates TRH-induced thyrotropin secretion. Proc Natl Acad Sci U S A 1990;87:4439-4443
  134. Pekary AE. Is Ps4 (prepro-TRH [160-169]) more than an enhancer for thyrotropin-releasing hormone? Thyroid 1998;8:963-968
  135. Nillni EA. Neuroregulation of ProTRH biosynthesis and processing. Endocrine 1999;10:185-199
  136. Carr FE, Fein HG, Fisher CU, Wessendorf MW, Smallridge RC. A cryptic peptide (160-169) of thyrotropin-releasing hormone prohormone demonstrates biological activity in vivo and in vitro. Endocrinology 1992;131:2653-2658
  137. Carr FE, Reid AH, Wessendorf MW. A cryptic peptide from the preprothyrotropin-releasing hormone precursor stimulates thyrotropin gene expression. Endocrinology 1993;133:809-814
  138. Nillni EA, Sevarino KA. The biology of pro-thyrotropin-releasing hormone-derived peptides. Endocr Rev 1999;20:599-648
  139. Engler D, Redei E, Kola I. The corticotropin-release inhibitory factor hypothesis: a review of the evidence for the existence of inhibitory as well as stimulatory hypophysiotropic regulation of adrenocorticotropin secretion and biosynthesis. Endocr Rev 1999;20:460-500
  140. Jessop DS. Review: Central non-glucocorticoid inhibitors of the hypothalamo-pituitary-adrenal axis. J Endocrinol 1999;160:169-180
  141. Koller KJ, Wolff RS, Warden MK, Zoeller RT. Thyroid hormones regulate levels of thyrotropin-releasing-hormone mRNA in the paraventricular nucleus. Proc Natl Acad Sci U S A 1987;84:7329-7333
  142. Segerson TP, Kauer J, Wolfe HC, Mobtaker H, Wu P, Jackson IM, Lechan RM. Thyroid hormone regulates TRH biosynthesis in the paraventricular nucleus of the rat hypothalamus. Science 1987;238:78-80
  143. Dyess EM, Segerson TP, Liposits Z, Paull WK, Kaplan MM, Wu P, Jackson IM, Lechan RM. Triiodothyronine exerts direct cell-specific regulation of thyrotropin-releasing hormone gene expression in the hypothalamic paraventricular nucleus. Endocrinology 1988;123:2291-2297
  144. Kakucska I, Rand W, Lechan RM. Thyrotropin-releasing hormone gene expression in the hypothalamic paraventricular nucleus is dependent upon feedback regulation by both triiodothyronine and thyroxine. Endocrinology 1992;130:2845-2850
  145. Strait KA, Zou L, Oppenheimer JH. Beta 1 isoform-specific regulation of a triiodothyronine-induced gene during cerebellar development. Mol Endocrinol 1992;6:1874-1880
  146. Kakucska I, Romero LI, Clark BD, Rondeel JM, Qi Y, Alex S, Emerson CH, Lechan RM. Suppression of thyrotropin-releasing hormone gene expression by interleukin-1-beta in the rat: implications for nonthyroidal illness. Neuroendocrinology 1994;59:129-137
  147. Wilber JF, Xu AH. The thyrotropin-releasing hormone gene 1998: cloning, characterization, and transcriptional regulation in the central nervous system, heart, and testis. Thyroid 1998;8:897-901
  148. Abel ED, Ahima RS, Boers ME, Elmquist JK, Wondisford FE. Critical role for thyroid hormone receptor beta2 in the regulation of paraventricular thyrotropin-releasing hormone neurons. J Clin Invest 2001;107:1017-1023.
  149. Riskind PN, Kolodny JM, Larsen PR. The regional hypothalamic distribution of type II 5'-monodeiodinase in euthyroid and hypothyroid rats. Brain Res 1987;420:194-198
  150. Rodriguez EM, Blazquez JL, Pastor FE, Pelaez B, Pena P, Peruzzo B, Amat P. Hypothalamic tanycytes: a key component of brain-endocrine interaction. Int Rev Cytol 2005;247:89-164
  151. Jansen J, Friesema EC, Milici C, Visser TJ. Thyroid hormone transporters in health and disease. Thyroid 2005;15:757-768
  152. Felmlee MA, Jones RS, Rodriguez-Cruz V, Follman KE, Morris ME. Monocarboxylate Transporters (SLC16): Function, Regulation, and Role in Health and Disease. Pharmacol Rev 2020;72:466-485
  153. Stromme P, Groeneweg S, Lima de Souza EC, Zevenbergen C, Torgersbraten A, Holmgren A, Gurcan E, Meima ME, Peeters RP, Visser WE, Honeren Johansson L, Babovic A, Zetterberg H, Heuer H, Frengen E, Misceo D, Visser TJ. Mutated Thyroid Hormone Transporter OATP1C1 Associates with Severe Brain Hypometabolism and Juvenile Neurodegeneration. Thyroid 2018;28:1406-1415
  154. Yamada M, Satoh T, Monden T, Mori M. Assignment of the thyrotropin-releasing hormone gene (TRH) to human chromosome 3q13.3-->q21 by in situ hybridization. Cytogenet Cell Genet 1999;87:275
  155. Chiamolera MI, Wondisford FE. Minireview: Thyrotropin-releasing hormone and the thyroid hormone feedback mechanism. Endocrinology 2009;150:1091-1096
  156. Nikrodhanond AA, Ortiga-Carvalho TM, Shibusawa N, Hashimoto K, Liao XH, Refetoff S, Yamada M, Mori M, Wondisford FE. Dominant role of thyrotropin-releasing hormone in the hypothalamic-pituitary-thyroid axis. J Biol Chem 2006;281:5000-5007
  157. Mittag J, Friedrichsen S, Strube A, Heuer H, Bauer K. Analysis of hypertrophic thyrotrophs in pituitaries of athyroid Pax8-/- mice. Endocrinology 2009;150:4443-4449
  158. Cintra A, Fuxe K, Wikstrom AC, Visser T, Gustafsson JA. Evidence for thyrotropin-releasing hormone and glucocorticoid receptor-immunoreactive neurons in various preoptic and hypothalamic nuclei of the male rat. Brain Res 1990;506:139-144
  159. Jackson IM. Thyrotropin-releasing hormone and corticotropin-releasing hormone--what's the message? Endocrinology 1995;136:2793-2794
  160. Luo LG, Bruhn T, Jackson IM. Glucocorticoids stimulate thyrotropin-releasing hormone gene expression in cultured hypothalamic neurons. Endocrinology 1995;136:4945-4950
  161. Shibusawa N, Yamada M, Hirato J, Monden T, Satoh T, Mori M. Requirement of thyrotropin-releasing hormone for the postnatal functions of pituitary thyrotrophs: ontogeny study of congenital tertiary hypothyroidism in mice. Mol Endocrinol 2000;14:137-146.
  162. Straub RE, Frech GC, Joho RH, Gershengorn MC. Expression cloning of a cDNA encoding the mouse pituitary thyrotropin-releasing hormone receptor. Proc Natl Acad Sci U S A 1990;87:9514-9518
  163. Hinuma S, Hosoya M, Ogi K, Tanaka H, Nagai Y, Onda H. Molecular cloning and functional expression of a human thyrotropin-releasing hormone (TRH) receptor gene. Biochim Biophys Acta 1994;1219:251-259
  164. Bilek R, Starka L. The computer modelling of human TRH receptor, TRH and TRH-like peptides. Physiol Res 2005;54:141-150
  165. Stephenson A, Lau L, Eszlinger M, Paschke R. The Thyrotropin Receptor Mutation Database Update. Thyroid 2020;30:931-935
  166. Collu R, Tang J, Castagne J, Lagace G, Masson N, Huot C, Deal C, Delvin E, Faccenda E, Eidne KA, Van Vliet G. A novel mechanism for isolated central hypothyroidism: inactivating mutations in the thyrotropin-releasing hormone receptor gene. J Clin Endocrinol Metab 1997;82:1561-1565
  167. Collu R. Genetic aspects of central hypothyroidism. J Endocrinol Invest 2000;23:125-134.
  168. Bonomi M, Busnelli M, Beck-Peccoz P, Costanzo D, Antonica F, Dolci C, Pilotta A, Buzi F, Persani L. A family with complete resistance to thyrotropin-releasing hormone. N Engl J Med 2009;360:731-734
  169. Koulouri O, Nicholas AK, Schoenmakers E, Mokrosinski J, Lane F, Cole T, Kirk J, Farooqi IS, Chatterjee VK, Gurnell M, Schoenmakers N. A Novel Thyrotropin-Releasing Hormone Receptor Missense Mutation (P81R) in Central Congenital Hypothyroidism. J Clin Endocrinol Metab 2016;101:847-851
  170. Gershengorn MC. Bihormonal regulation of the thyrotropin-releasing hormone receptor in mouse pituitary thyrotropic tumor cells in culture. J Clin Invest 1978;62:937-943
  171. Fujimoto J, Gershengorn MC. Evidence for dual regulation by protein kinases A and C of thyrotropin-releasing hormone receptor mRNA in GH3 cells. Endocrinology 1991;129:3430-3432
  172. Gershengorn MC. Thyrotropin-releasing hormone action: mechanism of calcium-mediated stimulation of prolactin secretion. Recent Prog Horm Res 1985;41:607-653
  173. Brenner-Gati L, Gershengorn MC. Effects of thyrotropin-releasing hormone on phosphoinositides and cytoplasmic free calcium in thyrotropic pituitary cells. Endocrinology 1986;118:163-169.
  174. Oron Y, Gillo B, Straub RE, Gershengorn MC. Mechanism of membrane electrical response to thyrotropin-releasing hormone in Xenopus oocytes injected with GH3 pituitary cell messenger ribonucleic acid. Mol Endocrinol 1987;1:918-925
  175. Heinflink M, Nussenzveig DR, Friedman AM, Gershengorn MC. Thyrotropin-releasing hormone receptor activation does not elevate intracellular cyclic adenosine 3',5'-monophosphate in cells expressing high levels of receptors. J Clin Endocrinol Metab 1994;79:650-652
  176. Martin TF. Thyrotropin-releasing hormone rapidly activates the phosphodiester hydrolysis of polyphosphoinositides in GH3 pituitary cells. Evidence for the role of a polyphosphoinositide-specific phospholipase C in hormone action. J Biol Chem 1983;258:14816-14822
  177. Nelson EJ, Hinkle PM. Characteristics of the Ca2+ spike and oscillations induced by different doses of thyrotropin-releasing hormone (TRH) in individual pituitary cells and nonexcitable cells transfected with TRH receptor complementary deoxyribonucleic acid. Endocrinology 1994;135:1084-1092
  178. Schaarschmidt J, Nagel MB, Huth S, Jaeschke H, Moretti R, Hintze V, von Bergen M, Kalkhof S, Meiler J, Paschke R. Rearrangement of the Extracellular Domain/Extracellular Loop 1 Interface Is Critical for Thyrotropin Receptor Activation. J Biol Chem 2016;291:14095-14108
  179. Nelson EJ, Hinkle PM. Thyrotropin-releasing hormone activates Ca2+ efflux. Evidence suggesting that a plasma membrane Ca2+ pump is an effector for a G-protein-coupled Ca(2+)-mobilizing receptor. J Biol Chem 1994;269:30854-30860
  180. Ashworth R, Hinkle PM. Thyrotropin-releasing hormone-induced intracellular calcium responses in individual rat lactotrophs and thyrotrophs. Endocrinology 1996;137:5205-5212
  181. Shupnik MA, Weck J, Hinkle PM. Thyrotropin (TSH)-releasing hormone stimulates TSH beta promoter activity by two distinct mechanisms involving calcium influx through L type Ca2+ channels and protein kinase C. Mol Endocrinol 1996;10:90-99
  182. Weintraub BD, Wondisford FE, Farr EA, Steinfelder HJ, Radovick S, Gesundheit N, Gyves PW, Taylor T, DeCherney GS. Pre-translational and post-translational regulation of TSH synthesis in normal and neoplastic thyrotrophs. Horm Res 1989;32:22-24
  183. Steinfelder HJ, Hauser P, Nakayama Y, Radovick S, McClaskey JH, Taylor T, Weintraub BD, Wondisford FE. Thyrotropin-releasing hormone regulation of human TSHB expression: role of a pituitary-specific transcription factor (Pit-1/GHF-1) and potential interaction with a thyroid hormone-inhibitory element. Proc Natl Acad Sci U S A 1991;88:3130-3134
  184. Steinfelder HJ, Wondisford FE. Thyrotropin (TSH) beta-subunit gene expression--an example for the complex regulation of pituitary hormone genes. Exp Clin Endocrinol Diabetes 1997;105:196-203
  185. Heuer H, Schafer MK, Bauer K. The thyrotropin-releasing hormone-degrading ectoenzyme: the third element of the thyrotropin-releasing hormone-signaling system. Thyroid 1998;8:915-920
  186. Steinfelder HJ, Radovick S, Mroczynski MA, Hauser P, McClaskey JH, Weintraub BD, Wondisford FE. Role of a pituitary-specific transcription factor (pit-1/GHF-1) or a closely related protein in cAMP regulation of human thyrotropin-beta subunit gene expression. J Clin Invest 1992;89:409-419
  187. Steinfelder HJ, Radovick S, Wondisford FE. Hormonal regulation of the thyrotropin beta-subunit gene by phosphorylation of the pituitary-specific transcription factor Pit-1. Proc Natl Acad Sci U S A 1992;89:5942-5945
  188. Howard PW, Maurer RA. Thyrotropin releasing hormone stimulates transient phosphorylation of the tissue-specific transcription factor, Pit-1. J Biol Chem 1994;269:28662-28669
  189. Parks JS, Brown MR, Hurley DL, Phelps CJ, Wajnrajch MP. Heritable disorders of pituitary development. J Clin Endocrinol Metab 1999;84:4362-4370
  190. Camper SA, Saunders TL, Katz RW, Reeves RH. The Pit-1 transcription factor gene is a candidate for the murine Snell dwarf mutation. Genomics 1990;8:586-590
  191. Pfaffle RW, DiMattia GE, Parks JS, Brown MR, Wit JM, Jansen M, Van der Nat H, Van den Brande JL, Rosenfeld MG, Ingraham HA. Mutation of the POU-specific domain of Pit-1 and hypopituitarism without pituitary hypoplasia. Science 1992;257:1118-1121
  192. Radovick S, Nations M, Du Y, Berg LA, Weintraub BD, Wondisford FE. A mutation in the POU-homeodomain of Pit-1 responsible for combined pituitary hormone deficiency. Science 1992;257:1115-1118
  193. Tatsumi K, Miyai K, Notomi T, Kaibe K, Amino N, Mizuno Y, Kohno H. Cretinism with combined hormone deficiency caused by a mutation in the PIT1 gene. Nat Genet 1992;1:56-58
  194. Parks JS, Kinoshita EI, Pfaffle RW. Pit-1 and hypopituitarism. TEM 1993;4:81
  195. Hashimoto Y, Cisternino M, Cohen LE. A novel nonsense mutation in the Pit-1 gene: evidence for a gene dosage effect. J Clin Endocrinol Metab 2003;88:1241-1247.
  196. Turton JP, Reynaud R, Mehta A, Torpiano J, Saveanu A, Woods KS, Tiulpakov A, Zdravkovic V, Hamilton J, Attard-Montalto S, Parascandalo R, Vella C, Clayton PE, Shalet S, Barton J, Brue T, Dattani MT. Novel mutations within the POU1F1 gene associated with variable combined pituitary hormone deficiency. J Clin Endocrinol Metab 2005;90:4762-4770
  197. Yamamoto M, Iguchi G, Takeno R, Okimura Y, Sano T, Takahashi M, Nishizawa H, Handayaningshi AE, Fukuoka H, Tobita M, Saitoh T, Tojo K, Mokubo A, Morinobu A, Iida K, Kaji H, Seino S, Chihara K, Takahashi Y. Adult combined GH, prolactin, and TSH deficiency associated with circulating PIT-1 antibody in humans. J Clin Invest 2011;121:113-119
  198. Bando H, Iguchi G, Fukuoka H, Yamamoto M, Hidaka-Takeno R, Okimura Y, Matsumoto R, Suda K, Nishizawa H, Takahashi M, Tojo K, Takahashi Y. Involvement of PIT-1-reactive cytotoxic T lymphocytes in anti-PIT-1 antibody syndrome. J Clin Endocrinol Metab 2014;99:E1744-1749
  199. Kanie K, Bando H, Iguchi G, Muguruma K, Matsumoto R, Hidaka-Takeno R, Okimura Y, Yamamoto M, Fujita Y, Fukuoka H, Yoshida K, Suda K, Nishizawa H, Ogawa W, Takahashi Y. Pathogenesis of Anti-PIT-1 Antibody Syndrome: PIT-1 Presentation by HLA Class I on Anterior Pituitary Cells. J Endocr Soc 2019;3:1969-1978
  200. Yamamoto M, Iguchi G, Bando H, Kanie K, Hidaka-Takeno R, Fukuoka H, Takahashi Y. Autoimmune Pituitary Disease: New Concepts With Clinical Implications. Endocr Rev 2020;41
  201. Cohen LE, Wondisford FE, Radovick S. Role of Pit-1 in the gene expression of growth hormone, prolactin, and thyrotropin. Endocrinol Metab Clin North Am 1996;25:523-540
  202. Gordon DF, Haugen BR, Sarapura VD, Nelson AR, Wood WM, Ridgway EC. Analysis of Pit-1 in regulating mouse TSH beta promoter activity in thyrotropes. Mol Cell Endocrinol 1993;96:75-84
  203. Haugen BR, Wood WM, Gordon DF, Ridgway EC. A thyrotrope-specific variant of Pit-1 transactivates the thyrotropin beta promoter. J Biol Chem 1993;268:20818-20824
  204. Haugen BR, Gordon DF, Nelson AR, Wood WM, Ridgway EC. The combination of Pit-1 and Pit-1T have a synergistic stimulatory effect on the thyrotropin beta-subunit promoter but not the growth hormone or prolactin promoters. Mol Endocrinol 1994;8:1574-1582
  205. Flynn TR, Hollenberg AN, Cohen O, Menke JB, Usala SJ, Tollin S, Hegarty MK, Wondisford FE. A novel C-terminal domain in the thyroid hormone receptor selectively mediates thyroid hormone inhibition. J Biol Chem 1994;269:32713-32716
  206. Haugen BR, McDermott MT, Gordon DF, Rupp CL, Wood WM, Ridgway EC. Determinants of thyrotrope-specific thyrotropin beta promoter activation. Cooperation of Pit-1 with another factor. J Biol Chem 1996;271:385-389
  207. Gordon DF, Lewis SR, Haugen BR, James RA, McDermott MT, Wood WM, Ridgway EC. Pit-1 and GATA-2 interact and functionally cooperate to activate the thyrotropin beta-subunit promoter. J Biol Chem 1997;272:24339-24347
  208. Bach I, Rhodes SJ, Pearse RV, 2nd, Heinzel T, Gloss B, Scully KM, Sawchenko PE, Rosenfeld MG. P-Lim, a LIM homeodomain factor, is expressed during pituitary organ and cell commitment and synergizes with Pit-1. Proc Natl Acad Sci U S A 1995;92:2720-2724
  209. Sornson MW, Wu W, Dasen JS, Flynn SE, Norman DJ, O'Connell SM, Gukovsky I, Carriere C, Ryan AK, Miller AP, Zuo L, Gleiberman AS, Andersen B, Beamer WG, Rosenfeld MG. Pituitary lineage determination by the Prophet of Pit-1 homeodomain factor defective in Ames dwarfism. Nature 1996;384:327-333
  210. Deladoey J, Fluck C, Buyukgebiz A, Kuhlmann BV, Eble A, Hindmarsh PC, Wu W, Mullis PE. "Hot spot" in the PROP1 gene responsible for combined pituitary hormone deficiency. J Clin Endocrinol Metab 1999;84:1645-1650
  211. Vieira TC, Dias da Silva MR, Cerutti JM, Brunner E, Borges M, Arnaldi LT, Kopp P, Abucham J. Familial combined pituitary hormone deficiency due to a novel mutation R99Q in the hot spot region of Prophet of Pit-1 presenting as constitutional growth delay. J Clin Endocrinol Metab 2003;88:38-44.
  212. Arimura A, Schally AV. Increase in basal and thyrotropin-releasing hormone (TRH)-stimulated secretion of thyrotropin (TSH) by passive immunization with antiserum to somatostatin in rats. Endocrinology 1976;98:1069-1072
  213. Beck-Peccoz P, Brucker-Davis F, Persani L, Smallridge RC, Weintraub BD. Thyrotropin-secreting pituitary tumors. Endocr Rev 1996;17:610-638
  214. Ross DS, Ellis MF, Milbury P, Ridgway EC. A comparison of changes in plasma thyrotropin beta- and alpha-subunits, and mouse thyrotropic tumor thyrotropin beta- and alpha-subunit mRNA concentrations after in vivo dexamethasone or T3 administration. Metabolism 1987;36:799-803
  215. Coiro V, Volpi R, Cataldo S, Capretti L, Caffarri G, Pilla S, Chiodera P. Dopaminergic and cholinergic involvement in the inhibitory effect of dexamethasone on the TSH response to TRH. J Investig Med 2000;48:133-136.
  216. Ahlquist JA, Franklyn JA, Wood DF, Balfour NJ, Docherty K, Sheppard MC, Ramsden DB. Hormonal regulation of thyrotrophin synthesis and secretion. Horm Metab Res Suppl 1987;17:86-89
  217. Glass CK, Holloway JM, Devary OV, Rosenfeld MG. The thyroid hormone receptor binds with opposite transcriptional effects to a common sequence motif in thyroid hormone and estrogen response elements. Cell 1988;54:313-323
  218. Weeke J, Hansen AP, Lundaek K. Inhibition by somatostatin of basal levels of serum thyrotropin (TSH) in normal men. J Clin Endocrinol Metab 1975;41:168-171
  219. Weeke J, Christensen SE, Hansen AP, Laurberg P, Lundbaek K. Somatostatin and the 24 h levels of serum TSH, T3, T4, and reverse T3 in normals, diabetics and patients treated for myxoedema. Acta Endocrinol (Copenh) 1980;94:30-37
  220. Lamberts SW, Zuyderwijk J, den Holder F, van Koetsveld P, Hofland L. Studies on the conditions determining the inhibitory effect of somatostatin on adrenocorticotropin, prolactin and thyrotropin release by cultured rat pituitary cells. Neuroendocrinology 1989;50:44-50
  221. Yang I, Woo J, Kim S, Kim J, Kim Y, Choi Y. Suppression of TRH-stimulated TSH secretion by glucose-induced hypothalamic somatostatin release. Horm Metab Res 1996;28:553-557
  222. Comi RJ, Gesundheit N, Murray L, Gorden P, Weintraub BD. Response of thyrotropin-secreting pituitary adenomas to a long-acting somatostatin analogue. N Engl J Med 1987;317:12-17
  223. Beck-Peccoz P, Mariotti S, Guillausseau PJ, Medri G, Piscitelli G, Bertoli A, Barbarino A, Rondena M, Chanson P, Pinchera A, et al. Treatment of hyperthyroidism due to inappropriate secretion of thyrotropin with the somatostatin analog SMS 201-995. J Clin Endocrinol Metab 1989;68:208-214
  224. Reisine T, Bell GI. Molecular biology of somatostatin receptors. Endocr Rev 1995;16:427-442
  225. Nabekura J, Mizuno Y, Oomura Y. Inhibitory effect of somatostatin on vagal motoneurons in the rat brain stem in vitro. Am J Physiol 1989;256:C155-159
  226. Nilsson T, Arkhammar P, Rorsman P, Berggren PO. Suppression of insulin release by galanin and somatostatin is mediated by a G-protein. An effect involving repolarization and reduction in cytoplasmic free Ca2+ concentration. J Biol Chem 1989;264:973-980
  227. De Groef B, Goris N, Arckens L, Kuhn ER, Darras VM. Corticotropin-Releasing Hormone (CRH)-Induced Thyrotropin Release is Directly Mediated Through CRH Receptor Type 2 on Thyrotropes. Endocrinology 2003;144:5537-5544
  228. De Groef B, Geris KL, Manzano J, Bernal J, Millar RP, Abou-Samra AB, Porter TE, Iwasawa A, Kuhn ER, Darras VM. Involvement of thyrotropin-releasing hormone receptor, somatostatin receptor subtype 2 and corticotropin-releasing hormone receptor type 1 in the control of chicken thyrotropin secretion. Mol Cell Endocrinol 2003;203:33-39.
  229. Kennedy AR, Todd JF, Stanley SA, Abbott CR, Small CJ, Ghatei MA, Bloom SR. Melanin-concentrating hormone (MCH) suppresses thyroid stimulating hormone (TSH) release, in vivo and in vitro, via the hypothalamus and the pituitary. Endocrinology 2000;142:3265-3268.
  230. Fukuda H, Greer MA. The effect of basal hypothalamic deafferentation on the nycthemeral rhythm of plasma TSH. Endocrinology 1975;97:749-752
  231. Roelfsema F, Veldhuis JD. Thyrotropin secretion patterns in health and disease. Endocr Rev 2013;34:619-657
  232. Ikegami K, Refetoff S, Van Cauter E, Yoshimura T. Interconnection between circadian clocks and thyroid function. Nat Rev Endocrinol 2019;15:590-600
  233. Spaulding SW, Burrow GN, Donabedian R, van Woert M. L-DOPA suppression of thyrotropin-releasing hormone response in man. J Clin Endocrinol Metab 1972;35:182-185
  234. Besses GS, Burrow GN, Spaulding SW, Donabedian RK. Dopamine infusion acutely inhibits the TSH and prolactin response to TRH. J Clin Endocrinol Metab 1975;41:985-988
  235. Leebaw WF, Lee LA, Woolf PD. Dopamine affects basal and augmented pituitary hormone secretion. J Clin Endocrinol Metab 1978;47:480-487
  236. Lewis BM, Dieguez C, Lewis MD, Scanlon MF. Dopamine stimulates release of thyrotrophin-releasing hormone from perfused intact rat hypothalamus via hypothalamic D2-receptors. J Endocrinol 1987;115:419-424
  237. Lewis BM, Dieguez C, Ham J, Page MD, Creagh FM, Peters JR, Scanlon MF. Effects of Glucose on Thyrotrophin-Releasing Hormone, Growth Hormone-Releasing Hormone, Somatostatin and Luteinizing Hormone-Releasing Hormone Release from Rat Hypothalamus in vitro. J Neuroendocrinol 1989;1:437-441
  238. Krulich L, Giachetti A, Marchlewska-Koj A, Hefco E, Jameson HE. On the role of the central noradrenergic and dopaminergic systems in the regulation of TSH secretion in the rat. Endocrinology 1977;100:496-505
  239. Krulich L, Mayfield MA, Steele MK, McMillen BA, McCann SM, Koenig JI. Differential effects of pharmacological manipulations of central alpha 1- and alpha 2-adrenergic receptors on the secretion of thyrotropin and growth hormone in male rats. Endocrinology 1982;110:796-804
  240. Gershengorn MC, Rebecchi MJ, Geras E, Arevalo CO. Thyrotropin-releasing hormone (TRH) action in mouse thyrotropic tumor cells in culture: evidence against a role for adenosine 3',5'-monophosphate as a mediator of TRH-stimulated thyrotropin release. Endocrinology 1980;107:665-670
  241. Dieguez C, Foord SM, Peters JR, Hall R, Scanlon MF. Alpha 1-adrenoreceptors and alpha 1-adrenoreceptor-mediated thyrotropin release in cultures of euthyroid and hypothyroid rat anterior pituitary cells. Endocrinology 1985;117:624-630
  242. Tekin S, Erden Y, Ozyalin F, Onalan EE, Cigremis Y, Colak C, Tekedereli I, Sandal S. Central irisin administration suppresses thyroid hormone production but increases energy consumption in rats. Neurosci Lett 2018;674:136-141
  243. Sharp B, Morley JE, Carlson HE, Gordon J, Briggs J, Melmed S, Hershman JM. The role of opiates and endogenous opioid peptides in the regulation of rat TSH secretion. Brain Res 1981;219:335-344
  244. Samuels MH, Kramer P, Wilson D, Sexton G. Effects of naloxone infusions on pulsatile thyrotropin secretion. J Clin Endocrinol Metab 1994;78:1249-1252
  245. Peuranen E, Vasar E, Koks S, Volke V, Lang A, Rauhala P, Mannisto PT. Further studies on the role of cholecystokinin-A and B receptors in secretion of anterior pituitary hormones in male rats. Neuropeptides 1995;28:1-11
  246. Santos CV, Pazos-Moura CC, Moura EG. Effect of gastrin-releasing peptide (GRP) and GRP antagonists on TSH secretion from rat isolated pituitaries. Life Sci 1995;57:911-915
  247. Fekete C, Kelly J, Mihaly E, Sarkar S, Rand WM, Legradi G, Emerson CH, Lechan RM. Neuropeptide Y has a central inhibitory action on the hypothalamic- pituitary-thyroid axis. Endocrinology 2001;142:2606-2613.
  248. Ciosek J, Stempniak B. The influence of vasopressin or oxytocin on thyroid-stimulating hormone and thyroid hormones' concentrations in blood plasma of euthyroid rats. J Physiol Pharmacol 1997;48:813-823
  249. Beak SA, Small CJ, Ilovaiskaia I, Hurley JD, Ghatei MA, Bloom SR, Smith DM. Glucagon-like peptide-1 (GLP-1) releases thyrotropin (TSH): characterization of binding sites for GLP-1 on alpha-TSH cells. Endocrinology 1996;137:4130-4138.
  250. Arvat E, Gianotti L, Ramunni J, Grottoli S, Brossa PC, Bertagna A, Camanni F, Ghigo E. Effect of galanin on basal and stimulated secretion of prolactin, gonadotropins, thyrotropin, adrenocorticotropin and cortisol in humans. Eur J Endocrinol 1995;133:300-304
  251. Nillni EA, Vaslet C, Harris M, Hollenberg A, Bjorbak C, Flier JS. Leptin regulates prothyrotropin-releasing hormone biosynthesis. Evidence for direct and indirect pathways. J Biol Chem 2000;275:36124-36133.
  252. Seoane LM, Carro E, Tovar S, Casanueva FF, Dieguez C. Regulation of in vivo TSH secretion by leptin. Regul Pept 2000;92:25-29.
  253. Flier JS, Harris M, Hollenberg AN. Leptin, nutrition, and the thyroid: the why, the wherefore, and the wiring. J Clin Invest 2000;105:859-861.
  254. Dubuis JM, Dayer JM, Siegrist-Kaiser CA, Burger AG. Human recombinant interleukin-1 beta decreases plasma thyroid hormone and thyroid stimulating hormone levels in rats. Endocrinology 1988;123:2175-2181
  255. Ozawa M, Sato K, Han DC, Kawakami M, Tsushima T, Shizume K. Effects of tumor necrosis factor-alpha/cachectin on thyroid hormone metabolism in mice. Endocrinology 1988;123:1461-1467
  256. Pang XP, Hershman JM, Mirell CJ, Pekary AE. Impairment of hypothalamic-pituitary-thyroid function in rats treated with human recombinant tumor necrosis factor-alpha (cachectin). Endocrinology 1989;125:76-84
  257. van der Poll T, Romijn JA, Wiersinga WM, Sauerwein HP. Tumor necrosis factor: a putative mediator of the sick euthyroid syndrome in man. J Clin Endocrinol Metab 1990;71:1567-1572
  258. Wassen FW, Moerings EP, Van Toor H, De Vrey EA, Hennemann G, Everts ME. Effects of interleukin-1 beta on thyrotropin secretion and thyroid hormone uptake in cultured rat anterior pituitary cells. Endocrinology 1996;137:1591-1598
  259. Melmed S. gp130-related cytokines and their receptors in the pituitary. TEM 1998;9:155
  260. Koenig JI, Snow K, Clark BD, Toni R, Cannon JG, Shaw AR, Dinarello CA, Reichlin S, Lee SL, Lechan RM. Intrinsic pituitary interleukin-1 beta is induced by bacterial lipopolysaccharide. Endocrinology 1990;126:3053-3058
  261. Rettori V, Dees WL, Hiney JK, Lyson K, McCann SM. An interleukin-1-alpha-like neuronal system in the preoptic-hypothalamic region and its induction by bacterial lipopolysaccharide in concentrations which alter pituitary hormone release. Neuroimmunomodulation 1994;1:251-258.
  262. Bartalena L, Grasso L, Brogioni S, Martino E. Interleukin 6 effects on the pituitary-thyroid axis in the rat. Eur J Endocrinol 1994;131:302-306
  263. van Haasteren GA, van der Meer MJ, Hermus AR, Linkels E, Klootwijk W, Kaptein E, van Toor H, Sweep CG, Visser TJ, de Greef WJ. Different effects of continuous infusion of interleukin-1 and interleukin-6 on the hypothalamic-hypophysial-thyroid axis. Endocrinology 1994;135:1336-1345
  264. Baur A, Bauer K, Jarry H, Kohrle J. Effects of proinflammatory cytokines on anterior pituitary 5'- deiodinase type I and type II. J Endocrinol 2000;167:505-515.
  265. Vankelecom H, Carmeliet P, Van Damme J, Billiau A, Denef C. Production of interleukin-6 by folliculo-stellate cells of the anterior pituitary gland in a histiotypic cell aggregate culture system. Neuroendocrinology 1989;49:102-106
  266. Spangelo BL, MacLeod RM, Isakson PC. Production of interleukin-6 by anterior pituitary cells in vitro. Endocrinology 1990;126:582-586
  267. Akieda-Asai S, Zaima N, Ikegami K, Kahyo T, Yao I, Hatanaka T, Iemura S, Sugiyama R, Yokozeki T, Eishi Y, Koike M, Ikeda K, Chiba T, Yamaza H, Shimokawa I, Song SY, Matsuno A, Mizutani A, Sawabe M, Chao MV, Tanaka M, Kanaho Y, Natsume T, Sugimura H, Date Y, McBurney MW, Guarente L, Setou M. SIRT1 Regulates Thyroid-Stimulating Hormone Release by Enhancing PIP5Kgamma Activity through Deacetylation of Specific Lysine Residues in Mammals. PLoS One 2010;5:e11755
  268. Prummel MF, Brokken LJ, Wiersinga WM. Ultra short-loop feedback control of thyrotropin secretion. Thyroid 2004;14:825-829
  269. Bockmann J, Winter C, Wittkowski W, Kreutz MR, Bockers TM. Cloning and expression of a brain-derived TSH receptor. Biochem Biophys Res Commun 1997;238:173-178
  270. Crisanti P, Omri B, Hughes E, Meduri G, Hery C, Clauser E, Jacquemin C, Saunier B. The expression of thyrotropin receptor in the brain. Endocrinology 2001;142:812-822.
  271. Prummel MF, Brokken LJ, Meduri G, Misrahi M, Bakker O, Wiersinga WM. Expression of the thyroid-stimulating hormone receptor in the folliculo- stellate cells of the human anterior pituitary. J Clin Endocrinol Metab 2000;85:4347-4353.
  272. Miura Y, Perkel VS, Papenberg KA, Johnson MJ, Magner JA. Concanavalin-A, lentil, and ricin lectin affinity binding characteristics of human thyrotropin: differences in the sialylation of thyrotropin in sera of euthyroid, primary, and central hypothyroid patients. J Clin Endocrinol Metab 1989;69:985-995
  273. Papandreou MJ, Persani L, Asteria C, Ronin C, Beck-Peccoz P. Variable carbohydrate structures of circulating thyrotropin as studied by lectin affinity chromatography in different clinical conditions. J Clin Endocrinol Metab 1993;77:393-398
  274. Wartofsky L, Dickey RA. The evidence for a narrower thyrotropin reference range is compelling. J Clin Endocrinol Metab 2005;90:5483-5488
  275. Surks MI, Goswami G, Daniels GH. The thyrotropin reference range should remain unchanged. J Clin Endocrinol Metab 2005;90:5489-5496
  276. Surks MI, Hollowell JG. Age-specific distribution of serum thyrotropin and antithyroid antibodies in the US population: implications for the prevalence of subclinical hypothyroidism. J Clin Endocrinol Metab 2007;92:4575-4582
  277. Surks MI, Boucai L. Age- and race-based serum thyrotropin reference limits. J Clin Endocrinol Metab 2010;95:496-502
  278. Mariotti S, Franceschi C, Cossarizza A, Pinchera A. The aging thyroid. Endocr Rev 1995;16:686-715
  279. Hansen PS, Brix TH, Sorensen TI, Kyvik KO, Hegedus L. Major genetic influence on the regulation of the pituitary-thyroid axis: a study of healthy Danish twins. J Clin Endocrinol Metab 2004;89:1181-1187
  280. Mariotti S, Naitza S, Cao A. Phosphodiesterase 8B (PDE8B) gene variants and TSH levels. Hot Thyroidtology 2010;3:http://www.hotthyroidology.com/editorial_211.html
  281. Arnaud-Lopez L, Usala G, Ceresini G, Mitchell BD, Pilia MG, Piras MG, Sestu N, Maschio A, Busonero F, Albai G, Dei M, Lai S, Mulas A, Crisponi L, Tanaka T, Bandinelli S, Guralnik JM, Loi A, Balaci L, Sole G, Prinzis A, Mariotti S, Shuldiner AR, Cao A, Schlessinger D, Uda M, Abecasis GR, Nagaraja R, Sanna S, Naitza S. Phosphodiesterase 8B gene variants are associated with serum TSH levels and thyroid function. Am J Hum Genet 2008;82:1270-1280
  282. Kuzuya N, Inoue K, Ishibashi M, Murayama Y, Koide Y, Ito K, Yamaji T, Yamashita K. Endocrine and immunohistochemical studies on thyrotropin (TSH)-secreting pituitary adenomas: responses of TSH, alpha-subunit, and growth hormone to hypothalamic releasing hormones and their distribution in adenoma cells. J Clin Endocrinol Metab 1990;71:1103-1111
  283. Faglia G, Beck-Peccoz P, Piscitelli G, Medri G. Inappropriate secretion of thyrotropin by the pituitary. Horm Res 1987;26:79-99
  284. Beck-Peccoz P, Persani L, Faglia G. Glycoprotein hormone alpha-subunit in pituitary adenomas. Trends Endocrinol Metab 1992;3:41-45
  285. Spencer CA, Lai-Rosenfeld AO, Guttler RB, LoPresti J, Marcus AO, Nimalasuriya A, Eigen A, Doss RC, Green BJ, Nicoloff JT. Thyrotropin secretion in thyrotoxic and thyroxine-treated patients: assessment by a sensitive immunoenzymometric assay. J Clin Endocrinol Metab 1986;63:349-355
  286. Spencer CA, LoPresti JS, Patel A, Guttler RB, Eigen A, Shen D, Gray D, Nicoloff JT. Applications of a new chemiluminometric thyrotropin assay to subnormal measurement. J Clin Endocrinol Metab 1990;70:453-460
  287. Spencer CA, Schwarzbein D, Guttler RB, LoPresti JS, Nicoloff JT. Thyrotropin (TSH)-releasing hormone stimulation test responses employing third and fourth generation TSH assays. J Clin Endocrinol Metab 1993;76:494-498
  288. Greenspan SL, Klibanski A, Schoenfeld D, Ridgway EC. Pulsatile secretion of thyrotropin in man. J Clin Endocrinol Metab 1986;63:661-668
  289. Brabant G, Brabant A, Ranft U, Ocran K, Kohrle J, Hesch RD, von zur Muhlen A. Circadian and pulsatile thyrotropin secretion in euthyroid man under the influence of thyroid hormone and glucocorticoid administration. J Clin Endocrinol Metab 1987;65:83-88.
  290. Brabant G, Prank K, Ranft U, Schuermeyer T, Wagner TO, Hauser H, Kummer B, Feistner H, Hesch RD, von zur Muhlen A. Physiological regulation of circadian and pulsatile thyrotropin secretion in normal man and woman. J Clin Endocrinol Metab 1990;70:403-409.
  291. Samuels MH, Veldhuis J, Ridgway EC. Copulsatile release of thyrotropin and prolactin in normal and hypothyroid subjects. Thyroid 1995;5:369-372.
  292. Weeke J. Circadian variation of the serum thyrotropin level in normal subjects. Scand J Clin Lab Invest 1973;31:337-342
  293. Bartalena L, Placidi GF, Martino E, Falcone M, Pellegrini L, Dell'Osso L, Pacchiarotti A, Pinchera A. Nocturnal serum thyrotropin (TSH) surge and the TSH response to TSH-releasing hormone: dissociated behavior in untreated depressives. J Clin Endocrinol Metab 1990;71:650-655
  294. Parker DC, Pekary AE, Hershman JM. Effect of normal and reversed sleep-wake cycles upon nyctohemeral rhythmicity of plasma thyrotropin: evidence suggestive of an inhibitory influence in sleep. J Clin Endocrinol Metab 1976;43:318-329.
  295. Parker DC, Rossman LG, Pekary AE, Hershman JM. Effect of 64-hour sleep deprivation on the circadian waveform of thyrotropin (TSH): further evidence of sleep-related inhibition of TSH release. J Clin Endocrinol Metab 1987;64:157-161
  296. Behrends J, Prank K, Dogu E, Brabant G. Central nervous system control of thyrotropin secretion during sleep and wakefulness. Horm Res 1998;49:173-177
  297. Yoshihara A, Noh JY, Watanabe N, Iwaku K, Kunii Y, Ohye H, Suzuki M, Matsumoto M, Suzuki N, Sugino K, Thienpont LM, Hishinuma A, Ito K. Seasonal Changes in Serum Thyrotropin Concentrations Observed from Big Data Obtained During Six Consecutive Years from 2010 to 2015 at a Single Hospital in Japan. Thyroid 2018;28:429-436
  298. Hirshberg B, Veldhuis JD, Sarlis NJ. Diurnal thyrotropin secretion in short-term profound primary hypothyroidism: does it ever persist? Thyroid 2000;10:1101-1106.
  299. Olsen T, Laurberg P, Weeke J. Low serum triiodothyronine and high serum reverse triiodothyronine in old age: an effect of disease not age. J Clin Endocrinol Metab 1978;47:1111-1115
  300. Sawin CT, Geller A, Kaplan MM, Bacharach P, Wilson PW, Hershman JM. Low serum thyrotropin (thyroid-stimulating hormone) in older persons without hyperthyroidism. Arch Intern Med 1991;151:165-168
  301. Barbesino G. Thyroid Function Changes in the Elderly and Their Relationship to Cardiovascular Health: A Mini-Review. Gerontology 2019;65:1-8
  302. Balsam A, Ingbar SH. Observations on the factors that control the generation of triiodothyronine from thyroxine in rat liver and the nature of the defect induced by fasting. J Clin Invest 1979;63:1145-1156
  303. Harris AR, Fang SL, Hinerfeld L, Braverman LE, Vagenakis AG. The role of sulfhydryl groups on the impaired hepatic 3',3,5-triiodothyronine generation from thyroxine in the hypothyroid, starved, fetal, and neonatal rodent. J Clin Invest 1979;63:516-524
  304. Kaplan MM, Tatro JB, Breitbart R, Larsen PR. Comparison of thyroxine and 3,3',5'-triiodothyronine metabolism in rat kidney and liver homogenates. Metabolism 1979;28:1139-1146
  305. Kinlaw WB, Schwartz HL, Oppenheimer JH. Decreased serum triiodothyronine in starving rats is due primarily to diminished thyroidal secretion of thyroxine. J Clin Invest 1985;75:1238-1241
  306. Wartofsky L, Burman KD. Alterations in thyroid function in patients with systemic illness: the "euthyroid sick syndrome". Endocr Rev 1982;3:164-217.
  307. Hugues JN, Burger AG, Pekary AE, Hershman JM. Rapid adaptations of serum thyrotrophin, triiodothyronine and reverse triiodothyronine levels to short-term starvation and refeeding. Acta Endocrinol (Copenh) 1984;105:194-199.
  308. Wehmann RE, Gregerman RI, Burns WH, Saral R, Santos GW. Suppression of thyrotropin in the low-thyroxine state of severe nonthyroidal illness. N Engl J Med 1985;312:546-552.
  309. Samuels MH, Kramer P. Differential effects of short-term fasting on pulsatile thyrotropin, gonadotropin, and alpha-subunit secretion in healthy men--a clinical research center study. J Clin Endocrinol Metab 1996;81:32-36.
  310. Samuels MH, Kramer P. Effects of metoclopramide on fasting-induced TSH suppression. Thyroid 1996;6:85-89.
  311. Carlson HE, Drenick EJ, Chopra IJ, Hershman JM. Alterations in basal and TRH-stimulated serum levels of thyrotropin, prolactin, and thyroid hormones in starved obese men. J Clin Endocrinol Metab 1977;45:707-713.
  312. Spencer CA, Lum SM, Wilber JF, Kaptein EM, Nicoloff JT. Dynamics of serum thyrotropin and thyroid hormone changes in fasting. J Clin Endocrinol Metab 1983;56:883-888.
  313. Blake NG, Eckland DJ, Foster OJ, Lightman SL. Inhibition of hypothalamic thyrotropin-releasing hormone messenger ribonucleic acid during food deprivation. Endocrinology 1991;129:2714-2718.
  314. Shi ZX, Levy A, Lightman SL. The effect of dietary protein on thyrotropin-releasing hormone and thyrotropin gene expression. Brain Res 1993;606:1-4.
  315. Hugues JN, Enjalbert A, Moyse E, Shu C, Voirol MJ, Sebaoun J, Epelbaum J. Differential effects of passive immunization with somatostatin antiserum on adenohypophysial hormone secretions in starved rats. J Endocrinol 1986;109:169-174.
  316. Kok P, Roelfsema F, Frolich M, Meinders AE, Pijl H. Spontaneous Diurnal Tsh Secretion Is Enhanced in Proportion to Circulating Leptin in Obese Premenopausal Women. J Clin Endocrinol Metab 2005;
  317. Kok P, Roelfsema F, Langendonk JG, Frolich M, Burggraaf J, Meinders AE, Pijl H. High circulating thyrotropin levels in obese women are reduced after body weight loss induced by caloric restriction. J Clin Endocrinol Metab 2005;90:4659-4663
  318. Legradi G, Emerson CH, Ahima RS, Flier JS, Lechan RM. Leptin prevents fasting-induced suppression of prothyrotropin-releasing hormone messenger ribonucleic acid in neurons of the hypothalamic paraventricular nucleus. Endocrinology 1997;138:2569-2576.
  319. Harris M, Aschkenasi C, Elias CF, Chandrankunnel A, Nillni EA, Bjoorbaek C, Elmquist JK, Flier JS, Hollenberg AN. Transcriptional regulation of the thyrotropin-releasing hormone gene by leptin and melanocortin signaling. J Clin Invest 2001;107:111-120.
  320. Kim MS, Small CJ, Stanley SA, Morgan DG, Seal LJ, Kong WM, Edwards CM, Abusnana S, Sunter D, Ghatei MA, Bloom SR. The central melanocortin system affects the hypothalamo-pituitary thyroid axis and may mediate the effect of leptin. J Clin Invest 2000;105:1005-1011.
  321. Guo F, Bakal K, Minokoshi Y, Hollenberg AN. Leptin signaling targets the thyrotropin-releasing hormone gene promoter in vivo. Endocrinology 2004;145:2221-2227
  322. Huo L, Munzberg H, Nillni EA, Bjorbaek C. Role of signal transducer and activator of transcription 3 in regulation of hypothalamic trh gene expression by leptin. Endocrinology 2004;145:2516-2523
  323. Mihaly E, Fekete C, Tatro JB, Liposits Z, Stopa EG, Lechan RM. Hypophysiotropic thyrotropin-releasing hormone-synthesizing neurons in the human hypothalamus are innervated by neuropeptide Y, agouti-related protein, and alpha-melanocyte-stimulating hormone. J Clin Endocrinol Metab 2000;85:2596-2603.
  324. Fekete C, Marks DL, Sarkar S, Emerson CH, Rand WM, Cone RD, Lechan RM. Effect of Agouti-related protein in regulation of the hypothalamic-pituitary-thyroid axis in the melanocortin 4 receptor knockout mouse. Endocrinology 2004;145:4816-4821
  325. Fekete C, Sarkar S, Rand WM, Harney JW, Emerson CH, Bianco AC, Beck-Sickinger A, Lechan RM. Neuropeptide Y1 and Y5 receptors mediate the effects of neuropeptide Y on the hypothalamic-pituitary-thyroid axis. Endocrinology 2002;143:4513-4519.
  326. Wittmann G, Liposits Z, Lechan RM, Fekete C. Medullary adrenergic neurons contribute to the neuropeptide Y-ergic innervation of hypophysiotropic thyrotropin-releasing hormone-synthesizing neurons in the rat. Neurosci Lett 2002;324:69-73.
  327. Kaplan MM, Larsen PR, Crantz FR, Dzau VJ, Rossing TH, Haddow JE. Prevalence of abnormal thyroid function test results in patients with acute medical illnesses. Am J Med 1982;72:9-16.
  328. Romijn JA, Adriaanse R, Brabant G, Prank K, Endert E, Wiersinga WM. Pulsatile secretion of thyrotropin during fasting: a decrease of thyrotropin pulse amplitude. J Clin Endocrinol Metab 1990;70:1631-1636.
  329. Kamat V, Hecht WL, Rubin RT. Influence of meal composition on the postprandial response of the pituitary-thyroid axis. Eur J Endocrinol 1995;133:75-79.
  330. Oppert JM, Dussault JH, Tremblay A, Despres JP, Theriault G, Bouchard C. Thyroid hormones and thyrotropin variations during long term overfeeding in identical twins. J Clin Endocrinol Metab 1994;79:547-553.
  331. Knudsen N, Laurberg P, Rasmussen LB, Bulow I, Perrild H, Ovesen L, Jorgensen T. Small differences in thyroid function may be important for body mass index and the occurrence of obesity in the population. J Clin Endocrinol Metab 2005;90:4019-4024
  332. An YM, Moon SJ, Kim SK, Suh YJ, Lee JE. Thyroid function in obese Korean children and adolescents: Korea National Health and Nutrition Examination Survey 2013-2015. Ann Pediatr Endocrinol Metab 2018;23:141-147
  333. Cho WK, Nam HK, Kim JH, Rhie YJ, Chung S, Lee KH, Suh BK. Thyroid Function in Korean Adolescents with Obesity: Results from the Korea National Health and Nutrition Examination Survey VI (2013-2015). Int J Endocrinol 2018;2018:6874395
  334. Song E, Ahn J, Oh HS, Jeon MJ, Kim WG, Kim WB, Shong YK, Kim TY. Sex-Dependent Association between Weight Change and Thyroid Dysfunction: Population-Level Analysis Using the Korean National Health and Nutrition Examination Survey. Eur Thyroid J 2019;8:202-207
  335. Hamblin PS, Dyer SA, Mohr VS, Le Grand BA, Lim CF, Tuxen DV, Topliss DJ, Stockigt JR. Relationship between thyrotropin and thyroxine changes during recovery from severe hypothyroxinemia of critical illness. J Clin Endocrinol Metab 1986;62:717-722.
  336. Faber J, Kirkegaard C, Rasmussen B, Westh H, Busch-Sorensen M, Jensen IW. Pituitary-thyroid axis in critical illness. J Clin Endocrinol Metab 1987;65:315-320.
  337. Ehrmann DA, Weinberg M, Sarne DH. Limitations to the use of a sensitive assay for serum thyrotropin in the assessment of thyroid status. Arch Intern Med 1989;149:369-372.
  338. Bartalena L, Martino E, Brandi LS, Falcone M, Pacchiarotti A, Ricci C, Bogazzi F, Grasso L, Mammoli C, Pinchera A. Lack of nocturnal serum thyrotropin surge after surgery. J Clin Endocrinol Metab 1990;70:293-296.
  339. Romijn JA, Wiersinga WM. Decreased nocturnal surge of thyrotropin in nonthyroidal illness. J Clin Endocrinol Metab 1990;70:35-42.
  340. Custro N, Scafidi V, Gallo S, Notarbartolo A. Deficient pulsatile thyrotropin secretion in the low-thyroid-hormone state of severe non-thyroidal illness. Eur J Endocrinol 1994;130:132-136.
  341. Van den Berghe G, de Zegher F, Veldhuis JD, Wouters P, Gouwy S, Stockman W, Weekers F, Schetz M, Lauwers P, Bouillon R, Bowers CY. Thyrotrophin and prolactin release in prolonged critical illness: dynamics of spontaneous secretion and effects of growth hormone-secretagogues. Clin Endocrinol (Oxf) 1997;47:599-612.
  342. Bacci V, Schussler GC, Kaplan TB. The relationship between serum triiodothyronine and thyrotropin during systemic illness. J Clin Endocrinol Metab 1982;54:1229-1235.
  343. Brent GA, Hershman JM, Braunstein GD. Patients with severe nonthyroidal illness and serum thyrotropin concentrations in the hypothyroid range. Am J Med 1986;81:463-466.
  344. Magner J, Roy P, Fainter L, Barnard V, Fletcher P, Jr. Transiently decreased sialylation of thyrotropin (TSH) in a patient with the euthyroid sick syndrome. Thyroid 1997;7:55-61.
  345. De Groot LJ. Dangerous dogmas in medicine: the nonthyroidal illness syndrome. J Clin Endocrinol Metab 1999;84:151-164.
  346. Fliers E, Alkemade A, Wiersinga WM. The hypothalamic-pituitary-thyroid axis in critical illness. Best Pract Res Clin Endocrinol Metab 2001;15:453-464.
  347. Hennessey J, Jackson IMD. The interface between thyroid hormones and psychiatry. The endocrinologist 1996;6:214
  348. Gold MS, Pottash AL, Extein I, Martin DM, Howard E, Mueller EA, 3rd, Sweeney DR. The TRH test in the diagnosis of major and minor depression. Psychoneuroendocrinology 1981;6:159-169.
  349. Chopra IJ, Solomon DH, Huang TS. Serum thyrotropin in hospitalized psychiatric patients: evidence for hyperthyrotropinemia as measured by an ultrasensitive thyrotropin assay. Metabolism 1990;39:538-543.
  350. Banki CM, Bissette G, Arato M, Nemeroff CB. Elevation of immunoreactive CSF TRH in depressed patients. Am J Psychiatry 1988;145:1526-1531.
  351. Loosen PT. Thyroid function in affective disorders and alcoholism. Endocrinol Metab Clin North Am 1988;17:55-82.
  352. Kirkegaard C, Carroll BJ. Dissociation of TSH adrenocortical disturbances in endogenous depression. Psychiatry Res 1980;3:253-264.
  353. O'Shanick GJ, Ellinwood EH, Jr. Persistent elevation of thyroid-stimulating hormone in women with bipolar affective disorder. Am J Psychiatry 1982;139:513-514.
  354. Fliers E, Guldenaar SE, Wiersinga WM, Swaab DF. Decreased hypothalamic thyrotropin-releasing hormone gene expression in patients with nonthyroidal illness. J Clin Endocrinol Metab 1997;82:4032-4036
  355. Delitala G, Tomasi P, Virdis R. Prolactin, growth hormone and thyrotropin-thyroid hormone secretion during stress states in man. Baillieres Clin Endocrinol Metab 1987;1:391-414.
  356. Chopra IJ, Sakane S, Teco GN. A study of the serum concentration of tumor necrosis factor-alpha in thyroidal and nonthyroidal illnesses. J Clin Endocrinol Metab 1991;72:1113-1116.
  357. Boelen A, Platvoet-Ter Schiphorst MC, Wiersinga WM. Association between serum interleukin-6 and serum 3,5,3'-triiodothyronine in nonthyroidal illness. J Clin Endocrinol Metab 1993;77:1695-1699.
  358. Davies PH, Black EG, Sheppard MC, Franklyn JA. Relation between serum interleukin-6 and thyroid hormone concentrations in 270 hospital in-patients with non-thyroidal illness. Clin Endocrinol (Oxf) 1996;44:199-205.
  359. Murai H, Murakami S, Ishida K, Sugawara M. Elevated serum interleukin-6 and decreased thyroid hormone levels in postoperative patients and effects of IL-6 on thyroid cell function in vitro. Thyroid 1996;6:601-606.
  360. Hermus RM, Sweep CG, van der Meer MJ, Ross HA, Smals AG, Benraad TJ, Kloppenborg PW. Continuous infusion of interleukin-1 beta induces a nonthyroidal illness syndrome in the rat. Endocrinology 1992;131:2139-2146.
  361. Sweep CG, van der Meer MJ, Ross HA, Vranckx R, Visser TJ, Hermus AR. Chronic infusion of TNF-alpha reduces plasma T4 binding without affecting pituitary-thyroid activity in rats. Am J Physiol 1992;263:E1099-1105.
  362. Bartalena L, Brogioni S, Grasso L, Martino E. Interleukin-6 and the thyroid. Eur J Endocrinol 1995;132:386-393.
  363. Kaptein EM, Spencer CA, Kamiel MB, Nicoloff JT. Prolonged dopamine administration and thyroid hormone economy in normal and critically ill subjects. J Clin Endocrinol Metab 1980;51:387-393.
  364. Nicoloff JT, Fisher DA, Appleman MD, Jr. The role of glucocorticoids in the regulation of thyroid function in man. J Clin Invest 1970;49:1922-1929.
  365. Re RN, Kourides IA, Ridgway EC, Weintraub BD, Maloof F. The effect of glucocorticoid administration on human pituitary secretion of thyrotropin and prolactin. J Clin Endocrinol Metab 1976;43:338-346
  366. Duick DS, Wahner HW. Thyroid axis in patients with Cushing's syndrome. Arch Intern Med 1979;139:767-772.
  367. Alkemade A, Unmehopa UA, Wiersinga WM, Swaab DF, Fliers E. Glucocorticoids decrease thyrotropin-releasing hormone messenger ribonucleic acid expression in the paraventricular nucleus of the human hypothalamus. J Clin Endocrinol Metab 2005;90:323-327
  368. Gharib H, Hodgson SF, Gastineau CF, Scholz DA, Smith LA. Reversible hypothyroidism in Addison's disease. Lancet 1972;2:734-736.
  369. Topliss DJ, White EL, Stockigt JR. Significance of thyrotropin excess in untreated primary adrenal insufficiency. J Clin Endocrinol Metab 1980;50:52-56.
  370. Surks MI, Sievert R. Drugs and thyroid function. N Engl J Med 1995;333:1688-1694.
  371. Faglia G, Beck-Peccoz P, Ferrari C, Ambrosi B, Spada A, Travaglini P. Enhanced plasma thyrotrophin response to thyrotrophin-releasing hormone following oestradiol administration in man. Clin Endocrinol (Oxf) 1973;2:207-210.
  372. Gershengorn MC, Marcus-Samuels BE, Geras E. Estrogens increase the number of thyrotropin-releasing hormone receptors on mammotropic cells in culture. Endocrinology 1979;105:171-176.
  373. Morley JE, Sawin CT, Carlson HE, Longcope C, Hershman JM. The relationship of androgen to the thyrotropin and prolactin responses to thyrotropin-releasing hormone in hypogonadal and normal men. J Clin Endocrinol Metab 1981;52:173-176.
  374. Arafah BM. Decreased levothyroxine requirement in women with hypothyroidism during androgen therapy for breast cancer. Ann Intern Med 1994;121:247-251.
  375. Porter BA, Refetoff S, Rosenfeld RL, De Groat LJ, Lang US, Stark O. Abnormal thyroxine metabolism in hyposomatotrophic dwarfism and inhibition of responsiveness to TRH during GH therapy. Pediatrics 1973;51:668-674.
  376. Lippe BM, Van Herle AJ, LaFranchi SH, Uller RP, Lavin N, Kaplan SA. Reversible hypothyroidism in growth hormone-deficient children treated with human growth hormone. J Clin Endocrinol Metab 1975;40:612-618.
  377. Giavoli C, Porretti S, Ferrante E, Cappiello V, Ronchi CL, Travaglini P, Epaminonda P, Arosio M, Beck-Peccoz P. Recombinant hGH replacement therapy and the hypothalamus-pituitary-thyroid axis in children with GH deficiency: when should we be concerned about the occurrence of central hypothyroidism? Clin Endocrinol (Oxf) 2003;59:806-810
  378. Jorgensen JO, Pedersen SA, Laurberg P, Weeke J, Skakkebaek NE, Christiansen JS. Effects of growth hormone therapy on thyroid function of growth hormone-deficient adults with and without concomitant thyroxine-substituted central hypothyroidism. J Clin Endocrinol Metab 1989;69:1127-1132.
  379. Trainer PJ, Holly J, Medbak S, Rees LH, Besser GM. The effect of recombinant IGF-I on anterior pituitary function in healthy volunteers. Clin Endocrinol (Oxf) 1994;41:801-807.
  380. Rogol AD, Reeves GD, Varma MM, Blizzard RM. Thyroid-stimulating hormone and prolactin responses to thyrotropin-releasing hormone during infusion of epinephrine and propranolol in man. Neuroendocrinology 1979;29:413-417.
  381. Little KY, Garbutt JC, Mayo JP, Mason G. Lack of acute d-amphetamine effects on thyrotropin release. Neuroendocrinology 1988;48:304-307.
  382. Zgliczynski S, Kaniewski M. Evidence for alpha-adrenergic receptors mediated TSH release in men. Acta Endocrinol (Copenh) 1980;95:172-176.
  383. Valcavi R, Dieguez C, Azzarito C, Artioli C, Portioli I, Scanlon MF. Alpha-adrenoreceptor blockade with thymoxamine reduces basal thyrotrophin levels but does not influence circadian thyrotrophin changes in man. J Endocrinol 1987;115:187-191.
  384. Lotti G, Delitala G, Devilla L, Alagna S, Masala A. Reduction of plasma triiodothyronine (T3) induced by propranolol. Clin Endocrinol (Oxf) 1977;6:405-410.
  385. Plosker SM, Rabinovici J, Montalvo M, Jaffe RB. Endogenous catecholamines suppress thyrotropin secretion during the early follicular phase of the menstrual cycle. J Clin Endocrinol Metab 1995;80:2530-2533.
  386. Hay ID, Bayer MF, Kaplan MM, Klee GG, Larsen PR, Spencer CA. American Thyroid Association assessment of current free thyroid hormone and thyrotropin measurements and guidelines for future clinical assays. The Committee on Nomenclature of the American Thyroid Association. Clin Chem 1991;37:2002-2008.
  387. Baloch Z, Carayon P, Conte-Devolx B, Demers LM, Feldt-Rasmussen U, Henry JF, LiVosli VA, Niccoli-Sire P, John R, Ruf J, Smyth PP, Spencer CA, Stockigt JR. Laboratory medicine practice guidelines. Laboratory support for the diagnosis and monitoring of thyroid disease. Thyroid 2003;13:3-126.
  388. Martino E, Bambini G, Bartalena L, Mammoli C, Aghini-Lombardi F, Baschieri L, Pinchera A. Human serum thyrotrophin measurement by ultrasensitive immunoradiometric assay as a first-line test in the evaluation of thyroid function. Clin Endocrinol (Oxf) 1986;24:141-148.
  389. Toft AD. Use of sensitive immunoradiometric assay for thyrotropin in clinical practice. Mayo Clin Proc 1988;63:1035-1042.
  390. Snyder PJ, Utiger RD. Response to thyrotropin releasing hormone (TRH) in normal man. J Clin Endocrinol Metab 1972;34:380-385
  391. Faglia G. The clinical impact of the thyrotropin-releasing hormone test. Thyroid 1998;8:903-908
  392. Sawin CT, Hershman JM, Chopra IJ. The comparative effect of T4 and T3 on the TSH response to TRH in young adult men. J Clin Endocrinol Metab 1977;44:273-278.
  393. Beck-Peccoz P, Persani L, Lania A. Thyrotropin-Secreting Pituitary Adenomas. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland HJ, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2018.
  394. Mehta A, Hindmarsh PC, Stanhope RG, Brain CE, Preece MA, Dattani MT. Is the thyrotropin-releasing hormone test necessary in the diagnosis of central hypothyroidism in children. J Clin Endocrinol Metab 2003;88:5696-5703
  395. Borst GC, Osburne RC, O'Brian JT, Georges LP, Burman KD. Fasting decreases thyrotropin responsiveness to thyrotropin-releasing hormone: a potential cause of misinterpretation of thyroid function tests in the critically ill. J Clin Endocrinol Metab 1983;57:380-383.
  396. Gow SM, Elder A, Caldwell G, Bell G, Seth J, Sweeting VM, Toft AD, Beckett GJ. An improved approach to thyroid function testing in patients with non-thyroidal illness. Clin Chim Acta 1986;158:49-58.
  397. Franklyn JA, Black EG, Betteridge J, Sheppard MC. Comparison of second and third generation methods for measurement of serum thyrotropin in patients with overt hyperthyroidism, patients receiving thyroxine therapy, and those with nonthyroidal illness. J Clin Endocrinol Metab 1994;78:1368-1371.
  398. Toft AD, Irvine WJ, Hunter WM, Ratcliffe JG, Seth J. Anomalous plasma TSH levels in patients developing hypothyroidism in the early months after 131I therapy for thyrotoxicosis. J Clin Endocrinol Metab 1974;39:607-609.
  399. Toft AD, Irvine WJ, McIntosh D, Seth J, Cameron EH, Lidgard GP. Temporary hypothyroidism after surgical treatment of thyrotoxicosis. Lancet 1976;2:817-818.
  400. Thein-Wai W, Larsen PR. Effects of weekly thyroxine administration on serum thyroxine, 3,5,3'-triiodothyronine, thyrotropin, and the thyrotropin response to thyrotropin-releasing hormone. J Clin Endocrinol Metab 1980;50:560-564.
  401. Vagenakis AG, Braverman LE, Azizi F, Portinay GI, Ingbar SH. Recovery of pituitary thyrotropic function after withdrawal of prolonged thyroid-suppression therapy. N Engl J Med 1975;293:681-684.
  402. Coya R, Carro E, Mallo F, Dieguez C. Retinoic acid inhibits in vivo thyroid-stimulating hormone secretion. Life Sci 1997;60:247-250
  403. Sherman SI, Gopal J, Haugen BR, Chiu AC, Whaley K, Nowlakha P, Duvic M. Central hypothyroidism associated with retinoid X receptor-selective ligands. N Engl J Med 1999;340:1075-1079
  404. Gow SM, Kellett HA, Seth J, Sweeting VM, Toft AD, Beckett GJ. Limitations of new thyroid function tests in pregnancy. Clin Chim Acta 1985;152:325-333.
  405. Emerson CH, Dysno WL, Utiger RD. Serum thyrotropin and thyroxine concentrations in patients recieving lithium carbonate. J Clin Endocrinol Metab 1973;36:338-346.
  406. Refetoff S, Sunthornthepvarakul T, Gottschalk ME, Hayashi Y. Resistance to thyrotropin and other abnormalities of the thyrotropin receptor. Recent Prog Horm Res 1996;51:97-120
  407. Abramowicz MJ, Duprez L, Parma J, Vassart G, Heinrichs C. Familial congenital hypothyroidism due to inactivating mutation of the thyrotropin receptor causing profound hypoplasia of the thyroid gland. J Clin Invest 1997;99:3018-3024
  408. Ahlbom BD, Yaqoob M, Larsson A, Ilicki A, Anneren G, Wadelius C. Genetic and linkage analysis of familial congenital hypothyroidism: exclusion of linkage to the TSH receptor gene. Hum Genet 1997;99:186-190
  409. Xie J, Pannain S, Pohlenz J, Weiss RE, Moltz K, Morlot M, Asteria C, Persani L, Beck-Peccoz P, Parma J, Vassart G, Refetoff S. Resistance to thyrotropin (TSH) in three families is not associated with mutations in the TSH receptor or TSH [see comments]. J Clin Endocrinol Metab 1997;82:3933-3940
  410. Boscato LM, Stuart MC. Heterophilic antibodies: a problem for all immunoassays. Clin Chem 1988;34:27-33.
  411. Despres N, Grant AM. Antibody interference in thyroid assays: a potential for clinical misinformation. Clin Chem 1998;44:440-454.
  412. Smith TP, Suliman AM, Fahie-Wilson MN, McKenna TJ. Gross variability in the detection of prolactin in sera containing big big prolactin (macroprolactin) by commercial immunoassays. J Clin Endocrinol Metab 2002;87:5410-5415
  413. Giusti M, Conte L, Repetto AM, Gay S, Marroni P, Mittica M, Mussap M. Detection of Polyethylene Glycol Thyrotropin (TSH) Precipitable Percentage (Macro-TSH) in Patients with a History of Thyroid Cancer. Endocrinol Metab (Seoul) 2017;32:460-465
  414. Hattori N, Aisaka K, Chihara K, Shimatsu A. Current Thyrotropin Immunoassays Recognize Macro-Thyrotropin Leading to Hyperthyrotropinemia in Females of Reproductive Age. Thyroid 2018;28:1252-1260
  415. Ohba K, Maekawa M, Iwahara K, Suzuki Y, Matsushita A, Sasaki S, Oki Y, Nakamura H. Abnormal thyroid hormone response to TRH in a case of macro-TSH and the cut-off value for screening cases of inappropriate TSH elevation. Endocr J 2020;67:125-130

 

 

Helminths And Endocrinology

ABSTRACT

Helminths are parasitic worms that can infect humans. They are broadly classified as flatworms (including Cestodes and Trematodes) and roundworms (nematodes). These worms infect organs such as intestines, liver, skin, as well as other tissues. These infections are more common in underdeveloped parts of the world affecting almost one-sixth of the world population. These infections can lead to a variety of endocrine manifestations. A decreased risk of developing type 2 diabetes mellitus has been observed in affected populations. Helminths modulate the host immunity towards a type 2 immune response which is anti-inflammatory in nature. An increase in T regulatory cells has also been seen which reduces T cell response to infections.  By virtue of these changes, chronic inflammation is suppressed in adipose tissues - this phenomenon may explain the protective effect in type 2 diabetes mellitus. A reduction in insulin resistance independent of BMI has been observed in animal as well as human studies. Hepatic lipid production can be reduced by the soluble egg antigen from certain schistosomes. The immunomodulatory effects of helminth infections can also protect against autoimmune endocrine conditions such as type 1 diabetes mellitus and Graves' disease. These observations may reflect the well-known "hygiene hypothesis". Thyroid nodules and hypothyroidism can occur in helminth infections. Insights into thyroid physiology, including thyroid hormone receptors and de-iodination pathways, have been obtained from studies in helminths. Certain helminth infections can impair osteoclast maturation suggesting potential implications for osteoporosis. Similarities between human and helminth bone morphogenetic protein pathways have been observed. Adrenal masses as well as adrenal insufficiency, have been observed in helminth infections. Infertility has frequently been reported with Schistosomiasis due to inflammation in the genital tracts. An estrogen like compound may be produced by schistosomes which can lead to hypogonadism in males. The helminth, Caenorhabditis elegans can serve as a model for studies on Kallman syndrome as the KAL-1 gene appears to be functionally conserved in this helminth.  A reduction in IGF-1 levels may be seen in adults infected with helminths. Apart from these manifestations, novel insights regarding endocrine disease mechanisms as well endocrine physiology can be derived from studies on helminths.

INTRODUCTION

The term helminth refers to parasitic worms which are broadly classified as flatworms (including Cestodes and Trematodes) and roundworms (nematodes) (1). These infections may be soil-transmitted and present as intestinal infections while others may invade different tissues such as blood vessels or other organs. These parasitic worms are endemic in several parts of the world, specifically in underdeveloped and parts of developing countries (1). It is estimated that approximately one-sixth of the world’s population is affected by helminth infections  (2). Helminths employ complex mechanisms to evade host immunity. They induce malnutrition in the host while simultaneously ensuring an adequate supply of nutrients for their own growth and reproduction.

Table 1. Classification of Helminths

PHYLUM

 

Affected Organ

Examples

Platyhelminths

Intestine

Cestodes -Taenia, Echinococcus

Liver

 Trematodes- Schistosoma, Fasciola

 

 Nematodes

Intestine

Ascaris, Enterobius, Necator, Ancylostoma, Trichuris, Strongyloides

Cutaneous

Strongyloides

Tissues

Onchocerca, Loa, Wuchereria

 

These immunological and metabolic interactions between helminth and the host may modulate the pathophysiology of several endocrine disorders including diabetes, thyroid disorders, and gonadal disorders apart from others. A discussion of each of these groups of disorders is presented below.

DIABETES MELLITUS

Diabetes mellitus is among the most common endocrine disorders with its rapidly growing prevalence earning it the designation of a pandemic. While type 1 diabetes mellitus is an autoimmune disease, type 2 diabetes is mediated by insulin resistance which is of multifactorial origin with genetics, environmental factors, and inflammation all playing their part. Among these factors, it has been noticed that areas with a high prevalence of soil-transmitted helminth infections have a relatively lower prevalence of diabetes (3). Although several other factors may also be operational in such areas, there are several proposed mechanisms by which helminth infections may influence diabetes and its pathogenesis.

Type 2 Diabetes Mellitus

Type 2 diabetes mellitus has been described as a chronic inflammatory disorder. Chronic inflammation in adipose tissues has been shown to be among the factors underlying this disease. The inflammatory process in adipose tissue involves infiltration by inflammatory cells such as lymphocytes, macrophages, and neutrophils.  Eosinophils on the other hand appear to have an anti-inflammatory effect. Apart from infiltration, several phenotypic changes occur in these cells which tip the balance towards inflammation. These include the predominance of T helper type 1 (Th1) and T helper type 17 (Th17) instead of the T helper type 2 (Th2) and the regulatory T cells (Tregs). The Th1 and Th17 cells promote the macrophage activation into classically activated macrophages (CAM) which in turn secrete inflammatory markers such as tumor necrosis factor-alpha (TNF-α), interleukin 6 and 12(IL6, IL12). TNF-α has been shown to interfere with insulin signaling. On the other hand, Th2 and T reg cells secrete IL-3 and IL 4 which stimulate the formation of alternatively activated macrophages (AAM) which are anti-inflammatory and express IL-10. Adipokines such as leptin, lipocalin 2, retinol-binding protein (RBP4), resistin, angiopoietin-like protein 2 (ANGPTL2), IL-6, IL-1, CC-chemokine ligand 2 (CCL2), CXC-chemokine ligand 5 (CXCL5) are also pro-inflammatory while adiponectin may have anti-inflammatory actions.

Helminth infections are associated with induction of type 2 immune response which involves increased activation of Th2 cells, eosinophilia, and production of IgE. The Th2 response in turn manifests as increased secretion of IL-4, IL-5, IL-9, IL-10, IL-13 which are anti-inflammatory. This also promotes the induction of anti-inflammatory AAMs.  Similarly, helminth infections are associated with an increase in T reg cells which mediate a state of T cell hypo-responsiveness. These changes on one hand limit the damage to host tissues by uncontrolled inflammation in response to helminth antigens and on the other hand prevent the clearance of the helminth from the host. The T cell hypo-responsiveness to parasite antigens can spill over to other antigens as well and this phenomenon has been invoked to explain the reduced prevalence of certain allergic and autoimmune disorders in helminth infected populations. Therefore, it has been hypothesized that since the immunological changes associated with helminth infections are anti-inflammatory in nature, they can reduce chronic inflammation in adipose and other tissues, thereby mitigating insulin resistance and the resultant type 2 diabetes.

The above hypothesis is supported by epidemiological data. In a study from China, previous schistosome infection was associated with a lower prevalence of obesity and metabolic syndrome as compared to those without such infection (4). Another study, which used ultrasonography to document chronic liver disease caused by schistosomiasis, found that metabolic syndrome prevalence was reduced to half of that seen in those without evidence of schistosomiasis (5). Serological evidence of chronic Strongyloides stercoralis infection was associated with a 61% lower risk of developing type 2 diabetes as compared to those who did not have this infection, despite adjustment for parameters such as age, BMI, and hypertension (6). In Indonesia, higher insulin sensitivity was demonstrated in patients who had infections with soil-transmitted helminths as evidenced by lower BMI and HOMA-IR levels (7). A randomized controlled trial to demonstrate the effect of ongoing helminth infection on insulin sensitivity has been conducted (8). This trial randomized households in an area endemic for helminth infection to receive albendazole or placebo over a period of time. In this trial, treatment of helminth infected subjects with albendazole lead to an increase in insulin resistance along with a reduction in IgE and eosinophil counts. However, at the community level, insulin resistance remained unchanged (9).

While the mechanisms of the reduction in type 2 diabetes have not been concretely studied in humans, animal studies provide support for the role of immunomodulation. Infections with Schistosoma mansoni, Nippostrongylus brasiliensis, and Litomosoides sigmodontis have been shown to increase eosinophils and AAMs in mice with diet-induced obesity (10–12). These animals had improved insulin sensitivity and glucose tolerance - this effect was lost in eosinophil deficient mice. S. mansoni soluble egg antigen and egg-derived ὠ-1 antigens stimulate innate lymphoid type 2 cells which produce IL-5 and IL-13 cytokines necessary for sustaining eosinophils and AAMs (13). L. sigmodontis Ag-treated obese mice had greater numbers of CD4+Foxp3+ Tregs in white adipose tissues as compared to controls indicating the upregulation of these cells as a mechanism of reducing insulin resistance (12).

Apart from these immunomodulatory mechanisms, the effect on body weight and gut microbiota are also potential mechanisms. The soluble egg antigen of S. japonicum, has been shown to reduce hepatic expression of microRNA 802 (miR802) which suppresses hepatic lipid production by upregulating the AMPK pathway. This has been proposed to be a future therapeutic target in obesity (14). However, the effect of helminth infection on insulin sensitivity has been shown to be independent of BMI in mice (10). This echoes the study on Australian aboriginals where the findings persisted despite adjusting for BMI (6).

Similarly, data on gut microbiota changes is scanty and conflicting. A few studies show an increase in gut bacterial diversity after helminth infection (15,16). Other authors have not found any significant changes in gut microbiota (17). While the mechanisms require further elucidation in both animal as well as human studies, there seems to be sufficient evidence to support the role of helminth infections in modulating the pathophysiology of type 2 diabetes.

Type 1 Diabetes Mellitus

Regarding type 1 diabetes, the type 2 immune response and suppressive regulatory environment induced by helminths may induce a protective effect. The incidence of type 1 diabetes has been increasing rapidly in developed countries and these regions are relatively less affected by helminth infections (18). The hygiene hypothesis has been invoked to explain this phenomenon (19). There are very few human studies which directly look at helminth infection and amelioration of type 1 diabetes risk. Enterobiasis did not reduce risk of type 1 diabetes in a population based study (19). Several animal studies do support the protective role of helminth infection. Axenic Caenorhabditis elegans antigen can protect against type 1 diabetes in the non-obese diabetes (NOD) mouse model (20). Trehalose produced by some helminths can alter intestinal microbiota leading to induction of CD8+ T cells which protect against type 1 diabetes in mice models (21). The severity of type 1 diabetes in mice models is ameliorated by recombinant Schistosoma japonicum cystatin and fructose-1,6-bisphosphate aldolase (22). Interestingly, children with schistosomiasis appear to have islet cell antibodies and defects in insulin secretion when compared to non infected siblings of children with insulin-dependent diabetes mellitus (23). Future studies may further shed light on this interesting topic.

THYROID DISORDERS

There appears to be a bidirectional relationship between the thyroid gland and helminth infections. On one hand, helminths appear to possess several proteins which are analogous to those involved in human thyroid physiology while on the other hand helminths can play a role in several thyroid diseases.

Thyroid hormone receptors which were earlier thought to be found only in chordates have been found to be present in S. mansoni (24). Two homologues of mammalian thyroid receptor (TR) has been isolated and characterized in S. mansoni (25), The thyroid hormone receptor beta from S. japonicum has also been characterized and evaluated as a vaccine candidate for this infection (26). Similarly, a nuclear hormone receptor has been identified in S. stercoralis which has some resemblance with steroid/thyroid hormone receptor found in humans (27). A transthyretin like protein has also been identified in Schistosoma dublin and Caenorhabditis elegans, although its function is unclear (28). Thyroid hormones may be essential for helminth growth and multiplication. In mice infected with Schistosoma mansoni, thyroid hormone therapy led to parasite multiplication and an increase in size whereas iodine deficient or thyroid hormone receptor knockout mice had lesser parasite numbers and smaller sized worms (29).

Some novel insights into thyroid physiology have also come from studies in helminths. Studies on the nematode Caenorhabditis have helped elucidate the mechanisms behind toxic effects of excess iodine - the dual oxidase maturation factor (DOXA-1) being among the implicated factors (30). Similarly, helminth studies have shown that iodotyrosine deiodinase may also have a role in regulating potassium channels in muscles (31).

Hypothyroidism and Thyroid Nodules

With respect to thyroid disorders, hypothyroidism and thyroid nodules appear to have some associations with helminth infections. S. stercoralis has been associated with hypothyroidism in one case report (32). Fasciola gigantica infection in buffaloes has been shown to lead to lymphocytic thyroiditis and hypothyroidism (33).

Hydatid cyst disease can mimic thyroid nodules and is often diagnosed by fine needle aspiration cytology (34). Cysticerosis may also present as a thyroid nodule (35). Similarly, microfilaria have also been found in fine needle aspirations from the thyroid (36). Schistosomiasis may interfere with technetium pertechnetate uptake in various tissues including the thyroid as demonstrated in mice- this may have implications for thyroid scan performed in infected humans (37).

Hyperthyroidism

Graves' disease, which is the most common cause of hyperthyroidism, may be affected by helminth infections. Considering that helminths affect host immune response and Graves' disease is an autoimmune process, such an association is not unexpected. Graves' disease is characterized by autoantibodies to the TSH receptor which leads to gland enlargement, hyperthyroidism, as well as extrathyroidal manifestations such as orbitopathy and dermopathy.  Animal models of Graves' disease have been developed which involve introduction of TSH receptor complementary DNA. It has been shown in such a mouse model that prior infection with S. mansoni may lead to a sustained Th2 type immune response towards the parasite egg antigens. This Th2 type immune response prevented the development of Graves' disease when mice were immunized with non-replicative recombinant adenovirus expressing the human TSHR. However, if given after disease onset, the Schistosoma infection could not cure the disease. Graves’ disease was once thought to be a Th2-type immune response, but recent studies have described a Th1-type as well as a Th2-type response suggesting that reversal of an activated immune response to the TSH receptor is not possible (38).  Based on similar findings with other infections, a hygiene hypothesis for Graves disease has also been proposed (39). 

BONES AND CALCIUM METABOLISM

There is limited information regarding calcium metabolism and bone health in helminth infections. Hydatid cyst disease involving the vertebrae has been described recently  and pathological hip fracture has also been reported (40,41). However, this is likely to represent direct involvement of the bone rather than alterations in bone metabolism.

Inflammatory arthritis is associated with secondary osteoporosis. The Th2 type immune response seen with helminth infections may attenuate inflammatory arthritis. N. brasiliensis was able to inhibit arthritis and bone loss in two experimental models of inflammatory arthritis (42). In vitro osteoclast differentiation has been shown to be inhibited by excretory/ secretory products from Heligmosomoides polygyrus bakeri, a murine helminth (43).  C-terminal sequence of Fasciola helminth defense molecule-1 (C-FhHDM-1) has been shown to reduce RANKL secretion as well as prevents both the formation of osteoclasts and acidification of lysosomes in animal models [6]. These features may be beneficial in osteoporotic states. However, in a study on pregnant mice, Heligmosomoides bakeri infection in late pregnancy and lactation led to a decrease in maternal bone mineral density and was associated with an increase in levels of inflammatory cytokines (IL-1 beta and IL-6) (44).

There may be several similarities between human and helminth physiology with respect to certain metabolic pathways. Homologues of  osteonectin, also called SPARC [Secreted Protein Acidic and Rich in Cysteine]), have been found in C. elegans while S. mansoni has homologues of TGF-beta receptor (45,46). More recently, it has been found that bone morphogenetic protein signaling may be conserved between humans and helminths, especially C. elegans. Secreted Modular Calcium-binding protein-1 (SMOC-1) gene identified in Caenorhabditis elegans may promote BMP signaling leading to the growth of the helminth (47,48). While BMP pathway plays several roles in human physiology including bone growth, the implications of these discoveries in helminths are still unclear.

 ADRENALS

Helminth infections in the adrenal with presentation as an adrenal mass have been reported in the literature.  Paragonimus westermani has been reported in a patient who had a lung cavity and an adrenal mass (49). Echinococcus multilocularis can infrequently present as a right adrenal mass detected incidentally (50). Adrenal schistosomiasis has also been reported (51). F. gigantica can cause adrenal insufficiency in animals (33). Acute adrenal insufficiency accompanied by adrenal hemorrhage has also been reported with S. stercoralis (52).

Activation of the hypothalamic-pituitary-adrenal axis leading to immunosuppression has been shown in mouse studies with Angiostrongylus cantonensis (53). While chronic immunosuppressive  therapy can lead to hyperinfection with helminths like S. stercoralis (54,55), adrenalectomized mice appeared to have higher worm burden and worm fecundity rates when infected with S mansoni (56). Previous mouse studies have shown adrenal hypertrophy and higher cortisol levels in S. mansoni infection (57). In vitro treatment of S. mansoni with adrenal hormones suggests that DHEA has a toxic effect with cercariae being more susceptible than schistosomula and adults (58).

GONADS

The manifestations of helminth infections with respect to gonads include hypogonadism and infertility. 

Hypogonadism

  1. mansoni infection has been associated with low normal testosterone and elevated estrogen levels in males although hepatic dysfunction may also play a role in these abnormalities (59). Patients infected with Loa loa and Mansonella perstans filariasis are more likely to have low testosterone and elevated gonadotropins as compared to control subjects (60). Further research in this area revealed that an estradiol-related compound was present in schistosome worm extracts (61). Later, the same authors confirmed the presence of this estradiol-related compound by mass spectrometry and also demonstrated that this compound has an antagonistic activity on the estrogen receptor and leads to a reduced expression of the estrogen receptor (62). Schistosome eggs can also convert estrogens to catechol-estrogens which in turn can be metabolized to active quinones. These quinones can cause DNA modifications and are implicated in the pathogenesis of malignancies related to schistosomiasis (63). These hormonal changes explain the pathogenesis of hypogonadism in schistosomiasis.

 Infertility

Schistosomiasis has been well recognized as a cause of infertility especially in females. Apart from hypogonadism caused by alterations in the estrogen axis, schistosomiasis can affect the genital tract leading to infertility. Genital involvement occurs in the form of granulomatous inflammation, fibrosis, and adhesion formation. The manifestations of this infection in females include tubal blockage, tubal pregnancy, tubal abortions, hemoperitoneum, preterm births, and miscarriages (64). Males can also have direct testicular inflammation along with, blockage of the genital ductal system and venous drainage leading to infarction. However, the involvement of male genital tract has been reported infrequently (65). Female infertility has also been reported with enterobiasis (66). Filarial involvement of male genitalia leading to hydrocele is well recognized (67). Male sterility can occur in such cases (68).

Some insights into genetic hypogonadism may come from helminth studies. The gene responsible for Kallman syndrome, KAL-1, appears to have a functionally conserved homologue in C. elegans. This gene plays a role in morphogenesis by influencing migration of epidermal cells in C. elegans. This discovery has established C. elegans as a model for study of Kallman syndrome for which a mouse model has proved to be elusive [33].

GROWTH

Children with helminth infections may have impaired growth- a phenomenon that can easily be attributed to malnutrition. However, helminth infections in adults are associated with significantly lower free IGF-1 which showed improvement after anti-helminth treatment(69). IGF-BP3 levels remain unchanged. Thus, direct effects on the GH-IGF-1 axis may occur in these infections.

CONCLUSION 

Table 2. Endocrine Manifestations of Helminth Infections

May protect against development of type 2 and type 1 diabetes mellitus

Hypothyroidism and thyroid nodules

May protect against Graves' disease

May protect against osteoporosis

Adrenal masses and acute adrenal insufficiency

Hypogonadism and infertility

Growth failure via reduction in IGF-1levels

 

In conclusion, helminths appear to play an important role in several endocrine disorders in endemic areas. Apart from contributing to the pathogenesis of disease, they may have a protective effect in some metabolic disorders. Novel insights regarding endocrine disease mechanisms as well endocrine physiology can be derived from studies using helminths. This is an interesting area for research which should encourage both helminthologists as well as endocrinologists.

REFERENCES

  1. Bethony J, Brooker S, Albonico M, Geiger S, Loukas A, D D, Pj H. Soil-transmitted helminth infections: ascariasis, trichuriasis, and hookworm. Lancet 2006;367(9521):1521–32.
  2. Pullan RL, Smith JL, Jasrasaria R, Brooker SJ. Global numbers of infection and disease burden of soil transmitted helminth infections in 2010. Parasit. Vectors 2014;7(1):37.
  3. de Ruiter K, Tahapary DL, Sartono E, Soewondo P, Supali T, Smit JWA, Yazdanbakhsh M. Helminths, hygiene hypothesis and type 2 diabetes. Parasite Immunol. 2017;39(5):e12404.
  4. Chen Y, Lu J, Huang Y, Wang T, Xu Y, Xu M, Li M, Wang W, Li D, Bi Y, Ning G. Association of Previous Schistosome Infection With Diabetes and Metabolic Syndrome: A Cross-Sectional Study in Rural China. J. Clin. Endocrinol. Metab. 2013;98(2):E283-7.
  5. Shen S-W, Lu Y, Li F, Shen Z-H, Xu M, Yao W-F, Feng Y-B, Yun J-T, Wang Y-P, Ling W, Qi H-J, Tong D-X. The potential long-term effect of previous schistosome infection reduces the risk of metabolic syndrome among Chinese men. Parasite Immunol. 2015;37(7):333–339.
  6. Hays R, Esterman A, Giacomin P, Loukas A, McDermott R. Does Strongyloides stercoralis infection protect against type 2 diabetes in humans? Evidence from Australian Aboriginal adults. Diabetes Res. Clin. Pract. 2015;107(3):355–361.
  7. Wiria AE, Hamid F, Wammes LJ, Prasetyani MA, Dekkers OM, May L, Kaisar MMM, Verweij JJ, Guigas B, Partono F, Sartono E, Supali T, Yazdanbakhsh M, Smit JWA. Infection with Soil-Transmitted Helminths Is Associated with Increased Insulin Sensitivity. PloS One 2015;10(6):e0127746.
  8. Tahapary DL, de Ruiter K, Martin I, van Lieshout L, Guigas B, Soewondo P, Djuardi Y, Wiria AE, Mayboroda OA, Houwing-Duistermaat JJ, Tasman H, Sartono E, Yazdanbakhsh M, Smit JWA, Supali T. Helminth infections and type 2 diabetes: a cluster-randomized placebo controlled SUGARSPIN trial in Nangapanda, Flores, Indonesia. BMC Infect. Dis. 2015;15(1):133.
  9. Tahapary DL, de Ruiter K, Martin I, Brienen EAT, van Lieshout L, Cobbaert CM, Soewondo P, Djuardi Y, Wiria AE, Houwing-Duistermaat JJ, Sartono E, Smit JWA, Yazdanbakhsh M, Supali T. Effect of Anthelmintic Treatment on Insulin Resistance: A Cluster-Randomized, Placebo-Controlled Trial in Indonesia. Clin. Infect. Dis. 2017;65(5):764–771.
  10. Hussaarts L, García-Tardón N, van Beek L, Heemskerk MM, Haeberlein S, van der Zon GC, Ozir-Fazalalikhan A, Berbée JFP, Willems van Dijk K, van Harmelen V, Yazdanbakhsh M, Guigas B. Chronic helminth infection and helminth-derived egg antigens promote adipose tissue M2 macrophages and improve insulin sensitivity in obese mice. FASEB J. 2015;29(7):3027–3039.
  11. Yang Z, Grinchuk V, Smith A, Qin B, Bohl JA, Sun R, Notari L, Zhang Z, Sesaki H, Urban JF, Shea-Donohue T, Zhao A. Parasitic nematode-induced modulation of body weight and associated metabolic dysfunction in mouse models of obesity. Infect. Immun. 2013;81(6):1905–1914.
  12. Berbudi A, Surendar J, Ajendra J, Gondorf F, Schmidt D, Neumann A-L, Wardani APF, Layland LE, Hoffmann LS, Pfeifer A, Hoerauf A, Hübner MP. Filarial Infection or Antigen Administration Improves Glucose Tolerance in Diet-Induced Obese Mice. J. Innate Immun. 2016;8(6):601–616.
  13. Hams E, Bermingham R, Wurlod FA, Hogan AE, O’Shea D, Preston RJ, Rodewald H-R, McKenzie ANJ, Fallon PG. The helminth T2 RNase ω1 promotes metabolic homeostasis in an IL-33- and group 2 innate lymphoid cell-dependent mechanism. FASEB J. 2016;30(2):824–835.
  14. Ni Y, Xu Z, Li C, Zhu Y, Liu R, Zhang F, Chang H, Li M, Sheng L, Li Z, Hou M, Chen L, You H, McManus DP, Hu W, Duan Y, Liu Y, Ji M. Therapeutic inhibition of miR-802 protects against obesity through AMPK-mediated regulation of hepatic lipid metabolism. Theranostics 2021;11(3):1079–1099.
  15. Lee SC, Tang MS, Lim YAL, Choy SH, Kurtz ZD, Cox LM, Gundra UM, Cho I, Bonneau R, Blaser MJ, Chua KH, Loke P. Helminth Colonization Is Associated with Increased Diversity of the Gut Microbiota. PLoS Negl. Trop. Dis. 2014;8(5):e2880.
  16. Kay G, Millard A, Sergeant M, Midzi N, R G, T M, A I, N N, F M, M P. Differences in the Faecal Microbiome in Schistosoma haematobium Infected Children vs. Uninfected Children. PLoS Negl. Trop. Dis. 2015;9(6):e0003861.
  17. Cooper P, Walker AW, Reyes J, Chico M, Salter SJ, Vaca M, Parkhill J. Patent Human Infections with the Whipworm, Trichuris trichiura, Are Not Associated with Alterations in the Faecal Microbiota. PLOS ONE 2013;8(10):e76573.
  18. Zaccone P, Hall SW. Helminth infection and type 1 diabetes. Rev. Diabet. Stud. RDS 2012;9(4):272–286.
  19. Bager P, Vinkel Hansen A, Wohlfahrt J, Melbye M. Helminth infection does not reduce risk for chronic inflammatory disease in a population-based cohort study. Gastroenterology 2012;142(1):55–62.
  20. Jackson-Thompson B, Torrero M, Mitre B, Long J, Packiam M, Mitre E. Axenic Caenorhabditis elegans antigen protects against development of type-1 diabetes in NOD mice. J. Transl. Autoimmun. 2020;3:100065.
  21. Shimokawa C, Kato T, Takeuchi T, Ohshima N, Furuki T, Ohtsu Y, Suzue K, Imai T, Obi S, Olia A, Izumi T, Sakurai M, Arakawa H, Ohno H, Hisaeda H. CD8+ regulatory T cells are critical in prevention of autoimmune-mediated diabetes. Nat. Commun. 2020;11(1):1922.
  22. Yan K, Wang B, Zhou H, Luo Q, Shen J, Xu Y, Zhong Z. Amelioration of type 1 diabetes by recombinant fructose-1,6-bisphosphate aldolase and cystatin derived from Schistosoma japonicum in a murine model. Parasitol. Res. 2020;119(1):203–14.
  23. Soliman AT, El-Nawawy AA, El-Azzouni OF, Amer EA, Demian SR, El-Sayed MH. High prevalence of islet cell antibody and defective insulin release in children with schistosomiasis. J. Trop. Pediatr. 1996;42(1):46–49.
  24. Wu W, Niles EG, LoVerde PT. Thyroid hormone receptor orthologues from invertebrate species with emphasis on Schistosoma mansoni. BMC Evol. Biol. 2007;7:150.
  25. Wu W, LoVerde PT. Nuclear hormone receptors in parasitic Platyhelminths. Mol. Biochem. Parasitol. 2019;233:111218.
  26. Qiu C, Liu S, Hong Y, Fu Z, Wei M, Ai D, Lin J. Molecular characterization of thyroid hormone receptor beta from Schistosoma japonicum and assessment of its potential as a vaccine candidate antigen against schistosomiasis in BALB/c mice. Parasit. Vectors 2012;5:172.
  27. Siddiqui AA, Stanley CS, Skelly PJ, Berk SL. A cDNA encoding a nuclear hormone receptor of the steroid/thyroid hormone-receptor superfamily from the human parasitic nematode Strongyloides stercoralis. Parasitol. Res. 2000;86(1):24–29.
  28. Richardson SJ, Hennebry SC, Smith BJ, Wright HM. Evolution of the thyroid hormone distributor protein transthyretin in microbes, C. elegans, and vertebrates. Ann. N. Y. Acad. Sci. 2005;1040:448–451.
  29. Saule P, Adriaenssens E, Delacre M, Chassande O, Bossu M, Auriault C, Wolowczuk I. Early variations of host thyroxine and interleukin-7 favor Schistosoma mansoni development. J. Parasitol. 2002;88(5):849–855.
  30. Zhaofa X, Jintao L, Yu L, Long M. The BLI-3/TSP-15/DOXA-1 dual oxidase complex is required for iodide toxicity in Caenorhabditis elegans. G3 Bethesda 2014;5(2):195–203.
  31. de la Cruz IP, Ma L, Horvitz HR. The Caenorhabditis elegans Iodotyrosine Deiodinase Ortholog SUP-18 Functions through a Conserved Channel SC-Box to Regulate the Muscle Two-Pore Domain Potassium Channel SUP-9. PLoS Genet. 2014;10(2):e1004175.
  32. Yue S, Min L. [Strongyloides stercoralis infection with hypothyroidism: one case report]. Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi 2017;29(3):393–394.
  33. Ganga G, Varshney JP, Sharma RL, Varshney VP, Kalicharan null. Effect of Fasciola gigantica infection on adrenal and thyroid glands of riverine buffaloes. Res. Vet. Sci. 2007;82(1):61–67.
  34. Dissanayake PI, Chennuri R, Tarjan G. Fine-needle aspiration diagnosis of primary hydatid disease of the thyroid; first reported case in the USA. Diagn. Cytopathol. 2016;44(4):334–337.
  35. Gupta S, Sodhani P. Clinically unsuspected thyroid involvement in cysticercosis: a case report. Acta Cytol. 2010;54(5 Suppl):853–856.
  36. Yenkeshwar PN, Kumbhalkar DT, Bobhate SK. Microfilariae in fine needle aspirates: a report of 22 cases. Indian J. Pathol. Microbiol. 2006;49(3):365–369.
  37. Góes VC, Neves RH, Arnóbio A, Bernardo-Filho M, Machado-Silva JR. Streptozotocin (STZ) and schistosomiasis mansoni change the biodistribution of radiopharmaceutical sodium (99m)Tc-pertechnetate in mice. Nucl. Med. Biol. 2016;43(9):581–586.
  38. Nagayama Y, Watanabe K, Niwa M, McLachlan SM, Rapoport B. Schistosoma mansoni and alpha-galactosylceramide: prophylactic effect of Th1 Immune suppression in a mouse model of Graves’ hyperthyroidism. J. Immunol. 2004;173(3):2167–2173.
  39. Nagayama Y, McLachlan SM, Rapoport B, Oishi K. Graves’ hyperthyroidism and the hygiene hypothesis in a mouse model. Endocrinology 2004;145(11):5075–5079.
  40. Meinel TR, Gottstein B, Geib V, Keel MJ, Biral R, Mohaupt M, Brügger J. Vertebral alveolar echinococcosis-a case report, systematic analysis, and review of the literature. Lancet Infect. Dis. 2018;18(3):e87–e98.
  41. Salman F, Khan MI, Hussain I, Abdullah HMA. Pathological fracture of femoral neck in a middle-aged woman: a rare presentation of primary hydatid cyst disease in humans. BMJ Case Rep. 2018;2018:bcr-2017-222980.
  42. Chen Z, Andreev D, Oeser K, Krljanac B, Hueber A, Kleyer A, Voehringer D, Schett G, Bozec A. Th2 and eosinophil responses suppress inflammatory arthritis. Nat. Commun. 2016;7:11596.
  43. Sarter K, Kulagin M, Schett G, Harris NL, Zaiss MM. Inflammatory arthritis and systemic bone loss are attenuated by gastrointestinal helminth parasites. Autoimmunity 2017;50(3):151–157.
  44. Odiere MR, Scott ME, Weiler HA, Koski KG. Protein deficiency and nematode infection during pregnancy and lactation reduce maternal bone mineralization and neonatal linear growth in mice. J. Nutr. 2010;140(9):1638–1645.
  45. Schwarzbauer JE, Spencer CS. The Caenorhabditis elegans homologue of the extracellular calcium binding protein SPARC/osteonectin affects nematode body morphology and mobility. Mol. Biol. Cell 1993;4(9):941–952.
  46. Beall MJ, Pearce EJ. Human transforming growth factor-beta activates a receptor serine/threonine kinase from the intravascular parasite Schistosoma mansoni. J. Biol. Chem. 2001;276(34):31613–31619.
  47. DeGroot MS, Shi H, Eastman A, McKillop AN, Liu J. The Caenorhabditis elegans SMOC-1 Protein Acts Cell Nonautonomously To Promote Bone Morphogenetic Protein Signaling. Genetics 2019;211(2):683–702.
  48. Schultz RD, Bennett EE, Ellis EA, Gumienny TL. Regulation of extracellular matrix organization by BMP signaling in Caenorhabditis elegans. PloS One 2014;9(7):e101929.
  49. Kwon YS, Lee HW, Kim HJ. Paragonimus westermani infection manifesting as a pulmonary cavity and adrenal gland mass: A case report. J. Infect. Chemother. Off. J. Jpn. Soc. Chemother. 2019;25(3):200–203.
  50. Spahn S, Helmchen B, Zingg U. Alveolar echinococcosis of the right adrenal gland: a case report and review of the literature. J. Med. Case Reports 2016;10(1):325.
  51. Poderoso WLS, Santana WB de, Costa EF da, Cipolotti R, Fakhouri R. Ectopic schistosomiasis: description of five cases involving skin, one ovarian case and one adrenal case. Rev. Soc. Bras. Med. Trop. 2008;41(6):668–671.
  52. Mutreja D, Sivasami K, Tewari V, Nandi B, Nair GL, Patil SD. A 36-year-old man with vomiting, pain abdomen, significant weight loss, hyponatremia, and hypoglycemia. Indian J. Pathol. Microbiol. 2015;58(4):500–505.
  53. Chen A-L, Sun X, Wang W, Liu J-F, Zeng X, Qiu J-F, Liu X-J, Wang Y. Activation of the hypothalamic-pituitary-adrenal (HPA) axis contributes to the immunosuppression of mice infected with Angiostrongylus cantonensis. J. Neuroinflammation 2016;13(1):266.
  54. Jones N, Cocchiarella A, Faris K, Heard SO. Pancreatitis associated with Strongyloides stercoralis infection in a patient chronically treated with corticosteroids. J. Intensive Care Med. 2010;25(3):172–174.
  55. Darlong J. Strongyloides hyper infection in a steroid dependent leprosy patient. Lepr. Rev. 2016;87(4):536–542.
  56. Morales-Montor J, Mohamed F, Damian RT. Schistosoma mansoni: the effect of adrenalectomy on the murine model. Microbes Infect. 2004;6(5):475–480.
  57. Antonios SN, Ismail HI, el-Nouby KA, Khalifa EA. Morphometric and histochemical study of the adrenal cortex in experimental schistosomiasis mansoni. J. Egypt. Soc. Parasitol. 2001;31(1):13–20.
  58. Morales-Montor J, Mohamed F, Ghaleb AM, Baig S, Hallal-Calleros C, Damian RT. In vitro effects of hypothalamic-pituitary-adrenal axis (HPA) hormones on Schistosoma mansoni. J. Parasitol. 2001;87(5):1132–1139.
  59. Saad AH, Abdelbaky A, Osman AM, Abdallah KF, Salem D. Possible role of Schistosoma mansoni infection in male hypogonadism. J. Egypt. Soc. Parasitol. 1999;29(2):307–323.
  60. Lansoud-Soukate J, Dupont A, De Reggi ML, Roelants GE, Capron A. Hypogonadism and ecdysteroid production in Loa loa and Mansonella perstans filariasis. Acta Trop. 1989;46(4):249–256.
  61. Botelho MC, Crespo M, Almeida A, Vieira P, Delgado ML, Araujo L, Machado JC, Correia da Costa JM. Schistosoma haematobium and Schistosomiasis mansoni: production of an estradiol-related compound detected by ELISA. Exp. Parasitol. 2009;122(3):250–253.
  62. Botelho MC, Soares R, Vale N, Ribeiro R, Camilo V, Almeida R, Medeiros R, Gomes P, Machado JC, Correia da Costa JM. Schistosoma haematobium: Identification of new estrogenic molecules with estradiol antagonistic activity and ability to inactivate estrogen receptor in mammalian cells. Exp. Parasitol. 2010;126(4):526–535.
  63. Cardoso R, Lacerda PC, Costa PP, Machado A, Carvalho A, Bordalo A, Fernandes R, Soares R, Richter J, Alves H, Botelho MC. Estrogen Metabolism-Associated CYP2D6 and IL6-174G/C Polymorphisms in Schistosoma haematobium Infection. Int. J. Mol. Sci. 2017;18(12):2560.
  64. Ribeiro AR, Luis C, Fernandes R, Botelho MC. Schistosomiasis and Infertility: What Do We Know? Trends Parasitol. 2019;35(12):964–971.
  65. Kini S, Dayoub N, Raja A, Pickering S, Thong J. Schistosomiasis-induced male infertility. Case Rep. 2009;2009:bcr0120091481.
  66. Dezsényi B, Sárközi L, Kaiser L, Tárkányi K, Nikolova R, Belics Z. Gynecological and obstetrical aspects of Enterobius vermicularis infection. Acta Microbiol. Immunol. Hung. 2018;65(4):459–465.
  67. Mand S, Debrah AY, Klarmann U, Mante S, Kwarteng A, Batsa L, Marfo-Debrekyei Y, Adjei O, Hoerauf A. The role of ultrasonography in the differentiation of the various types of filaricele due to bancroftian filariasis. Acta Trop. 2011;120 Suppl 1:S23-32.
  68. Panda DK, Mohapatra DP. Bancroftian filariasis associated with male sterility. BMJ Case Rep. 2018;2018:bcr-2017-223236.
  69. Kurniawan F, Tahapary DL, de Ruiter K, Yunir E, Biermasz NR, Smit JWA, Supali T, Sartono E, Yazdanbakhsh M, Soewondo P. Effect of anthelmintic treatment on serum free IGF-1 and IGFBP-3: a cluster-randomized-controlled trial in Indonesia. Sci. Rep. 2020;10(1):19023.

 

 

Endocrine Disruptor Chemicals

ABSTRACT

 

Endocrine Disrupting Chemicals (EDCs) impact health and disease. Scientific research conducted over the last few decades has solidified our knowledge of the health impacts of these chemicals. Intrauterine exposure of EDCs can have transgenerational effects, thus laying the foundation for disease in later life, when exposure may not be documentable. The meticulously orchestrated endocrine system is often a target for these chemicals. As the endocrine system is central to the body’s physiological and biological functions, EDCs can lead to perturbations in the functioning of an individual. Exposure to EDCs can occur right from children’s products to personal care products, food containers to pesticides and herbicides. Moreover, there are many unsuspected chemicals which may be contributing to the disease burden in the society, which have never been studied. The dose response relationship may not always be predictable for the different EDCs as even low-level exposures that may occur in everyday life can have significant effects in a susceptible individual. Although individual compounds have been studied in detail, the effects of a combination of these chemicals are yet to be studied in order to understand the real-life situation, where human beings are exposed to a cocktail of these EDCs. This chapter aims to summarize the available literature regarding these EDCs and their effects on endocrine physiology.

 

INTRODUCTION

 

Endocrine Disrupting Chemicals (EDCs) are a ubiquitous problem. This is a global issue and health hazard not well addressed due to lack of evidence and testing. Only a few EDCs are known and the others are suspected or yet to be explored (1). EDCs represent a broad class of natural or synthetic chemicals which are widely dispersed in the environment. This can be ingested or consumed or inhaled and may be found in larger quantities or trace amounts in serum, placenta, fat, umbilical cord blood etc. Exposure to EDCs can occur as early as in gestational period or childhood and can impact later stages of life. EDCs can alter normal physiological mechanisms in our body leading to a myriad of endocrinological problems both in children and adults.

 

The Endocrine Society defined EDC as “an exogenous chemical, or mixture of chemicals, that interfere with any aspect of hormone action.” In other words, the chemical substances that can affect the endocrine system resulting in adverse effects are called Endocrine Disruptor Chemicals (EDCs) (2). These chemicals often bind to the endogenous receptors (e.g.: estrogen receptor, steroid receptor) and interfere with the normal function of brain, reproductive organs, development, immune system, and other organs (3).

 

The common EDCs are bisphenol A (BPA), perchlorate, dioxins, phthalates, phytoestrogens, polychlorinated biphenyls (PCB), polybrominated diphenyl ethers (PBDE), triclosan, perfluoroalkyl and polyfluoroalkyl substances (PFAS), pesticides like dichlorodiphenyldichloroethylene (DDT) and its metabolite dichlorodiphenyldichloroethylene (DDE), organophosphorus compounds, alkylphenols(surfactants), parabans, methoxychlor,  diethylstilbestrol (DES), fungicide vinclozolin, and natural hormones (2) (4) (5). Among these, BPA is the most commonly encountered EDC, which has both estrogenic and antiandrogenic properties. EDCs are  mostly lipophilic in nature and resistant to metabolism (6). EDCs are usually present in food, beverages, pesticides, or air. People who get exposed to any of these EDCs may have hormonal imbalance. Even a small amount of EDC consumed can result in hormonal imbalance especially in children (2). Sometimes they are stored in body fats, and transferred to the developing fetus via the placenta (6).

 

Studies on animal models and humans reveal that the mechanisms through which the EDCs act involve divergent pathways. The EDC`s can act like endogenous hormones and thereby increase or decrease the cellular response. Also, they can block the effects of hormones and stimulate or inhibit the production of hormones. They can thus interfere with synthesis, transport, action, and degradation of hormones (7). EDCs can act via nuclear receptors, nonsteroidal receptors, transcription coactivators, and certain enzymatic pathways (5).

 

HISTORY OF EDCs

 

The effect of EDCs was first noticed by pig farmers in USA. Farmers observed pigs fed on moldy grain did not reproduce. Later it was found that moldy grain contained mycoestrogens. Several other incidents with such EDCs were noticed by farmers in other parts of the world. In 1940, diethylstilbestestrol (DES), a synthetic estrogen, was prescribed to women in their first trimester of pregnancy to prevent threatened miscarriage. Later in 1971, a rare vaginal cancer in daughters born to mothers who had taken DES was noted. All these events inspired Rachel Carlson to write a book named ‘Silent Spring’. In this book the author warned about long- term consequences of the use of pesticides and herbicides. In another book ‘Our Stolen Future ‘by Theo Colbron, Dianne Dumankosi, and John Peterson Meyers additional evidence on EDC was described. The hypothesis and evidence generated by this book was used for future research on EDC. This booked paved the path for the US regulators to create the United States Environment Protect Agency.

 

ARE HORMONES AND EDCs THE SAME?

 

EDCs are not the same as hormones but they can mimic hormones, and produce ill effects in the body.

 

Table 1. The Difference Between Hormone and Endocrine Disruptor Chemicals. (4)(8)

Hormones

EDCs

(1)  These are chemical substances produced by the body and transported via bloodstream to the cells and organs which carry receptors for the hormone and on which it has a specific regulatory effect.

(1) Exogenous substance that alters function(s) of the endocrine system and consequently causes adverse effects in an intact organism, or its progeny, or populations.

(2) They act via specific receptors and produces class effects

(2) They act via hormone and other receptors and produces abnormal functions and interactions.  

(3) No bio accumulation

(3) Results in bioaccumulation

(4)  Non-linear dose response with saturable kinetics

(4) Non-linear dose response with saturable kinetics

E.g.; steroid hormones, thyroid hormones

E.g.; Perchlorate, Dioxins, Phthalates

 

EDCs AND HUMAN HEALTH

 

EDCs can affect several systems in our body resulting in many ill health effects. There is evidence showing various diseases are linked to EDCs as shown in Table 2.

 

Table 2. Examples of EDCs and Their Possible Mechanisms Resulting in Clinical Conditions. (4)(9)(10)

 EDCs

Main Sources

Possible Mechanism

Clinical condition

Alkylphenols

Detergents Shampoos

Pesticide

Mimics estrogen

Breast cancer

Phthalates

Plastic products

Personal care products (perfume, moisturizer)

Not yet known

Testicular and ovarian toxicants

Polychlorinated biphenyls

(chlorinated/ halogenated/

TBBPA)

Paints

Plastics

Lubricants

Electrical applications

Estrogenic and anti-androgenic activity

Indirectly regulate circulating gonadal hormones.

Inducers of CYP1A and CYPIIB

Decreased NMDA receptor binding in striatum, frontal cortex and hippocampus, cerebellum 

Reduced glutamate and dopamine

Acts at AhR signaling pathways resulting in cytotoxic effects

 

Neurobehavioral defects like cognitive deficits in children

Neurotoxicity

Thyroid toxicity

Susceptibility to infections

Cancers (especially Breast Cancer)

Infertility

TBBPA- Tetrabromobisphenol A, CYP - cytochrome P450 enzymes, NMDA- N-methyl-D-aspartate, AhR - aryl hydrocarbon receptor

 

EFFECT OF EDCs ON ENDOCRINE SYSTEM

 

Neuro- Hypothalamic Effects

 

According to recent studies one in eight children 2-9 years of age suffer from neurodevelopmental disorders (NDDs) in India. NDDs include speech and language disorders, autism, cerebral palsy, epilepsy, vision impairment, ADHD, learning disorders, etc. EDCs are one among other risk factors associated with development of NDDs in children. NDD burden can be lessened by eliminating the causative factors or by preventing exposure to them. The major EDCs associated with NDDs are PCB and polybrominated diphenyl ethers (PBDEs).  Other EDCs that are linked to NDDs but lack firm evidence are brominated flame retardants, perfluorinated compounds, and pesticides. Animal studies reveal that EDCs can alter or affect neuronal development, synaptic organization, neurotransmitter synthesis and release, and structural development of the brain (11). Studies of pregnant women who lived near Lake Michigan, with high levels of exposure to PCBs, revealed that children of mothers with the highest exposure levels were much more likely to have lower average IQ levels and poorer performance on reading comprehension (12). BPA and phthalates have also been shown to be associated with behavioral problems in children, including anxiety and depression (13,14). Prenatal pesticide exposure has been linked to increased likelihood of children having autism spectrum disorder or developmental delay (15).

 

EDCs can cause perturbations of the neuroendocrine processes originating in the hypothalamus, and can also act on the steroid hormone receptors and other signaling pathways that occur widely throughout the brain. The critical period of exposure is important because even minor alterations in hormones can alter the neurobiological outcome during development. Our knowledge in this area is predominantly derived from animal studies as human studies (postmortem studies, accurate measurement of hypothalamic releasing hormones) are not feasible. Animal studies have shown the variable effects of BPA exposure on ER α and β protein and mRNA expression in different areas of the brain (16,17,18,19). Treatment of adult male and female rats for 4 days with low-dose BPA had significant effects on mRNAs for aromatase (increased in both sexes) and 5α-reductase 1 (decreased in females) in the prefrontal cortex (20). Although we know that developmental EDC exposure can alter the expression of genes and proteins for steroid hormone receptors, we cannot draw generalized conclusions from these animal models and future research should target especially this area of early EDC exposure.

 

EDC exposure can also have neuroendocrine effects. Animal studies have reported on the stimulatory as well as inhibitory actions of BPA on GnRH and kisspeptin systems (21,22). Studies on PCBs and phthalates have shown mixed results. Animal studies have shed some light on the effect of EDCs on the developing hypothalamic pituitary adrenal (HPA) axis. BPA exposure has been found to be associated with an increase in adrenal weight and an attenuated stress response (23). Basal corticosterone, as well as CRH- or ACTH-induced corticosterone release, has been found to be significantly suppressed in PCB exposed rats (24). These effects of EDCs on the HPA axis leading to aberrant stress response needs to be evaluated further in humans. Animal studies have opened up some new and interesting possibilities of EDC exposure with changes in AVP and oxytocin levels and social behavior (25,26).

 

Thyroid Function

 

EDCs can interfere with thyroid hormone synthesis, release, transport, metabolism and clearance.

 

Table 3.  EDCs Effect on Thyroid Function (27,28)

EDC

Source

Possible Outcome

Perchlorate

Oxidant in solid rocket propellants, fireworks, airbag deployment systems, etc.

Interferes with the uptake of iodide into the thyrocyte by sodium/iodide symporter (NIS)

Thiocyanates

Cigarettes

Interferes with the uptake of iodide

Isoflavones

(Phytoestrogens)

Soy protein

TPO inhibitors resulting in goiter in children

PCB

Paints

Plastics

 

They can act as TR agonist or antagonist, or reduce circulating levels of T4 resulting in relative hypothyroidism, increase in expression of glial fibrillar acidic protein leading to neurotoxicity in children.

BPA

 

Plastics

Food cans

Dental sealants

Binds to TRb and antagonizesT3 activation.

It can block T3-induced oligodendrocyte development from precursor cells, resulting in ADHD. Halogenated BPA can act as TR agonists, TBBPA bind to TR and induces GH3 cell proliferation and GH production.

 

 

PCB - Polychlorinated biphenyls, BPA - Bisphenol A, ADHD- Attention Deficit Hyperactivity Disorder, TBBPA - Tetrabromobisphenol A, T4 - tetraiodothyronine (thyroxine), T3 - triiodothyronine, TPO – Thyroperoxidase, TR – Thyroid Receptor, TH - Thyroid hormone, GH- Growth Hormone

 

Adipose Tissue and Metabolic Disorders

 

OBESITY

 

Obesity can result in the metabolic syndrome, reproductive problems, and cardiovascular risk factors. “Obesogens” are defined as “xenobiotic chemicals that can disrupt the normal developmental and homeostatic controls over adipogenesis and/or energy balance” (29). The “obesogen hypothesis” suggests that prenatal or early-life exposure to certain EDCs compounded by sedentary lifestyle and improper nutrition predisposes certain individuals to become obese later in life (30).  

 

In DES exposed mice an increase in body fat, leptin, adiponectin, interleukin (IL) - 6, triglyceride (TG) was observed. EDCs cause upregulation of gene expression involved in adipocyte differentiation and lipid metabolism resulting in fat accumulation (31). PPARg (peroxisome proliferator-activated receptors), a major regulator of adipogenesis, are expressed in adipocytes. It promotes adipocyte differentiation and the induction of lipogenic enzymes. During activation, PPARg along with retinoid X receptor (RXR), forms a heterodimer complex which then binds to PPAR response elements for regulation of fatty acids and repression of lipolysis. EDCs like tributyltin (TBT) and triphenyltin acts as PPARg and RXR agonists and increases adipose tissue mass.

 

Phytoestrogens mimic endogenous estrogens and exert various biological actions. They can bind to estrogen receptor (ER)a and estrogen receptor (ER)b and influence lipogenesis. One of the major sources of phytoestrogens is soy protein which contains genistein, a phytoestrogen. At low doses genistein inhibits lipogenesis whereas at high doses it can promote lipogenesis (27). EDCs like BPA, phthalates, dioxins perfluorinated compounds, and some pesticides are emerging as potential obeso­gens warranting further research.

 

Table 4. Potential Obesogenic Actions of EDCs

·      Agonist at PPARᵧ and RXRα (32)

·      Promotion of adipogenesis through ERs (33)

·      Increase in enzymatic activity of 11-β hydroxysteroid dehydrogenase type 1 (11-β HSD type 1) (34)

·      Increase in insulin stimulated lipogenesis (35)

·      Alterations in blood levels of insulin, leptin, and adiponectin (36)

·      Alteration of central energy regulatory pathways (37)

·      Decreased TRH expression and type 4 melanocortin receptors in the paraventricular nucleus of the hypothalamus and stimulation of orexigenic pathways (38)

·       Epigenetic transgenerational inheritance of adult-onset obesity (39)

 

DIABETES AND GLUCOSE HOMEOSTASIS

 

EDCs can disrupt glucose homeostasis in our body by affecting both insulin- and glucagon-secretory cells. Any toxic chemical that kills β cells or disrupts their function has been termed a “diabetogen”. The “diabetogen hypothesis” suggests that “every EDC circulating in plasma able to produce insulin resistance, independently of its obesogenic potential and its accumulation in adipocytes, may be considered a risk factor for metabolic syndrome and type 2 diabetes” (40). The obesogenic EDCs are risk factors for type 2 diabetes as well and lead to the dangerous combination of obesity and diabetes or “Diabesity”. However, certain EDCs may directly cause insulin resistance and defects in insulin production and secretion, without significantly affecting the weight of the individual. Studies have shown that acute treatment with BPA causes a temporary hyperinsulinemia, whereas longer-term exposure suppresses adiponectin release, and aggravates insulin resistance, obesity related syndromes, and development of diabetes mellitus. The hyperinsulinemia is attributed to the very rapid closure of ATP-sensitive K+ channels, potentiation of glucose-stimulated Ca2+ signals, and release of insulin via binding at extranuclear ER (41). Low doses of DES have been shown to impair the molecular signaling that regulates glucagon production through non genomic mechanism (27). POPs have been demonstrated to have direct effects on insulin signaling (42). They can lead to insulin resistance by causing adipose tissue inflammation. Heavy metals such as arsenic and mercury have also been considered as potential diabetogens. Intake of a high fat diet along with exposure to a cocktail of these EDCs (DEHP, BPA, PCB153, and TCDD) has been found to have sex specific alterations in the metabolic milieu in offspring. In males, there was alteration in the cholesterol metabolism whereas in females, there was pronounced effect on the glucose metabolism through a decrease in ER α expression and estrogen target genes (43).

 

The causal relationship between EDCs and type 1 diabetes is an area warranting research as animal studies have shown exposure to EDCs associated with insulitis (44).

 

Reproductive System

 

Over the past few decades there has been a surge in the incidence of reproductive system related disorders among both the males and females. EDCs can be attributed to this surge. Exposure to EDCs especially phytoestrogens have resulted in early menarche and polycystic ovarian diseases (PCODs) in adolescent girls. Infertility affects up to 15% of couples in the reproductive age group worldwide. The EDCs and their effect on reproductive system is summarized in Table 5.

 

Table 5. Effects of EDCs on the Reproductive System (27)(9)

EDC

Possible Mechanism

Possible Clinical Condition

Males

Females

Vinclozolin

Epigenetic (altered DNA methylation in germ cell lines)

AR antagonism

Hypospadias

Undescended testes

Delayed puberty

Prostate disease/cancer

Dysregulates the gland development Formation of

mammary tumor

DES

Increased ER expression in

Epididymis

Epigenetic silencing of

mRNA

Hypospadias Cryptorchidism Micropenis

Epididymal cysts

Vaginal adenocarcinoma

Ectopic pregnancy

Infertility

DDT/DDE

Antiandrogen

Antiprogestin

Induction of aromatase

Reduced insulin-like factor

 

Cryptorchidism

Infertility

 

Risk of breast cancer in females

Precocious and early puberty

Infertility

PCB

Estrogen agonist / Estrogen antagonist / antiandrogenic activity

Prostate cancer

Early onset of menarche

Delayed pubertal

development

Accumulates in breast adipose tissue

Phthalates

ER agonist/antagonist

Antiandrogen and decreases testosterone synthesis

 

Reduced anogenital distance and

Leydig cell function

HypospadiasCryptorchidism

Increased cell proliferation in

the breast

BPA

ER agonist

Antiandrogen

Inhibition of apoptotic activity in breast

Increased number of progesterone receptor positive

epithelial cells

Nongenomic activation of ERK1/2

Reduced sulfotransferase inactivation of estradiol

Prostate cancer

Testicular cancer in fetus

Altered breast development

Early puberty

Dioxins

ER agonist

Antiandrogen

Interfere with sex-steroid

synthesis

Inhibition of cyclooxygenase2 via AhR

Cryptorchidism

Premature thelarche

Endometriosis

Breast cancer

DES – Diethylstilbestrol, DDT – dichlorodiphenyldichloroethylene, DDE - dichlorodiphenyldichloroethylen, DNA – Deoxyribonucleicacid, AR – Androgen Receptor, ER – Estrogen Receptor, AhR - aryl hydrocarbon receptor, ERK1/2 - extracellular signal-regulated kinase 2, mRNA – messenger RNA

 

EFFECTS ON THE FEMALE REPRODUCTIVE SYSTEM

 

In vivo animal studies and thereafter in vitro studies indicate that exposure to BPA (1–30M) impairs meiotic progression in human fetal oocytes, increased levels of recombination, and induces epigenetic changes that may contribute to chromosome congression failure (45,46). Studies in rats have shown that neonatal BPA exposure decreased the numbers of all follicle types and increased atretic follicles during adulthood (47). In vitro animal studies have demonstrated the toxic effect of phthalates on the follicle growth and inhibition of estradiol production (48). Similar toxic effects of pesticides and environmental pollutants on gene expression, follicle growth and oocyte quality have been confirmed in animal studies. BPA and phthalates have also been implicated in altered steroidogenesis in the gonads (49). Findings of alteration in uterine structure and function after exposure to EDCs is more concerning as it may lead to abnormalities in implantation and recurrent abortions (50). BPA exposure has been associated with increased risk of implantation failure and miscarriages (51). Animal studies have also pointed towards the transgenerational effect of prenatal BPA exposure on female fertility (52). Experimental studies have shown an association between phthalate exposure and reduced fertility (53). The findings of these studies need to be confirmed in the human ovary to fully understand the impact of these EDCs on fertility and reproductive health as well as the transgenerational impact. EDCs have also been found to have adverse effects on menstrual cyclicity in women. Fungicide exposure has been associated with a significant decrease in bleeding (54). BPA and pesticides may accelerate ovarian failure and may lead to premature menopause in women (55). In utero exposure to DES increases the lifetime risk of premature menopause (56). Propyl paraben, a preservative in personal care product, was associated with lower antral follicle counts as well as higher day-3 FSH levels indicating accelerated ovarian aging (57). It may not be exposure to just a single EDC and more often than not it may be a cocktail of these that could lead to early reproductive senescence. In animal studies, late gestational exposure to DES causes ovarian hyperandrogenism and menstrual abnormalities similar to those in women with PCOS (58). A few epidemiological studies have pointed towards an association between phthalate exposure and risk of endometriosis, possibly due to increased viability of cells (59). 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure may disrupt cannabinoid signaling in the human endometrium and lead to increased inflammation in the endometrium (60). TCCD exposure can cause a progesterone-resistant phenotype that may persist over multiple generations, suggesting that TCDD exposure has transgenerational effects on endometriosis (61). TCDD increases the expression of thymus-expressed chemokine and promotes the invasiveness of endometrial stromal cells by increasing the expression of matrix metalloproteinase-2 and -9(62). TCDD also reduces the expression of CD82 (a wide-spectrum tumor metastasis suppressor that inhibits the mobility and invasiveness of cells), and increases the expression of CCL2-CCR2, which recruits macrophages and further down-regulates CD82 (63). Pesticides like fenvalerate stimulate the growth of uterine fibroid cells by enhancing cell cycle progression and inhibiting apoptosis through an ER-independent pathway (64). DES exposure has been shown to increase the occurrence of early onset fibroids in the Sister Study and Nurses’ Health Study II (65,66). Given the multiplicity of effects of EDCs on the female reproductive system, there remains an urgent need for future studies to confirm the findings of experimental and animal studies and understand the underlying mechanisms.

 

EFFECTS ON THE MALE REPRODUCTIVE SYSTEM

 

EDCs, by virtue of their antiandrogenic and estrogenic effects can have a profound influence on the male reproductive physiology. Studies on the causative effect of EDCs on hypospadias have not given consistent results due to the small number of subjects studied. Levels of chlorinated pesticides have been found to be higher in breast milk of mothers with cryptorchid boys (77). Studies on the incidence of cryptorchidism with xenoestrogen exposure showed detectable levels of lindane and mirex in placenta with higher cryptorchidism risk (78). Higher dioxin levels in breast milk and dibutyltin concentrations in placenta were associated with cryptorchidism in Danish boys (79). Dioxins may have estrogenic effects through interaction of the dioxin-AhR nuclear translocator complex with estrogen receptor. High exposure to DDE and PCBs also has a higher risk of cryptorchidism (80). Environmental factors play an important role in the development of testicular cancers. Cryptorchidism and hypospadias are well-characterized risk factors of testicular germ cell cancers (TGCC). Although TGCC is probably a condition with fetal origins, it has been practically difficult to prove the association between pre and postnatal exposure of EDCs and TGCC, given the long lag time between exposure and effect. A positive association of TGCC with DDE (81) and chlordane exposure (cis-nonachlor and trans-nonachlor) has been found (82). Intra uterine exposure to EDCs that affect the spermatogonial stem cells or Sertoli cells, can cause irreversible changes that result in permanently low adult sperm number. PCB exposure may affect sperm DNA integrity and motility (83). DDE exposure has been inversely associated with sperm motility and total sperm count (84), and positively correlated with defects in sperm chromatin condensation and morphology (85). Fetal and perinatal exposure via breast-feeding to dioxin in the Seveso accident was associated with reductions in sperm concentration, number of motile sperm, and total sperm number (86). PBDEs used as flame retardants have been found to negatively affect sperm concentration, testicular size and sperm motility (87). The major impediment to establishing a causal role of these effects of EDCs is the long lag time between the critical exposure and the manifestation of the adverse outcomes.

 

EFFECTS ON PUBERTY  

 

There has been a decrease in the age of breast development but the age of menarche has not changed significantly. This finding alerted researchers about the possible interfering role of EDCs in pubertal mechanisms. Results of epidemiological studies have been equivocal on the effects of BPA and phthalates on pubertal onset (67,68). Studies have pointed towards higher kisspeptin levels in girls exposed to phthalates, which may promote precocious puberty (69). There have been inconsistencies between animal and human studies and hence, inconclusive data on the effects of other EDCs like pesticides and environmental contaminants on puberty. Apparently innocuous substances like lavender oil and tea tree oil present in lotions and creams can lead to prepubertal gynecomastia by their estrogenic effects (70). A very interesting hypothesis has been put forward to explain the role of EDCs in precocious puberty seen in immigrant girls from developing countries. Early and temporary exposure to weakly estrogenic dichlorodiphenyltrichloroethane (DDT) in developing countries, where the exposure is still high, could stimulate hypothalamic maturation while the pituitary gonadotrophins are inhibited via a negative feedback that prevents manifestation of central maturation. This negative feedback disappears after withdrawal from the exposure, as happens when the child migrates to a different environment. This could precipitate precocious puberty in these migrant children (71). High exposure to endosulfan has been shown to be associated with pubertal delay, due to its antisteroidogenic properties (72). Dioxins act through aryl hydrocarbon receptors and thereby interact with other nuclear receptors. Exposure in boys has been associated with delayed puberty and in girls with delayed thelarche due to its antiestrogenic effects (73,74). Lead exposure has been implicated in delayed puberty in both boys and girls (75,76). Endocrine disrupters may alter the levels of endogenous hormones and their ratios by influencing their production, secretion, binding to carriers, metabolism and excretion. When studying these compounds, one needs to keep in mind about their active metabolites and the multiplicity of effects on the complex endocrine milieu.  

 

Hormone Responsive Cancer

 

Most cancer occur due to genetic predisposition or exposure to environmental or occupational hazards. EDCs can alter the genes and result in uncontrolled proliferation of cells. Almost all the EDCs identified are known to cause cancer. People working in certain industries like coal, steel, rubber, textile, paper manufacturing, paint are at higher risk of developing cancers due to increased exposure to these EDCs. Studies have shown that early exposure to these EDCs BPA, PCBs, perflourinated compounds, phthalates, and some pesticides can increase cancer risk(1).Several EDCs that mimic endogenous estrogens are potential carcinogens. The estrogen-responsive cancers including breast, endometrial, ovarian, and prostate cancers are caused due to several chemical xenoestrogens and phytoestrogens (88). EDC exposure during the critical periods of mammary gland development like gestation, puberty, and pregnancy may predispose to carcinogenesis. Dioxin exposure, especially TCDD has been found to increase the incidence of breast cancer (89). Inconsistent results have been obtained with regards to pesticide exposure and breast cancer risk, possibly due to individual chemicals studied whereas in real life, humans are often exposed to a mixture of them. Breast cancer patients present more frequently with a combination of aldrin, DDE, and DDD, and this mixture has not been found in healthy women (90). Exposure to diethyl phthalate, the parent may be associated with a 2-fold increase in breast cancer risk (91). EDCs may influence other estrogen dependent cancers as well. In women previously exposed to chlorotriazine herbicides, there was a significant 2.7-fold increased risk for ovarian neoplasms (92). Higher PFOA levels are associated with ovarian cancer (93). In males, those EDCs that can interfere with androgen and estrogen signaling pathways can increase the risk of prostate cancer. A classic example of developmental exposure and onset of latent disease is the progeny of mothers exposed to DES during pregnancy. Although prostatic structural abnormalities have been documented in this cohort (94), the exact effect on prostate cancer is yet to be ascertained as the cohort is still being followed up. Pesticide exposure and carcinogenesis has garnered much interest after the Agricultural Health Study (AHS) in the United States. Specific organophosphate insecticides like fonofos, malathion, terbufos, and aldrin) have been associated with increased risk of aggressive prostate cancer (95). Certain organophosphates like coumaphos and organochlorine (aldrin) pesticides increase prostate cancer risk in men with a family history of the disease (96). Compounds like chlorpyrifos, coumaphos, fonofos, and phorate strongly inhibit the hepatic CYP1A2 and CYP3A4 enzymes that metabolize testosterone, estradiol, and estrone (97) and thereby act as EDCs apart from causing DNA damage by oxidative stress. TCDD, the most toxin dioxin in the Agent Orange herbicide spray has been found to have a strong positive association in the incidence and aggressiveness of prostate cancer in the Vietnam veterans (98). Trace elements like arsenic and cadmium, have been classified as EDCs due to their ability to act as a ligand and/or interact with members of the steroid receptor superfamily and have been implicated in prostate cancer although more conclusive studies are needed.

 

Effect on The Adrenals

 

The adrenal gland is probably one of the most ignored glands in toxicology, despite it being very sensitive to toxins. By virtue of its intense vascularity, its capacity for uptake and storage of lipophilic agents and high local concentrations of enzymes of CYP family with potential for bioactivation of toxins, the adrenals are very susceptible to the toxic effects of EDCs. The results of toxicological research on adrenals may not always be straightforward because of the dynamic nature of the HPA axis. Thus, even in the face of compromised adrenal steroidogenesis, it is not surprising to find relatively normal levels of circulating cortisol, albeit with an increased ACTH drive. Hence, scientists studying the toxic effect of EDCs on the adrenals, need to take into account the ACTH and cortisol levels as well as the adrenal weight. One of the earliest evidences for an adrenal disruptor was the use of the anesthetic, etomidate, which inhibits CYP11B1, leading to adrenal insufficiency. Another direct inhibitor of adrenal steroidogenic enzymes is a derivative of the pesticide DDD, mitotane (o,p’-DDD), which is used to treat Cushing’s syndrome. Polychlorinated biphenyl 126 (PCB126) causes an increase in aldosterone biosynthesis by increasing expression of CYP11B2, the enzyme which catalyzes the final step of aldosterone biosynthesis. High concentrations PCB126 has been shown to increase expression of the Angiotensin 1 (AT1) receptor, enhancing angiotensin II responsiveness of adrenal cells. Lead has also been reported to increase aldosterone synthesis by a mechanism consistent with upregulation of CYP11B2. It has also been reported that a class of herbicides (2-chloro-s-triazine herbicides) increase the expression of CYP19, which encodes aromatase, raising the possibility of increased adrenal estrogen secretion (99). The lack of a clear understanding of the adrenal toxicology can be overcome by the use of sophisticated endocrine studies, which take into account the dynamicity of the HPA axis.

 

EFFECT OF EDCs DURING PREGNANCY

 

Studies on animals have shown that EDCs can affect germ cell lines. In a cohort study of 47,540 women with history of exposure to diethylstilbestrol (DES) during pregnancy and ADHD diagnosis were followed up to three generations (F0, F1, F2) to know consequences of exposure to DES. This study revealed that the progeny of mothers who used DES in the 1st trimester of pregnancy had higher risk of developing ADHD. BPA is another EDC which can lead to neuroendocrinal problems (100). This highlights the ill effects of EDCs in vertical transmission. EDCs like perfluorooctanoic acid have been implicated in pregnancy induced hypertension. There have been some pointers towards an association between BPA and preterm birth but it has not been conclusively proven in experimental animal studies (101).

 

Phthalate exposure during pregnancy may be associated with increased odds of prematurity (102). The possible mechanisms are interference with the placental function via effects on trophoblast differentiation and placental steroidogenesis which could increase the risk of preterm birth. Similar genetic effects of pesticides have also been shown to result in increased prematurity and preterm birth. This risk has been shown to be magnified in those with certain genetic mutations, highlighting the gene- environment interaction (103). Environmental contaminants like TCCD exert pro-inflammatory effects on the placenta, leading to infection-mediated preterm birth (104). EDCs have also been implicated in adverse birth outcomes. In the Generation R study in The Netherlands, prenatal BPA exposure was associated with reduced fetal weight and head circumference (105). The same study also showed that maternal phthalate exposure was associated with an increased time-to-pregnancy (106) and impaired fetal growth during pregnancy and decreased placental weight (107). In a similar Japanese study, maternal urinary MEHP levels were negatively associated with anogenital distance (AGD) in male offspring (108). Pesticide exposure during the second trimester of pregnancy have been negatively associated with birth weight, birth length, and head circumference as shown in the data from Center for Health Assessment of Mothers and Children of Salinas (CHAMACOS) (109). Increased incidence of infants being born as small for gestational age has also been reported in mothers who were exposed to pesticides (110). A sex dependent nature of these adverse birth outcomes has been demonstrated in a Chinese study with a decrease in gestational duration in girls but not boys (111). Similarly, in the Hokkaido Study on Environment and Children’s Health, an ongoing cohort study in Japan, PCDF and PCDD exposures were negatively associated with birth weight and infant development, with males being more susceptible than females (112). However, not all studies are shown these consistent adverse effects of EDCs. Hence future studies should confirm these preliminary findings and also study certain EDCs which have never been studied so far in experimental and epidemiological studies.

 

DETECTION OF EDCs

 

EDCs may be in complex forms or in trace amounts in biological fluids or environment which makes it difficult to identify or detect them. The methods used for the detection of these compounds should be highly sensitive and specific. These include liquid chromatography, gas chromatography and capillary electrophoresis. The bioassay techniques (Receptor binding assay, Receptor gene assay, DNA binding assay) are either qualitative or quantitative and can be helpful to know the biological effects of the complex samples. Due to the complexities and trace amount of the EDC, preconcentration is required (5). However, the limitations in sensitivity, reproductivity, difficulty in separation, and affordability still remain.

 

 

Newer methods are being explored to predict the effect of chemical disruptors using artificial intelligence (AI).  Combining artificial neural network (ANN) and chemical similarity approaches, a significant role of AI in chemical endocrine receptor disruption prediction has been demonstrated. For example, isoflavone genistein, a phytoestrogenfrom soy was found to be active or disruptive whereas isoflavone daidzein from the soy was predicted to be inactive or non-disruptive (113).  ANN can be used to predict chemical activity against estrogen and androgen receptors. Machine learning and ANN can more accurately and precisely predict EDCs in future.

 

Biosensors are newer devices which can detect chemicals up to femtomolar limit of detection.  Aptasensors, Nanotubes, Molecularly imprinted polymer (MIP)-based sensors are the emerging EDC detectors (114). Recently a device called ‘Tethys’ has been invented to detect presence of lead in water. Lead is known to affect the hormone signaling and central nervous system. This device works on the basis of nanocarbon tubes and could send water quality information via Bluetooth (115).

 

Among several computer aided approaches,  invitro and in silico predictions are now used to predict large number of chemical disruptors in the environment (116). Also the ligand-based models, like QSAR models which can predict biological activities of EDC and structure-based models can be combined with Artificial Intelligence technology for more accurate EDC predictions (117).

 

EDCs IN THE TROPICS

 

Pesticide use has increased over the years due to intensification of agricultural practices in the tropical countries. While the developed countries do have a well-established legal framework for pesticide environmental risk assessments, such requirements are either not available or inadequately implemented in tropical countries. Added to these woes are the fact that cheap compounds that are environmentally persistent and highly toxic, banned from agriculture use in developed countries, still remain popular in developing countries (118). These may lead to soil and water contamination with pesticide residues. The effect of these compounds on the applicators as well as the consumers are manyfold. In a multi-centric study to assess the pesticide residues in selected food commodities (Surveillance of Food Contaminants in India, 1993), DDT residues were found in about 82% of the 2205 samples of bovine milk. Data on 186 samples of 20 commercial brands of infants’ formulae showed the presence of residues of DDT and Hexachlorocyclohexane (HCH) isomers in about 70 and 94% of the samples with their maximum level of 4.3 and 5.7 mg/kg respectively (119). The average daily intake of HCH and DDT by Indians was reported to be 115 and 48 mg per person respectively, which were higher than those observed in most of the developed countries (120). Over these continuous levels of exposure through food, water and soil are the occasional spillovers and accidents that lead to greater exposure. Although these exposures have been documented well in literature, there are sparse studies from the tropical areas on the long-term effects, especially in relation to the endocrine system. Although there are compelling social and economic benefits for the rampant use of EDCs, the policymakers need to be made aware of the long term and sometimes transgenerational effects of these molecules.

 

CONCLUSION

 

EDCs are an emerging global health problem that requires urgent attention and action. The most common EDCs that we encounter in our day-to-day life are BPA, PCBs, paraben etc. This results in endocrinological problems in all the age groups. There is an urgent need of novel biomarkers, detectors or assays using novel technologies for the early detection of EDCs. The novel technologies like Artificial Intelligence, OMICS (Genomics, Epigenomics, Mitochondriomics) and Nano technology are the new-way forward in this regard. Food and Health authorities play a vital role in curbing this problem. Food and safety laws should be more stringent and higher throughput screening for EDCs should be done prior to approval of any products. BPA free, paraben free products should be encouraged. Industrialists and others manufacturers must make sure not to pollute the water with the industrial wastes. All these measures will help in eliminating EDCs related health problems.

 

REFERENCES

 

  1. Gore AC, Crews D, Doan LL, Merrill ML, Patisaul H, Zota A. INTRODUCTION TO ENDOCRINE DISRUPTING CHEMICALS (EDCs). :76.
  2. Endocrine Disruptors [Internet]. National Institute of Environmental Health Sciences. [cited 2021 Feb 7]. Available from: https://www.niehs.nih.gov/health/topics/agents/endocrine/index.cfm
  3. Research NC for T. Endocrine Disruptor Knowledge Base [Internet]. FDA. FDA; 2019 [cited 2021 Feb 7]. Available from: https://www.fda.gov/science-research/bioinformatics-tools/endocrine-disruptor-knowledge-base
  4. WHO | Global assessment of the state-of-the-science of endocrine disruptors [Internet]. WHO. World Health Organization; [cited 2021 Feb 8]. Available from: https://www.who.int/ipcs/publications/new_issues/endocrine_disruptors/en/
  5. Jones L, Regan F. Endocrine Disrupting Chemicals. In: Reference Module in Chemistry, Molecular Sciences and Chemical Engineering [Internet]. Elsevier; 2018 [cited 2021 Feb 7]. p. B9780124095472144000. Available from: https://linkinghub.elsevier.com/retrieve/pii/B9780124095472145123
  6. Elobeid MA, Allison DB. Putative environmental-endocrine disruptors and obesity: a review. Curr Opin Endocrinol Diabetes Obes. 2008 Oct;15(5):403–8.
  7. Schneider M, Pons J-L, Labesse G, Bourguet W. In Silico Predictions of Endocrine Disruptors Properties. Endocrinology. 2019 Nov 1;160(11):2709–16.
  8. Bergman Å, United Nations Environment Programme, World Health Organization. State of the science of endocrine disrupting chemicals - 2012 an assessment of the state of the science of endocrine disruptors [Internet]. Geneva: WHO : UNEP; 2013 [cited 2021 Feb 7]. Available from: http://www.who.int/ceh/publications/endocrine/en/index.html
  9. Lucaccioni L, Trevisani V, Marrozzini L, Bertoncelli N, Predieri B, Lugli L, et al. Endocrine-Disrupting Chemicals and Their Effects during Female Puberty: A Review of Current Evidence. Int J Mol Sci. 2020 Mar 18;21(6):2078.
  10. Bell MR. Endocrine-disrupting actions of PCBs on brain development and social and reproductive behaviors. Curr Opin Pharmacol. 2014 Dec;19:134–44.
  11. Arora NK, Nair MKC, Gulati S, Deshmukh V, Mohapatra A, Mishra D, et al. Neurodevelopmental disorders in children aged 2–9 years: Population-based burden estimates across five regions in India. PLOS Med. 2018 Jul 24;15(7):e1002615.
  12. Jacobson JL, Jacobson SW. Intellectual impairment in children exposed to polychlorinated biphenyls in utero. N Engl J Med. 1996;335:783–789.
  13. Harley KG, Gunier RB, Kogut K, et al. Prenatal and early childhood bisphenol A concentrations and behavior in school-aged children. Environ Res. 2013;126:43–50.
  14. Braun JM, Kalkbrenner AE, Calafat AM, et al. Impact of early-life bisphenol A exposure on behavior and executive function in children. Pediatrics. 2011;128:873–882.
  15. Shelton JF, Geraghty EM, Tancredi DJ, et al. Neurodevelopmental disorders and prenatal residential proximity to agricultural pesticides: the CHARGE Study. Environ Health Perspect. 2014;122:1103–1109.
  16. Chen F, Zhou L, Bai Y, Zhou R, Chen L. Sex differences in the adult HPA axis and affective behaviors are altered by perinatal exposure to a low dose of bisphenol A. Brain Res. 2014;1571:12–24.
  17. Cao J, Rebuli ME, Rogers J, et al. Prenatal bisphenol A exposure alters sex-specific estrogen receptor expression in the neonatal rat hypothalamus and amygdala. Toxicol Sci. 2013;133:157–173.
  18. Monje L, Varayoud J, Muñoz-de-Toro M, Luque EH, Ramos JG. Exposure of neonatal female rats to bisphenol A disrupts hypothalamic LHRH pre-mRNA processing and estrogen receptor expression in nuclei controlling estrous cyclicity. Reprod Toxicol. 2010;30:625–634.
  19. Yu B, Chen QF, Liu ZP, et al. Estrogen receptor and expressions in hypothalamus-pituitary-ovary axis in rats exposed lactationally to soy isoflavones and bisphenol A. Biomed Environ Sci. 2010;23:357–362.
  20. Castro B, Sánchez P, Torres JM, Preda O, del Moral RG, Ortega E. Bisphenol A exposure during adulthood alters expression of aromatase and 5 alpha-reductase isozymes in rat prostate. PLoS One. 2013;8:e55905.
  21. Bai Y, Chang F, Zhou R, et al. Increase of anteroventral periventricular kisspeptin neurons and generation of E2- induced LH-surge system in male rats exposed perinatally to environmental dose of bisphenol-A. Endocrinology.2011;152:1562–1571.
  22. Patisaul HB, Todd KL, Mickens JA, Adewale HB. Impact of neonatal exposure to the ER agonist CD-1, bisphenol- A or phytoestrogens on hypothalamic kisspeptin fiber density in male and female rats. Neurotoxicology.2009;30:350–357.
  23. Panagiotidou E, Zerva S, Mitsiou DJ, AlexisMN,Kitraki E. Perinatal exposure to low-dose bisphenol A affects the neuroendocrine stress response in rats. J Endocrinol.2014;220:207–218.
  24. Meserve LA, Murray BA, Landis JA. Influence of maternal ingestion of Aroclor 1254 (PCB) or FireMaster BP-6 (PBB) on unstimulated and stimulated corticosterone levels in young rats. Bull Environ Contam Toxicol. 1992;48:715–720.
  25. Wolstenholme JT, Taylor JA, Shetty SR, Edwards M, Connelly JJ, Rissman EF. Gestational exposure to low dose bisphenol A alters social behavior in juvenile mice. PLoS One. 2011;6:e25448.
  26. Sullivan AW, Beach EC, Stetzik LA, et al. A novel model for neuroendocrine toxicology: neurobehavioral effects of BPA exposure in a prosocial species, the prairie vole (Microtus ochrogaster). Endocrinology.2014;155:3867–3881.
  27. Diamanti-Kandarakis E, Bourguignon J-P, Giudice LC, Hauser R, Prins GS, Soto AM, et al. Endocrine-Disrupting Chemicals: An Endocrine Society Scientific Statement. Endocr Rev. 2009 Jun 1;30(4):293–342.
  28. Lauretta R, Sansone A, Sansone M, Romanelli F, Appetecchia M. Endocrine Disrupting Chemicals: Effects on Endocrine Glands. Front Endocrinol. 2019 Mar 21;10:178.
  29. Grün F, Blumberg B. Environmental obesogens: organotins and endocrine disruption via nuclear receptor signaling. Endocrinology. 2006;147:S50–S55.
  30. Grün F, Watanabe H, Zamanian Z, et al. Endocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebrates. Mol Endocrinol. 2006;20:2141–2155.
  31. Newbold RR, Padilla-Banks E, Snyder RJ, Phillips TM, Jefferson WN. Developmental exposure to endocrine disruptors and the obesity epidemic. Reprod Toxicol Elmsford N. 2007 May;23(3):290–6.
  32. Kanayama T, Kobayashi N, Mamiya S, Nakanishi T, Nishikaw J. Organotin compounds promote adipocyte differentiation as agonists of the peroxisome proliferator activated receptor /retinoid X receptor pathway. Mol Pharmacol. 2005;67:766–774.
  33. Masuno H, Iwanami J, Kidani T, Sakayama K, Honda K. Bisphenol A accelerates terminal differentiation of 3T3–L1 cells into adipocytes through the phosphatidylinositol 3-kinase pathway. Toxicol Sci. 2005;84:319–327.
  34. Wang J, Sun B, Hou M, Pan X, Li X. The environmental obesogen bisphenol A promotes adipogenesis by increasing the amount of 11ẞ-hydroxysteroid dehydrogenase type 1 in the adipose tissue of children. Int J Obes (Lond).2013;37:999–1005.
  35. Neel BA, Brady MJ, SargisRM.The endocrine disrupting chemical tolylfluanid alters adipocyte metabolism via glucocorticoid receptor activation. Mol Endocrinol. 2013;27:394–406.
  36. Angle BM, Do RP, Ponzi D, et al. Metabolic disruption in male mice due to fetal exposure to low but not high doses of bisphenolA(BPA): evidence for effects on body weight,food intake, adipocytes, leptin, adiponectin, insulin and glucose regulation. Reprod Toxicol. 2013;42:256–268.
  37. Batista TM, Alonso-Magdalena P, Vieira E, et al. Shortterm treatment with bisphenol-A leads to metabolic abnormalities in adult male mice. PLoS One. 2012;7:e33814.
  38. Decherf S, Demeneix BA. The obesogen hypothesis: a shift of focus from the periphery to the hypothalamus. J Toxicol Environ Health B Crit Rev. 2011;14:423–448.
  39. Manikkam M, Tracey R, Guerrero-Bosagna C, Skinner MK. Plastics derived endocrine disruptors (BPA, DEHP and DBP) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations. PLoS One. 2013;8:e55387.
  40. Alonso-Magdalena P, Quesada I, Nadal A. Endocrine disruptors in the etiology of type 2 diabetes mellitus. Nat Rev Endocrinol. 2011;7:346–353.
  41. Soriano S, Alonso-Magdalena P, García-Arévalo M, et al.Rapid insulinotropic action of low doses of bisphenol-A on mouse and human islets of Langerhans: role of estrogen receptor . PLoS One. 2012;7:e31109.
  42. Ruzzin J, Petersen R, Meugnier E, et al. Persistent organic pollutant exposure leads to insulin resistance syndrome. Environ Health Perspect. 2010;118:465–471.
  43. Naville D, Pinteur C, Vega N, et al. Low-dose food contaminants trigger sex-specific, hepatic metabolic changes in the progeny of obese mice. FASEB J. 2013;27:3860–3870.
  44. Bodin J, Bølling AK, Becher R, Kuper F, Løvik M, Nygaard UC. Transmaternal bisphenol A exposure accelerates diabetes type 1 development in NOD mice. Toxicol Sci. 2014;137:311–323.
  45. Brieño-Enríquez MA, Reig-Viader R, Cabero L, et al. Gene expression is altered after bisphenol A exposure in human fetal oocytes in vitro. Mol Hum Reprod. 2012;18:171–183.
  46. Trapphoff T, Heiligentag M, El Hajj N, Haaf T, Eichenlaub-Ritter U. Chronic exposure to a low concentration of bisphenolA during follicle culture affects the epigenetic status of germinal vesicles and metaphase II oocytes. Fertil Steril. 2013;100:1758–1767.
  47. Li Y, Zhang W, Liu J, et al. Prepubertal bisphenol A exposure interferes with ovarian follicle development and its relevant gene expression. Reprod Toxicol. 2014;44:33– 40.
  48. Craig ZR, Hannon PR, Wang W, Ziv-Gal A, Flaws JA. Di-n-butyl phthalate disrupts the expression of genes involved in cell cycle and apoptotic pathways in mouse ovarian antral follicles. Biol Reprod. 2013;88:23.
  49. Peretz J, Gupta RK, Singh J, Hernández-Ochoa I, Flaws JA. Bisphenol A impairs follicle growth, inhibits steroidogenesis, and downregulates rate-limiting enzymes in the estradiol biosynthesis pathway. Toxicol Sci. 2011;119:209–217.
  50. Chang CC, Hsieh YY, Hsu KH, Tsai HD, Lin WH, Lin CS. Deleterious effects of arsenic, benomyl and carbendazim on human endometrial cell proliferation in vitro. Taiwan J Obstet Gynecol. 2010;49:449–454.
  51. Ehrlich S, Williams PL, Missmer SA, et al. Urinary bisphenol A concentrations and implantation failure among women undergoing in vitro fertilization. Environ Health Perspect. 2012;120:978–983.
  52. Ziv-Gal A, Wang W, Zhou C, Flaws JA. The effects of in utero bisphenol A exposure on reproductive capacity in several generations of mice. Toxicol Appl Pharmacol.2015;284:354–362.
  53. Schmidt JS, Schaedlich K, Fiandanese N, Pocar P, Fischer B. Effects of di(2-ethylhexyl) phthalate (DEHP) on female fertility and adipogenesis inC3H/N mice. Environ Health Perspect. 2012;120:1123–1129.
  54. Buck Louis GM, Rios LI, McLain A, Cooney MA, Kostyniak PJ, Sundaram R. Persistent organochlorine pollutants and menstrual cycle characteristics. Chemosphere. 2011;85:1742–1748.
  55. Grindler NM, Allsworth JE, Macones GA, Kannan K, Roehl KA, Cooper AR. Persistent organic pollutants and early menopause in U.S. women. PLoS One. 2015;10:e0116057.
  56. Hoover RN, Hyer M, Pfeiffer RM, et al. Adverse health outcomes in women exposed in utero to diethylstilbestrol. N Engl J Med. 2011;365:1304–1314.
  57. Smith KW, Souter I, Dimitriadis I, et al. Urinary paraben concentrations and ovarian aging among women from a fertility center. Environ Health Perspect. 2013;121:1299–1305.
  58. Abbott DH, Nicol LE, Levine JE, Xu N, Goodarzi MO, Dumesic DA. Nonhuman primate models of polycystic ovary syndrome. Mol Cell Endocrinol. 2013;373:21–28.
  59. Kim SH, Chun S, Jang JY, Chae HD, Kim CH, Kang BM. Increased plasma levels of phthalate esters in women with advanced-stage endometriosis: a prospective case-control study. Fertil Steril. 2011;95:357–359.
  60. Resuehr D, Glore DR, Taylor HS, Bruner-Tran KL, Osteen KG. Progesterone-dependent regulation of endometrial cannabinoid receptor type 1 (CB1-R) expression is disrupted in women with endometriosis and in isolated stromal cells exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Fertil Steril. 2012;98:948–956.e1.
  61. Bruner-Tran KL, Ding T, Osteen KG. Dioxin and endometrial progesterone resistance. Semin Reprod Med. 2010;28:59–68.
  62. Wang Y, Yu J, Luo X, et al. Abnormal regulation of chemokine TECK and its receptor CCR9 in the endometriotic milieu is involved in pathogenesis of endometriosis by way of enhancing invasiveness of endometrial stromal cells. Cell Mol Immunol. 2010;7:51–60.
  63. Li MQ, Hou XF, Lv SJ, et al. CD82 gene suppression in endometrial stromal cells leads to increase of the cell invasiveness in the endometriotic milieu. J Mol Endocrinol. 2011;47:195–208.
  64. Gao X, Yu L, Castro L, et al. An endocrine-disrupting chemical, fenvalerate, induces cell cycle progression and collagen type I expression in human uterine leiomyoma and myometrial cells. Toxicol Lett. 2010;196:133–141.
  65. D’Aloisio AA, Baird DD, DeRoo LA, Sandler DP. Association of intrauterine and early-life exposures with diagnosis of uterine leiomyomata by 35 years of age in the Sister Study. Environ Health Perspect. 2010;118:375–381.
  66. Mahalingaiah S, Hart JE, Wise LA, Terry KL, Boynton-Jarrett R, Missmer SA. Prenatal diethylstilbestrol exposure and risk of uterine leiomyomata in the Nurses’ Health Study II. Am J Epidemiol. 2014;179:186–191.
  67. Peretz J, Vrooman L, Ricke WA, et al. Bisphenol A and reproductive health: update of experimental and human evidence, 2007–2013. Environ Health Perspect. 2014;122:775–786.
  68. Chakraborty TR, Alicea E, Chakraborty S. Relationships between urinary biomarkers of phytoestrogens, phthalates, phenols, and pubertal stages in girls. Adolesc Health Med Ther. 2012;3:17–26.
  69. Chen CY, Chou YY, Wu YM, Lin CC, Lin SJ, Lee CC. Phthalates may promote female puberty by increasing kisspeptin activity. Hum Reprod. 2013;28:2765–2773.
  70. Henley, D.V., Lipson, N., Korach, K.S., Bloch, C.A., 2007. Prepubertal gynecomastia linked to lavender and tea tree oils. N. Engl. J. Med. 356, 479–485.
  71. Rasier, G., Toppari, J., Parent, A.-S., Bourguignon, J.-P., 2006. Female sexual maturation and reproduction after prepubertal exposure to estrogens and endocrine disrupting chemicals: a review of rodent and human data. Mol. Cell. Endo.254–255, 187–201.
  72. Saiyed, H., Dewan, A., Bhatnagar, V., Shenoy, U., Shenoy, R., Rajmohan, H., Patel, K., Kashyap, R., Kulkarni, P., Rajan, B., Lakkad, B., 2003. Effect of endosulfan on male reproductive development. Environ. Health Perspect. 111, 1958–1962.
  73. . Guo, Y.L., Lambert, G.H., Hsu, C.C., Hsu, M.M., 2004. Yucheng: health effects of prenatal exposure to polychlorinated biphenyls and dibenzofurans. Int. Arch. Occup. Environ. Health 77, 153–158.
  74. Leijs,M.M., Koppe, J.G., Olie, K., van Aalderen,W.M.C., de Voogt, P., Vulsma, T.,Westra, M., ten Tusscher, G.W., 2008. Delayed initiation of breast development in girls with higher prenatal dioxin exposure; a longitudinal cohort study. Chemosphere 73, 999–1004.
  75. Hauser, R., Sergeyev, O., Korrick, S., Lee,M.M., Revich, B., Gitin, E., Burns, J.S.,Williams, P.L., 2008. Association of blood lead levels with onset of puberty in Russian boys. Environ. Health Perspect. 116, 976–980.
  76. Selevan, S.G., Rice, D.C., Hogan, K.A., Euling, S.Y., Pfahles-Hutchens, A., Bethel, J., 2003. Blood lead concentration and delayed puberty in girls. N. Engl. J. Med.348, 1527–1536.
  77. Damgaard IN, Skakkebaek NE, Toppari J, et al. Persistent pesticides in human breast milk and cryptorchidism. Environ Health Perspect. 2006;114:1133–1138.
  78. Fernandez MF, Olmos B, Granada A, et al. Human exposure to endocrine-disrupting chemicals and prenatal risk factors for cryptorchidism and hypospadias: a nested case-control study. Environ Health Perspect. 2007;115(suppl 1):8 –14.
  79. Virtanen HE, Koskenniemi JJ, Sundqvist E, et al. Associations between congenital cryptorchidism in newborn boys and levels of dioxins and PCBs in placenta. Int J Androl. 2012;35:283–293.
  80. Brucker-Davis F, Wagner-Mahler K, Delattre I, et al. Cryptorchidism at birth in Nice area (France) is associated with higher prenatal exposure to PCBs and DDE, as assessed by colostrum concentrations. Hum Reprod.2008;23:1708–1718.
  81. Cook MB, Trabert B, McGlynn KA. Organochlorine compounds and testicular dysgenesis syndrome: human data. Int J Androl. 2011;34:e68–e84; discussion e84–e65.
  82. Trabert B, Longnecker MP, Brock JW, Klebanoff MA, McGlynn KA. Maternal pregnancy levels of trans-nonachlor and oxychlordane and prevalence of cryptorchidism and hypospadias in boys. Environ Health Perspect.2012;120:478–482.
  83. Vested A, Giwercman A, Bonde JP, Toft G. Persistent organic pollutants and male reproductive health. Asian J Androl. 2014;16:71–80.
  84. Aneck-Hahn NH, Schulenburg GW, Bornman MS, Farias P, de Jager C. Impaired semen quality associated with environmental DDT exposure in young men living in a malaria area in the Limpopo Province, South Africa. J Androl. 2007;28:423–434.
  85. De Jager C, Farias P, Barraza-Villarreal A, et al. Reduced seminal parameters associated with environmental DDT exposure and p,p’-DDE concentrations in men in Chiapas, Mexico: a cross-sectional study. J Androl. 2006;27:16–27.
  86. Mocarelli P, Gerthoux PM, Needham LL, et al. Perinatal exposure to low doses of dioxin can permanently impair human semen quality. Environ Health Perspect. 2011;119:713–718.
  87. Abdelouahab N, Ainmelk Y, Takser L. Polybrominated diphenyl ethers and sperm quality. Reprod Toxicol.2011;31:546–550.
  88. Hwang K-A, Choi K-C. Endocrine-Disrupting Chemicals with Estrogenicity Posing the Risk of Cancer Progression in Estrogen-Responsive Organs. In: Advances in Molecular Toxicology [Internet]. Elsevier; 2015 [cited 2021 Feb 7]. p. 1–33. Available from: https://linkinghub.elsevier.com/retrieve/pii/B9780128022290000013.
  89. Warner M, Eskenazi B, Mocarelli P, et al. Serum dioxin concentrations and breast cancer risk in the Seveso Women’s Health Study. Environ Health Perspect. 2002;110:625–628.
  90. Boada LD,ZumbadoM,Henríquez-Hernández LA, et al. Complex organochlorine pesticide mixtures as determinant factor for breast cancer risk: a population-based case-control study in the Canary Islands (Spain). Environ Health. 2012;11:28.
  91. López-Carrillo L, Hernández-Ramírez RU, Calafat AM, et al. Exposure to phthalates and breast cancer risk in northern Mexico. Environ Health Perspect. 2010;118:539–544.
  92. Donna A, Crosignani P, Robutti F, et al. Triazine herbicides and ovarian epithelial neoplasms. Scand J Work Environ Health. 1989;15:47–53.
  93. VieiraVM,HoffmanK, ShinHM,Weinberg JM, Webster TF, Fletcher T. Perfluorooctanoic acid exposure and cancer cancer outcomes in a contaminated community: a geographic analysis. Environ Health Perspect. 2013;121:318–323.
  94. Driscoll SG, Taylor SH. Effects of prenatal maternal estrogen on the male urogenital system. Obstet Gynecol.1980;56:537–542.
  95. Koutros S, Beane Freeman LE, Lubin JH, et al. Risk of total and aggressive prostate cancer and pesticide use in the Agricultural Health Study. Am J Epidemiol. 2013;177:59–74.
  96. ChristensenCH,Platz EA, Andreotti G, et al. Coumaphos exposure and incident cancer among male participants in the Agricultural Health Study (AHS). Environ Health Perspect. 2010;118:92–96.
  97. Usmani KA, ChoTM,Rose RL, Hodgson E. Inhibition of the human liver microsomal and human cytochrome P450 1A2 and 3A4 metabolism of estradiol by deployment-related and other chemicals. Drug Metab Dispos.2006;34:1606–1614.
  98. Institute of Medicine. Veterans and Agent Orange: Update 2012. Washington DC: The National Academy of Sciences; 2014.
  99. Hinson, J. P., & Raven, P. W. (2006). Effects of endocrine-disrupting chemicals on adrenal function. Best Practice & Research Clinical Endocrinology & Metabolism, 20(1), 111–120.
  100. Kioumourtzoglou M-A, Coull BA, O’Reilly ÉJ, Ascherio A, Weisskopf MG. Association of Exposure to Diethylstilbestrol During Pregnancy With Multigenerational Neurodevelopmental Deficits. JAMA Pediatr. 2018 Jul 1;172(7):670–7.
  101. Cantonwine D, Meeker JD, Hu H, et al. Bisphenol A exposure in Mexico City and risk of prematurity: a pilot nested case control study. Environ Health. 2010;9:62.
  102. Ferguson KK, McElrath TF, Meeker JD. Environmental phthalate exposure and preterm birth. JAMA Pediatr.2014;168:61–67.
  103. Mustafa MD, Banerjee BD, Ahmed RS, Tripathi AK, Guleria K. Gene-environment interaction in preterm delivery with special reference to organochlorine pesticides. Mol Hum Reprod. 2013;19:35–42.
  104. Peltier MR, Arita Y, Klimova NG, et al. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) enhances placental inflammation. J Reprod Immunol. 2013;98:10–20.
  105. Snijder CA, Heederik D, Pierik FH, et al. Fetal growth and prenatal exposure to bisphenol A: the generationRstudy. Environ Health Perspect. 2013;121:393–398.
  106. Burdorf A, Brand T, Jaddoe VW, Hofman A, Mackenbach JP, Steegers EA. The effects of work-related maternal risk factors on time to pregnancy, preterm birth and birth weight: the Generation R Study. Occup Environ Med. 2011;68:197–204.
  107. Snijder CA, te Velde E, Roeleveld N, Burdorf A. Occupational exposure to chemical substances and time to pregnancy: a systematic review. Hum Reprod Update. 2012;18:284–300.
  108. Suzuki Y, Yoshinaga J, Mizumoto Y, Serizawa S, Shiraishi H. Foetal exposure to phthalate esters and anogenital distance in male newborns. Int J Androl. 2012;35:236–244.
  109. Gemmill A, Gunier RB, Bradman A, Eskenazi B, Harley KG. Residential proximity to methyl bromide use and birth outcomes in an agricultural population in California. Environ Health Perspect. 2013;121:737–743.
  110. Migeot V, Albouy-Llaty M, Carles C, et al. Drinking water exposure to a mixture of nitrate and low-dose atrazine metabolites and small-for-gestational age (SGA) babies: a historic cohort study. Environ Res. 2013;122:58–64.
  111. Wang P, Tian Y, Wang XJ, et al. Organophosphate pesticide exposure and perinatal outcomes in Shanghai, China. Environ Int. 2012;42:100–104.
  112. Kishi R, Sasaki S, Yoshioka E, Yuasa M, Sata F, Saijo Y, Kurahashi N, Tamaki J, Endo T, Sengoku K, Nonomura K, Minakami H; Hokkaido Study on Environment and Children's Health. Cohort profile: the Hokkaido study on environment and children's health in Japan. Int J Epidemiol. 2011 Jun;40(3):611-8.
  113. Paulose R, Jegatheesan K, Balakrishnan GS. A big data approach with artificial neural network and molecular similarity for chemical data mining and endocrine disruption prediction. Indian J Pharmacol. 2018 Jul 1;50(4):169.
  114. Jaffrezic-Renault N, Kou J, Tan D, Guo Z. New trends in the electrochemical detection of endocrine disruptors in complex media. Anal Bioanal Chem. 2020 Sep;412(24):5913–23.
  115. Street ME, Audouze K, Legler J, Sone H, Palanza P. Endocrine Disrupting Chemicals: Current Understanding, New Testing Strategies and Future Research Needs. Int J Mol Sci. 2021 Jan 19;22(2):933.
  116. Artificial Intelligence Facilitates Chemical Toxicity Evaluation: the Case of Bisphenol S [Internet]. Newsroom | Inserm. 2019 [cited 2021 Feb 8]. Available from: https://presse.inserm.fr/en/artificial-intelligence-facilitates-chemical-toxicity-evaluation-the-case-of-bisphenol-s/34525/
  117. Porta N, ra Roncaglioni A, Marzo M, Benfenati E. QSAR Methods to Screen Endocrine Disruptors. Nucl Recept Res [Internet]. 2016 [cited 2021 Feb 17];3. Available from: http://www.kenzpub.com/journals/nurr/2016/101203/
  118. Carvalho, F.P., 2006. Agriculture, pesticides, food security and food safety. Environ. Sci.Policy 9, 685–692.
  119. Toteja G.S, Dasgupta J, Saxena B.N, Kalra R.L, editors. Report of an ICMR Task Force Study (Part 1). New Delhi: Indian Council of Medical Research; 1993. Surveillance of Food Contaminants in India.
  120. Kannan K, Tanabe S, Ramesh A, Subramanian A, Tatsukawa R. Persistent organochlorine residues in food stuffs from India and their implications on human dietary exposure. J Agric Food Chem. 1992;40:518.

 

 

Diabetes Mellitus and Infections

ABSTRACT

 

Diabetes presents a significant risk factor for all kinds of infections. It has been well described to increase rates of outpatient infection as well as the incidence of infections requiring hospitalization. This appears to be related to deficits in the immune system, particularly changes seen in innate immunity. Respiratory infections, skin and soft tissue infections, gastrointestinal and genitourinary infections all appear to occur more frequently in those with DM. Not only are they more frequent, but these infections appear to have a poorer response to therapy and more rapid progression to severe forms of infection. There is good evidence that reduction of hyperglycemia can improve outcomes. Among the antihyperglycemic agents available, translational and clinical data exists that insulin can help to improve immune function and potentially metformin as well.

 

INTRODUCTION

 

The 2020 release of the US Centers for Disease Control and Prevention National Diabetes Statistics Report revealed that in 2018, 34.2 million individuals had diabetes mellitus (DM), representing approximately 10.5% of the US population and a total cost of care in excess of US $300 billion (1).  Part of these numbers reflects the impact of DM on infection rates and morbidity/mortality. Both type 1 and type 2 DM are associated with a significantly higher risk of infection, both in the outpatient and inpatient context, and the outcomes are generally worse than in those without diabetes. We will discuss here the impact of diabetes on the immune system, specific infections which are commonly seen in diabetes, and the influence of various therapies targeted both at glycemic control and also on immunomodulation on infection outcomes.

 

EPIDEMIOLOGY

 

DM, both type 1 and type 2, is associated with a high risk of infection. A large retrospective study of primary care patients revealed that diabetes is likely to account for 6% of infection-related hospitalizations and 12% of infection-related deaths, with the strongest associations being for bone and joint infections, development of sepsis, and cellulitis (2).

 

Outpatient

 

A number of studies have been performed on the rate of infection among patients with diabetes in the primary care and other outpatient settings. In a Canadian cohort of 1,779 patients with DM matched to 11,066 without DM the patients with DM had an increased risk of infection (adjusted odds ratio 1.21, adjusted for confounding variables). with skin and soft tissue infections having the strongest association with having DM.  Interestingly, DM was not found in this study to be associated with head and neck, musculoskeletal, or viral infections (3). Another large Canadian study including more than a million individuals matched those with DM to those without, and assessed all physician and hospital claims for infectious disease. It found that almost half of all individuals with DM had a claim for an infectious disease within a cohort year compared with 38% of those without DM. The risk ratio was skewed most towards those with DM for upper respiratory tract infections, cystitis, and pneumonia (4).

 

Inpatient

 

In one study, having diabetes led to a 2-fold increased risk for hospitalization when presenting with an infection to the emergency room, and up to 12% of inpatient admissions in patients with diabetes were the consequence of an infection (5). A South Korean showed that those with diabetes had a significantly greater risk of infection-related ICU admission and death when hospitalized with infections of skin or soft tissue, central nervous system infection, or bone and joints (6). The previously-mentioned Canadian retrospective cohort study showed that, while the overall risk ratio for infection in those with diabetes versus without was 1.21, this number rose to 2.17 and 1.92 when considering infection which led to hospitalization and death, respectively (4). These and other studies (7,8) have revealed that not only is diabetes associated with (and causative of) an increased risk of infection, but also with higher rates of hospitalization, ICU stays, and death related to these infections. Of note, many of these patients with DM have other comorbidities which may not be able to be fully controlled for in these epidemiologic studies demonstrating higher estimates of the risk of infection with DM.

 

One important entity to consider in the inpatient arena is sepsis. The studies on sepsis are not consistent —though some show worse outcomes from DM, others have suggested either no effect or even a protective effect from DM (9). Data for the latter come from studies assessing acute respiratory failure and respiratory distress syndrome in the ICU, and it may be that the blunted immune response that we see in some patients with DM are responsible for the findings (i.e., reduced inflammation and injury related to impaired neutrophil function as described in section on Innate Immunity) (10). More studies are needed to better understand in what specific clinical contexts DM results in higher risk.

 

PATHOPHYSIOLOGY OF DIABETES AND IMMUNE SYSTEM

 

There is well-known disruption of the immune system in diabetes which occurs at multiple levels. Innate and adaptive immunity are affected along with cytokine signaling within both. This dysregulation occurs both in those with type 1 and 2 DM. Microvascular complications such as neuropathy also increase susceptibility to an accidental lesion in the barrier of the skin which forms one of the first lines of defense. Furthermore, poor vascular flow to sites of infection can further compromise an appropriate immune response and healing leading to worsening or secondary infections (11). In our discussion on alterations of the immune system, it is important to note that we have focused on alterations in function as opposed to baseline differences in the number of immune cells or cytokine levels between those with and without DM. Table 1 provides a summary of the alterations in immune dysfunction that are known.

 

Innate Immunity

 

COMPLEMENT SYSTEM

 

The complement system plays a critical role in both innate and adaptive immunity and leads to the opsonization, lysis and phagocytosis of pathogens along with recruitment of immune cells to the site of infection. There is known to be a reduction in the complement factor 4 (C4) level in those with type 1 DM, although it is unclear if that alone can precipitate increased risk of infection (12).  A variety of studies demonstrate that hyperglycemia can inhibit phagocytosis through the system, potentially by reducing complement binding to immunoglobulins. Glycosylation of C3 can also impair its ability to attach to the pathogen surfaces (13).

 

Table 1. Impact of Diabetes and Hyperglycemia on the Immune System

Innate Immunity

 

Complement System

- Reduction in C4 levels (Unclear relevance for infection risk)

- Reduction complement binding to immunoglobulins

- Glycosylation of C3 can impair binding to pathogen surface

Recruitment and Pathogen Recognition

- Reduced CAM expression leading to reduced leukocyte recruitment

- In setting of hyperglycemia reduced production of chemokine in response to bacterial LPS

- Advanced glycation end products inhibit neutrophil transendothelial migration

- Reduced expression TLR (which allows for recognition LPS)

Cellular Dysfunction

- Reduced H2O2 production leading to reduced bactericidal ability in both macrophage and neutrophils

- Impairment of macrophage phagocytic ability through complement pathway

- Impaired metabolism of glucose in macrophages reducing activity

- NK cell activity reduced through reduced expression activating receptors NKG2D and NKp46

Adaptive Immunity

- Depletion and dysfunction memory CD4+ cells

- Not as well described as alterations in innate immunity

Cytokine Signaling

- Deficiency of IL-1b, IL-2, IL-6, IL-10, IL-22, IFN-γ, TNF-a

Skin and Mucosal Barriers

- Vascular compromise leading to impaired healing

- Neuropathy making breakage of the skin more likely

 

RECRUITMENT AND PATHOGEN RECOGNITION

 

A number of older studies reported reduced chemotaxis of polymorphonuclear leukocytes (PMNs) in patients who have DM (14,15). One of the mechanisms appears to be related to disruption of cellular adhesion molecules (CAMs) which are critical for the recruitment of leukocytes to the site of infection. This was seen when db/db (a diabetic mouse model) and wild type mice were infected with West Nile Virus and subsequent analysis of the brains of the db/db showed less leukocyte recruitment consistent with reduced CAM expression compared with the brains of the wild type mice (16). When hyperglycemia was induced in mice exposed to Klebsiella pneumoniae, there was a reduced recruitment of granulocytes to the site of infection as compared with control mice. This was felt to be related to a reduction in the chemokine production able to be induced by the bacterial lipopolysaccharide (LPS) (17). There are also in vitro data which reveal that advanced glycation end products are able to inhibit neutrophil transendothelial migration (18). A reduction in the expression of Toll-like receptors (TLR, which bind to LPS and allow for pathogen recognition) in patients with poorly controlled diabetes has been described as well (19).

 

SPECIFIC CELLULAR DYSFUNCTION

 

Multiple immune cells are impacted in DM. Neutrophil recruitment is not only reduced, but there are also good data demonstrating their reduced phagocytic activity and hydrogen peroxide production, leading to reduced bactericidal ability (20-23). The mechanism for the alteration appears may be partially linked to impaired metabolism of glucose and glutamine, as was demonstrated in streptozotocin-induced diabetic rats (24). Macrophages are also similarly affected. Examination of cells from patients with type 2 DM have revealed that there is impairment of both the complement and Fc-gamma receptor-mediated pathways by which macrophages are able to phagocytize pathogens (25). When macrophages from diabetic mice were cultured in normal versus high glucose, there was a reduction in the phagocytic and bactericidal activity, apparently through a defect similar to that seen in neutrophils, specifically impaired metabolism of glucose (26). NK cell activity is also known to be reduced in a manner which is related to level of glycemic control, demonstrated in multiple studies comparing NK cells from patients with DM, prediabetes, and also without DM (27). The mechanism of the reduced activity may be in part related to decreased expression of activating receptors NKG2D and NKp46 on the NK cells in patients with DM (28).

 

Adaptive Immunity

 

The adaptive immune system is activated in response to specific pathogens and involves the immunologic memory for those pathogens. There are two components of adaptive immunity, the humoral and cellular, which carry out the major purposes of generating an antibody and cellular (involving B and T cells) response to a specific antigen. The adaptive humoral immunity is involved in antibody production. This appears to be preserved in those with DM on the basis of overall appropriate response to various vaccines (29,30). However, there may be some dysregulation of the adaptive cellular immunity. There is a depletion of memory CD4+ cells that has been noted prior to the development of type 1 DM (31). Furthermore, a dysfunction of and an impaired response of these cells to Streptococcus pneumoniae has been described (32). However, the dysfunction seen in DM with innate immunity is better understood than that the dysfunction in adaptive immunity (33).

 

Cytokine Signaling Defects

 

Cytokines play a vital role in the signaling cascades which underpin the immune system, allowing for full activation of both innate and adaptive immune responses. Multiple points of dysregulation have been identified in DM. With stimulation, multiple of these cytokines have been shown to be secreted at lower levels than would be typical with stimulation. In vitro studies done on monocytes isolated from patients without DM showed suppression of IL-1b, IL-2, IL-6, and IL-10 secretion in the presence of hyperglycemia (34-36). In diabetic mice, there was noted to be immune dysregulation and also inflammation which was related to IL-22 deficiency reversed with provision of IL-22 (37). There is also evidence for impaired interferon gamma (IFN-γ) and TNF alpha (TNF-a) production from T cells in the setting of methylglyoxal, a compound which is increased in those with DM (38). The end result of these deficits is that there is attenuation of the phagocytic and cellular immune response.

 

COMMON INFECTIONS, OUTCOMES, AND GENERAL DRUG OF CHOICE

 

In addition to many infections having a worse course in those with diabetes, specific types of infections are also significantly more common in those with diabetes. We will review these diabetes-predominant infections (39-42). Table 2 also provides an overview of common infections and considerations in those with DM.

 

Table 2. Common infections seen in patients with diabetes with attention to diagnosis, common responsible organisms, management, and outcomes

Respiratory Infections

 

Pneumonia

- Higher rates of hospitalization and also mortality in those with DM compared with those without

- Less commonly presents with purulent cough and pleuritic chest pain, more commonly with altered consciousness

- Aspiration and skin colonization common etiologies

- S pneumonia, S aureus, and K pneumoniae among most common organism

- Rx with amoxicillin/clavulanate or cephalosporin + macrolide/doxycycline vs fluoroquinolone

Tuberculosis

- Higher risk contracting with risk corresponding with level glycemic control

- Higher risk of treatment failure

- Isoniazid needs to be taken with pyridoxine to prevent neuropathy

- Rifampin can cause hyperglycemia and also induces cyp450 leading to increased clearance of various DM agents

Skin and Soft Tissue

 

Cellulitis/Abscess

- Most common SSTI seen in those with DM

- Most common organism Staph species

- For abscess culture is needed to determine organism and resistance

- Oral abx: Doxycycline, clindamycin, TMP-SMX, cephalexin

- Presence SIRS or other complication: IV vancomycin, linezolid, ceftaroline

Necrotizing Fasciitis

- Comorbid DM much more common

- Limb loss seen more often

- Polymicrobial typically but can be only K pneumoniae

- Surgical rx most common and need broad spectrum coverage

Fournier Gangrene

- More commonly seen in those with DM

- Anaerobic and aerobic bacteria such as S aureus and Pseudomonas species

- Debridement a must

- Seen with SGLT2i

Sternal Wound Infection

- DM one of strongest predictors for infection

- Improved glycemic control with insulin shown to reduce rate of infection

Gastrointestinal

 

Hepatitis

- HCV outcomes worse with more frequent cirrhosis and failure of antivirals

Emphysematous Cholecystitis

- Diagnosis through sonography or CT typically as first step

- Most common organism C perfringens and E coli

- Rx is typically cholecystectomy but can try abx in mild case

Genitourinary

 

Urinary Tract Infection

- Higher rate of infection and failure/relapse with rx

- Most common organism E choli and Enterobacteriaceae

- Urine culture is strongly recommended

- Do not treat asx bactiuria

- Decision for abx is based on local organism and resistance trend

- Higher risk of progression to pyelonephritis which is more severe and often bilateral

Head and Neck

 

Necrotizing Otitis Externa

- DM higher risk of abscess formation requiring draining

- Vascular compromise and pseudomonal vasculitis much more commonly seen in DM

- P aeruginosa most common organism

- Confirm with CT

- Systemic abx with antipseudomonal action and local therapy to the canal including cleaning/debridement

Fungal Infections

 

Onychomycosis

- Potentially up to 1/3 of all patients with DM impacted

- Diagnosis based on fungal culture/microscopy

- Oral agents most effective

Genitourinary

- Most common Candida specie

- Increased ability to bind with receptor in DM

UTI

- Communicate with lab on culture that Candida specie is suspected

- If symptomatic, then fluconazole first line

Mucormycosis

- Causative agents are the mucormycetes

- Most commonly sinus +/- cerebrum/orbits

- Respiratory tract second most common

- Skin third most common and has ulcerative necrotic lesion

- Tissue biopsy needed and imaging helpful to identify extent of infection

- Debulking of infection with adjuvant

Abbreviations: Rx (Treatment), Abx (Antibiotics), Asx (Asymptomatic), SSTI (Skin and soft tissue infections), UTI (Urinary tract infection)

 

Respiratory Infections

 

Pneumonia is a frequently-seen infection in those with DM. In a large Danish population-based case-control study of 34,239 patients, the relative risk for hospitalization from community-acquired pneumonia was 1.26 compared with patients without DM. Furthermore, the risk appeared to be correlated with level of glycemic control with relative risk (RR) for those with HbA1c <7% being 1.22, versus a RR of 1.6 when HbA1c was ≥9% (43). A Portuguese study similarly showed DM prevalence was higher in those with pneumonia and that outcomes were worse with a longer hospital stay and significantly higher mortality in patients with DM versus those without (15.2% vs 13.5%) (44). These trends were also seen in another Danish study which showed mortality was greater in those with type 2 DM compared with other patients at both 30 and 90 days after the initial pneumonia episode (45). The presentation of pneumonia is potentially different in those with DM as typically bacterial pneumonia presents with a purulent cough and pleuritic chest pain, symptoms which are less commonly seen in those with DM. The hypothesis is that the lowered immune defense results in a decreased inflammatory response and symptoms. Notably, altered consciousness is more common on presentation with pneumonia in those with DM. The causative agent of pneumonia is similar in those with and without DM with the most common being Streptococcus pneumoniae (46). However, there is an over-representation of organisms such as Staphylococcus aureus and Klebsiella pneumoniae related to skin colonization and more frequent aspiration in those with DM. Management is using combination therapy with amoxicillin/clavulanate or cephalosporin and a macrolide or doxycycline versus monotherapy with respiratory fluoroquinolone (47).

 

Diabetes also represents an important risk factor for contracting tuberculosis (TB). The odds of developing tuberculosis appear to be higher in those with DM compared to those without with the odds ratio ranging from 2.44-8.33 in various studies. Furthermore, severity of DM appears to be correlated with greater risk of contracting TB based on studies comparing the incidence of TB in those with insulin-dependent versus non-insulin dependent DM. Risk of treatment failure despite good adherence to medication regimen and also death from tuberculosis all appear to be increased (48). There are side effects of medications targeted at tuberculosis with particular relevance in DM. Isoniazid can cause peripheral neuropathy that could be mistaken for diabetic neuropathy, and pyridoxine should be administered to ameliorate this risk. Rifampicin has the ability to cause hyperglycemia. Rifampicin also is a powerful inducer of the cytochrome P450 system leading to increased clearance of multiple DM agents (i.e., sulfonylurea, pioglitazone, meglitinides). Hence while the regimen to treat TB seen in patients with DM is the same as the regimen in those without, special attention must be paid to these DM specific issues.

 

Skin and Soft Tissue Infections

 

There is a significantly increased risk of skin and soft tissue infections (SSTI) in DM. Using a large administrative claims database (HealthCore Integrated Research Database), Suaya et al were able to demonstrate complications from SSTI were five times higher, and hospitalization four times higher, in patients with DM than those without DM in their cohort (49). The most common agent of infection is Staphylococcus aureus (S aureus). The foot represents the most common site of infection as well which has been expertly covered in another chapter (50).

 

For cellulitis, the diagnosis is typically clinical, as opposed to an abscess which often is cultured for organism identification and resistance profiling. The decision for antibiotics in cellulitis and with abscess is often empiric, and in those with DM it is imperative that the choice cover Staphylococcus species. Potential first line therapy for outpatient oral antibiotics includes doxycycline, clindamycin, trimethoprim-sulfamethoxazole, and cephalexin. If there is admission due to SIRS criteria being present or a suspected complication, then the recommendations change to IV predominant choices including vancomycin, linezolid, and ceftaroline (51).

 

Necrotizing fasciitis is a life-threatening condition involving the subcutaneous fat and deep fascial layers. A retrospective report of 59 necrotizing fasciitis cases at a single center revealed that 11 of the cases had DM, and another study showed that 51% of 84 patients in the cohort with necrotizing fasciitis had DM (52,53). Though most commonly a polymicrobial infection, Klebsiella pneumoniae is also commonly seen as a single isolate (53). Prognosis appears to be poorer in those with DM, with a higher rate of limb loss than that seen in those without DM. Broad spectrum antibiotics are utilized, related to the frequent polymicrobial nature of the infection. However, ischemia compromises appropriate antibiotic concentration at the site, therefore the management is primarily surgical involving a combination of debridement, necrosectomy, and fasciotomy frequently (54).

 

Fournier gangrene represents a particularly serious SSTI, defined as a necrotizing skin infection of the scrotum and penis or vulva. Typically, patients are between the age of 50-60 years and DM represents a serious risk factor for development. Usually, the infection will begin in the perianal or retroperitoneal region and then spreads to the genitalia or as a urinary tract infection which then also moves towards the genitalia. There will be necrosis and crepitus which is an indication of involvement of the underlying skin and soft tissue. The etiology is usually a mix of aerobic and anaerobic bacteria which can commonly include S aureus and Pseudomonas (51). Surgical debridement is typically necessary. This condition has become of particular relevance with the release of a warning from the US Food and Drug Administration in August 2018 that Fournier’s represented a rare but serious complication of sodium-glucose cotransporter 2 inhibitors (SGLT2i), a newer but increasingly utilized class of DM medications (55). There have been 55 cases of Fournier’s reported to the FDA between March 2013 and January 2019 (56). However, it is important to note that while this number appears to be higher than for other anti-hyperglycemic agents, that there has not been a clear establishment of causality here. In fact, other studies including a meta-analysis of randomized controlled trials involving SGLT2i and a “real-world” study using IBM MarketScan were unable to confirm an increased risk of Fournier’s Gangrene for those using SGLT2i versus those who did not (57,58).

 

Sternal wound infections after surgery are also known to occur more frequently in those with DM. After coronary artery bypass, presence of DM is one the strongest predictors for a deep sternal wound infection (odds ratio 2.6) (59). Improved glycemic control in the post-operative period has been shown to be able to significantly reduce the rate of sternal wound infection (60-62). Furnary et al in their seminal study were able to demonstrate in a prospective manner that use of an IV insulin infusion to keep glucose <200 mg/dL resulted in a 66% reduction in risk of sternal infection compared with nonrandomized historical controls (60).

 

Gastrointestinal Infections

 

The presence of DM has been known to worsen viral hepatitis. The outcome in chronic hepatitis C infection is worse in those with DM as compared to those without, corresponding to a significantly increased risk for cirrhosis and a reduced response to antiviral therapy in those with DM and hepatitis C (63). Emphysematous cholecystitis is a rare progression of acute cholecystitis, defined as presence of gas in the gallbladder wall for which DM serves as a major risk factor. The prevalence of DM among those with emphysematous cholecystitis has been described as high as 50% in the literature (64). Sonography is able to detect the presence of gas within the gallbladder wall and an abdominal radiograph will show a curvilinear lucency around the gallbladder. CT can also detect the condition with nearly 100% sensitivity. The two most common causative agents are Clostridium perfringens and Escherichia coli. The treatment is often surgical with prompt cholecystectomy although for a mild case antibiotic therapy can be initiated but without improvement within 3-4 days then the recommendation is still for cholecystectomy (65).

 

Genitourinary Infections

 

Urinary tract infections (UTI) are significantly more common in patients with DM with a large UK study showing an incidence of UTI of 46.9 per 1000 person-years among those who had type 2 DM versus 29.9 for those without DM (66). The most common pathogens in those with DM are Escherichia coli and the Enterobacteriaceae (Klebsiella, Proteus, Enterobacter, etc.). There is an increased risk of drug-resistant organisms being present. In making the diagnosis, beyond the typical clinical symptoms of dysuria and increased frequency, it is important to note that a urine culture really should be obtained in all individuals with DM prior to therapy (67). Outcomes are worse in those with DM, being associated with increased relapse and reinfection (at 7.1% and 15.9% respectively in those with DM versus 2% and 4.1% in those without) (68). Asymptomatic bacteriuria should not be treated even in those with DM. In those with symptoms, trimethoprim-sulfamethoxazole and ciprofloxacin are good choices but the general consensus is to follow local infection and resistance trends and tailor antibiotic therapy towards those organisms (67). From a lower UTI, there is also more risk of progression to pyelonephritis (both emphysematous and nonemphysematous) which tends to be more severe requiring hospitalization and are often bilateral in those with DM (69,70).

 

Head and Neck Infections

 

Consistent with the findings in many types of infections, head and neck infections are more common and appear to be more severe in those with DM. An assessment of 185 patients at the National Taiwan University Hospital with deep neck infections found that those with DM had a significantly higher rate of abscess formation than those without (89.3% vs 71.3%) and that surgical drainage was required more frequently as well (86% vs 65.2%) (71). One concerning entity commonly associated with DM is necrotizing external otitis. This term references an infection which has spread to the temporal and adjacent bones at the base of the skull. The causative agent is often Pseudomonas aeruginosa. The increased frequency of this in those with DM appears to be related in part to the vascular compromise seen in DM combined with a pseudomonal vasculitis (72). A certain level of suspicion for this condition needs to be present when there is external otitis in a patient at risk of necrotizing progression. Imaging is often needed to confirm with CT used. Systemic antibiotics are a requirement along with local treatment of the canal (i.e., cleaning, antimicrobial topicals, etc.). The antibiotic chosen needs to have antipseudomonal action such as the fluoroquinolones with the understanding that poor vascularization of the area often means higher doses are required (72).

 

Fungal Infections

 

Infection with Candida species is common in those with DM (73). Skin and soft tissue along with mucosa can be commonly impacted. Assessment of mouth swabs from patients with and without DM revealed that there was a higher frequency of Candida infection in the patients with DM (74). This appears to be related to decreased salivary pH and salivary flow which promoted colonization with the yeast. But there is also the possibility that in DM there is increased ability of Candida to bind to its receptor (75). Onychomycosis, a fungal infection of the nails, is also very common with DM with some studies suggesting up to 1/3 of individuals with DM are impacted. Diagnosis is made based on a positive fungal culture for a dermatophyte or microscopy showing fungus prior to initiation of therapy. Oral agents tend to be the most efficacious but topical lacquers are used as well (76).

 

Beyond the skin and mucosa, there is a high rate of genitourinary infections with Candida species. Among fungal infections of the urinary tract system, the vast majority involve Candida species. For both outpatient and inpatient urinary tract infections where Candida species was the causative agent, DM is present as a comorbid condition in 29% and 39% of cases respectively. Initial work-up would be similar to that of any other for urinary tract infection including urinalysis and culture. If there is a concern for Candida then that should be communicated with the reporting lab as there can be slow growth on certain cultures. It is important to remember that symptomatic urinary tract infections with Candida are rare. However, if truly a symptomatic infection is felt to be present, then fluconazole is typically the first line therapy of choice because of its ability to accumulate in high concentrations within the urine. If there is progression to pyelonephritis then speciation and sensitivities will be required given the presence of resistant strains and often dual agent therapy is required (77).

 

Mycotic genital infections are driven by Candida species. in both men and women (78). A very large percentage of women will experience symptomatic vulvovaginal candidiasis within their lifetime, and DM represents a risk factor for development of infection, with worsening glycemic control predisposing to an even higher risk (73). This appears to be related to known defects in immune cell function (PMNs and macrophages) and the impact of glucose in the urine, leading to worsening virulence in addition to improved adherence of yeast. Clinically, women present with pruritis and discomfort. Diagnosis is best made with microscopy and can be suspected in those with a negative amine (“whiff”) test and normal pH. Treatment is typically with a short course of intravaginal imidazole or triazole as opposed to oral fluconazole (78). For male patients, a similar finding of increased risk of balanitis (i.e., inflammation of the glans penis and prepuce) with DM and with increasing HbA1c is noted (73,79). This condition also occurs almost exclusively in males who have not been circumcised. Diagnosis is best made based on clinical presentation (burning and itching of penis worse after intercourse) in combination with a subpreputial culture. Imidazole or triazole creams represent the mainstay of therapy (78).

 

It is worth mentioning that SGLT2i as a class are known to be associated with genital infections, particularly mycotic genital infections caused by Candida species. Using information from two US based health insurance databases, Dave et al were able to demonstrate in a retrospective cohort study that use of an SGLT2i compared with dipeptidyl peptidase 4 inhibitors and glucagon-like peptide 1 receptor agonists led to an approximately three-fold increase in risk of genital infection (80). This might be mitigated in part by improved hygiene (i.e., rinsing the genital area with water after every episode of urination and before going to bed) (81). Interestingly, the association between use of SGLT2i and genital infections does not appear to extend to UTIs (82). This may be related to increased volume and flow through the urinary tract preventing excess bacterial load from developing (83). As SGLT2i are increasingly a mainstay of therapy for patients with type 2 DM, this association with mycotic genital infections needs to be carefully considered.

 

Mucormycosis is a rare but life-threatening fungal infection caused by a mucormycetes, a group of molds (commonly the mucor and rhizppus species). A metanalysis of cases of mucormycosis showed that DM was the commonest underlying condition present in 40% of cases (84). Frequently the DM associated is type 2 and uncontrolled. It carries with it a high fatality rate which ranges from 32-57%. The most common site of infection (66% of cases in a large 929 case series) is the sinus-cavity leading to rhinocerebral infection which can also impact the orbits. In these cases, there are often symptoms of sinus congestion/inflammation, fever, facial swelling along with ophthalmoplegia, cellulitis, and cranial nerve palsies. Necrotic eschars in the nasal cavity and on the hard palate are classic findings and also indicate a rapidly progressive infection. The respiratory tract is the second most commonly affected location (16%) with endobronchial lesions commonly found in those with DM. The subsequent invasion of the vasculature can lead to more distant infection. Finally, the third most common site is the skin (10%). Cutaneous mucormycosis presents with erythematous or ulcerative necrotic lesions which can also lead to osteomyelitis (85,86). Tissue biopsy is a requirement for diagnosis while various imaging modalities can help provide further evidence of an infection. Debulking of the infection through surgery is necessary. Amphotericin B and isavuconazole have been utilized as adjunctive therapies (85,87).

 

COVID-19 and DM

 

Becoming increasingly clear is the interaction between DM and SARS-CoV-2 (causative agent of coronavirus disease 2019 (COVID-19)). Multiple risk factors for contracting and having a more severe course of COVID-19 have been identified, including advanced age and male gender, but both type 1 and type 2 DM are now known to be important risk factors for morbidity and mortality with the disease (88). An assessment of patients who contracted COVID-19 and were tested within the Vanderbilt University Medical system demonstrated that there was a significantly increased risk for hospitalization in those with DM compared to without DM (odds ratio 3.36 for type 2 and 3.9 for type 1) and also for more severe disease course (odds ratio 3.42 for type 2 and 3.35 for type 1) (89). From a cohort of patients in England, there is evidence that poorly-controlled DM as compared with well-controlled DM (HbA1c 6.5-7% versus ³10%) results in significantly increased mortality in both type 1 and type 2 DM (hazard ratio 2.23 and 1.61 respectively) (90). Increased mortality has also been seen in another English cohort (83). A more comprehensive review on this topic is offered by Lim et al, where the multiple points at which COVID-19 and DM interact – including the impact of glucotoxicity on the lungs, increased thromboembolic risk, worsened oxidative stress. and inappropriately high levels of cytokine production leading to organ damage – are outlined (91).

 

IMPACT OF GLYCEMIC CONTROL AND OTHER THERAPIES

 

Glycemic Control and Diabetes Therapies

 

There is good evidence that glycemic control is correlated with infection. A study of 69,318 patients with type 2 DM in Denmark revealed an association between increased risk for community- and hospital-treated infection in those with higher HbA1c ≥10.5% compared with HbA1c 5.5-<6.4% (92). Similarly, in a large English cohort there was an increasing risk of infection in parallel with HbA1c for patients with both type 1 and type 2 (2). In a Taiwanese study looking at outcomes from a community-based health screening program, the authors found that fasting plasma glucose >200 mg/dL and DM was associated with the highest risk of infection and also a 3-fold higher risk of death than those without DM (93). Looking at an older population, the risk of certain infections was significantly higher in those with poor glycemic control HbA1c >8.5% compared with good glycemic control (relative risk infections ranging from 1.28-2.38) (94). Intervening to lower glucose appears to mitigate the risks. Zerr et al assessed incidence of sternal wound infection in patients with and without DM before and after implementation of a postoperative continuous IV insulin protocol to keep blood glucose <200 mg/dL. They found that lower glucose in the first 2 days postoperatively was associated with a decrease in deep wound infection from 2.4% to 1.5% (62).

 

Insulin, in both translational and clinical studies, has been suggested to have a protective effect against infection risk in those with DM (Table 3). A large surgical ICU trial assessing tight (80-110 mg/dL) versus conventional (treatment with insulin only if glucose >215 mg/dL) glycemic control using IV insulin found a lower mortality with tight glycemic control, and the greatest reduction in mortality was seen in those with sepsis leading to multi-organ dysfunction. In those treated with IV insulin, there was a significant reduction in the risk of developing sepsis (46%) (95). While these data were later brought into question by the findings of the NICE-SUGAR trial (96) which demonstrated increased mortality with intensive glycemic control using IV insulin, other studies have suggested that there is improvement in rates of infection with use of insulin (60,97) particularly in the post-cardiac surgery setting for sternal wound infections. Furthermore, the increased risk of mortality in the intensive glycemic control arm in NICE-SUGAR is felt to be related to an excess of hypoglycemia seen in the cohort compared with normal care. If improved glycemic control can be achieved without causing a significant increase in hypoglycemia, a different outcome may be able to be achieved (98). Translational studies have been able to show in vivo that T cells which lack insulin receptor expression are unable to proliferate and produce cytokines properly and that insulin enables the cells to take up nutrient which supports their function (99). As mentioned earlier chemotaxis of PMNs is impaired in patients with DM and it has been noted that provision of glucose and insulin can restore these to baseline (15).

 

While data for immune function improvement and infection using other antihyperglycemic medications are relatively sparse, there are some suggestions that therapies beyond insulin can have an immune-restorative effect (Table 3). Metformin has demonstrated an ability to increase the number and action of CD8+ tumor-infiltrating lymphocytes, resulting in improved production of cytokines IL-2, TNFα, and IFNγ (100). In a mice model with an absence of the TNF receptor-associated factor 6 (TRAF6), there is a relative inability to generate memory T cells that is related to defects in fatty acid metabolism. When TRAF6-deficient antigen specific effector T cells and TRAF6-deficient mice were exposed to metformin, there was a restoration of the production of memory T cells (101). While a number of other diabetes therapies have well-established anti-inflammatory effects (i.e., peroxisome proliferator-activated receptor-γ agonists, glucagon-like peptide-1 receptor agonists, sodium-glucose cotransporter 2 inhibitors) data are lacking as to their specific impact on immune function with regards to infection risk apart from their reduction of hyperglycemia (102-104).

 

Table 3. Anti-Hyperglycemic Agents Associated with a Reduced Risk of Infection

Insulin

- T cells in vivo which lack insulin receptor are unable to proliferate and produce cytokines due to the inability to take up nutrients

- Chemotaxis in PMNs impaired in DM which is restored with glucose and insulin

Metformin

- Increase number of tumor-infiltrating lymphocytes and improved cytokine production

- Restoration of production of memory T cells from effector T cells

 

Immunomodulating Therapies

 

There are data showing that the use of granulocyte-colony stimulating factor (G-CSF), which induces differentiation and release of PMN from marrow, is able to assist with healing in foot infections (105). While a subsequent meta-analysis did not show that the use of G-CSF was able to significantly impact the resolution or healing of wounds, there was a reduction in risk of needing amputation or surgical intervention (106).

 

While not a “medication”, physical activity has long been known to be associated with improvement in the immune system which are known to extend as well to those with DM (107). In diabetic rats, exercise was able to improve the neutrophil and lymphocyte count significantly (108).

 

CONCLUSION

 

Diabetes represents an incredibly important risk factor for infection raising the likelihood of infection for both outpatient treated conditions and those which lead to hospitalization. Beyond raising the risk for contracting an infection, prognosis is frequently worse for many of these conditions which increases the frequency of rare and life-threatening infectious processes seen in those with DM. This is the consequence of disturbances in the immune system which have been well described involving both innate and adaptive immunity. However, glucose lowering therapies appear to be able to counteract some of the increased risk of infection and worsened prognosis by improving function of immune cells. More work is needed to fully elucidate if and how newer diabetes agents may be able to reduce risk of infection.

 

REFERENCES

 

  1. Centers for Disease Control and Prevention. National Diabetes Statistics Report, 2020. Atlanta, GA: Centers for Disease Control and Prevention, U.S. Dept of Health and Human Services; 2020.
  2. Carey IM, Critchley JA, DeWilde S, Harris T, Hosking FJ, Cook DG. Risk of Infection in Type 1 and Type 2 Diabetes Compared With the General Population: A Matched Cohort Study. Diabetes Care. 2018;41(3):513-521.
  3. Abu-Ashour W, Twells LK, Valcour JE, Gamble JM. Diabetes and the occurrence of infection in primary care: a matched cohort study. BMC Infect Dis. 2018;18(1):67.
  4. Shah BR, Hux JE. Quantifying the risk of infectious diseases for people with diabetes. Diabetes Care. 2003;26(2):510-3.
  5. Korbel L, Spencer JD. Diabetes mellitus and infection: an evaluation of hospital utilization and management costs in the United States. J Diabetes Complications. 2015;29(2):192-5.
  6. Kim EJ, Ha KH, Kim DJ, Choi YH. Diabetes and the Risk of Infection: A National Cohort Study. Diabetes Metab J. 2019;43(6):804-814.
  7. Muller LM, Gorter KJ, Hak E, Goudzwaard WL, Schellevis FG, Hoepelman AI, Rutten GE. Increased risk of common infections in patients with type 1 and type 2 diabetes mellitus. Clin Infect Dis. 2005;41(3):281-8.
  8. Fine MJ, Smith MA, Carson CA, Mutha SS, Sankey SS, Weissfeld LA, Kapoor WN. Prognosis and outcomes of patients with community-acquired pneumonia. A meta-analysis. JAMA. 1996;275(2):134-41.
  9. Schuetz P, Castro P, Shapiro NI. Diabetes and sepsis: preclinical findings and clinical relevance. Diabetes Care. 2011 Mar;34(3):771-8.
  10. Esper AM, Moss M, Martin GS. The effect of diabetes mellitus on organ dysfunction with sepsis: an epidemiological study. Crit Care 2009;13:R18
  11. Forbes J.M., Cooper M.E. Mechanisms of diabetic complications. Physiological Reviews. 2013;93(1):137–188.
  12. Vergani D, Johnston C, B-Abdullah N, Barnett AH. Low serum C4 concentrations: an inherited predisposition to insulin dependent diabetes?. Br Med J (Clin Res Ed). 1983;286(6369):926-928.
  13. Jafar N, Edriss H, Nugent K. The Effect of Short-Term Hyperglycemia on the Innate Immune System. Am J Med Sci. 2016;351(2):201-11.
  14. Delamaire M, Maugendre D, Moreno M, Le Goff MC, Allannic H, Genetet B. Impaired leucocyte functions in diabetic patients. Diabet Med. 1997;14(1):29-34.
  15. Mowat A, Baum J. Chemotaxis of polymorphonuclear leukocytes from patients with diabetes mellitus. N Engl J Med. 1971;284(12):621-7.
  16. Kumar M, Roe K, Nerurkar PV, Orillo B, Thompson KS, Verma S, Nerurkar VR.. Reduced immune cell infiltration and increased pro-inflammatory mediators in the brain of Type 2 diabetic mouse model infected with West Nile virus. J Neuroinflammation. 2014;11:80.
  17. Martinez N, Ketheesan N, Martens GW, West K, Lien E, Kornfeld H. Defects in early cell recruitment contribute to the increased susceptibility to respiratory Klebsiella pneumoniae infection in diabetic mice. Microbes Infect. 2016;18(10):649-655.
  18. Collison KS, Parhar RS, Saleh SS, Meyer BF, Kwaasi AA, Hammami MM, Schmidt AM, Stern DM, Al-Mohanna FA. RAGE-mediated neutrophil dysfunction is evoked by advanced glycation end products (AGEs). J Leukoc Biol. 2002;71(3):433-44.
  19. Gupta S, Maratha A, Siednienko J, Natarajan A, Gajanayake T, Hoashi S, Miggin S. Analysis of inflammatory cytokine and TLR expression levels in Type 2 Diabetes with complications. Sci Rep. 2017 Aug 9;7(1):7633. doi: 10.1038/s41598-017-07230-8. Erratum in: Sci Rep. 2018;8(1):5768.
  20. Bagdade JD, Nielson KL, Bulger RJ. Reversible abnormalities in phagocytic function in poorly controlled diabetic patients. Am J Med Sci. 1972;263(6):451-6.
  21. Bybee JD, Rogers DE. The phagocytic activity of polymorphonuclear leukocytes obtained from patients with diabetes mellitus. J Lab Clin Med. 1964;64:1-13.
  22. Inoue S, Lan Y, Muran J, Tsuji M. Reduced hydrogen peroxide production in neutrophils from patients with diabetes. Diabetes Res Clin Pract. 1996;33(2):119-27.
  23. Repine JE, Clawson CC, Goetz FC. Bactericidal function of neutrophils from patients with acute bacterial infections and from diabetics. J Infect Dis. 1980;142(6):869-75.
  24. Alba-Loureiro TC, Hirabara SM, Mendonça JR, Curi R, Pithon-Curi TC. Diabetes causes marked changes in function and metabolism of rat neutrophils. J Endocrinol. 2006;188(2):295-303.
  25. Restrepo BI, Twahirwa M, Rahbar MH, Schlesinger LS. Phagocytosis via complement or Fc-gamma receptors is compromised in monocytes from type 2 diabetes patients with chronic hyperglycemia. PLoS One. 2014 Mar 26;9(3):e92977.
  26. Pavlou S, Lindsay J, Ingram R, Xu H, Chen M. Sustained high glucose exposure sensitizes macrophage responses to cytokine stimuli but reduces their phagocytic activity. BMC Immunol. 2018;19(1):24.
  27. Kim JH, Park K, Lee SB, Kang S, Park JS, Ahn CW, Nam JS. Relationship between natural killer cell activity and glucose control in patients with type 2 diabetes and prediabetes. J Diabetes Investig. 2019;10(5):1223-1228.
  28. Berrou J, Fougeray S, Venot M, Chardiny V, Gautier JF, Dulphy N, Toubert A, Peraldi MN. Natural killer cell function, an important target for infection and tumor protection, is impaired in type 2 diabetes. PLoS One. 2013;8(4):e62418.
  29. Beam, T.R.J., Crigler, E.D., Goldman, J.R. and Schifmann, G. Antibody response to polyvalent pneumococcal polysaccharide vaccine in diabetics. J Am Med Assoc. 1980;244:2641-2644.
  30. Diepersloot RJ, Bouter KP, Beyer WE, Hoekstra JB, Masurel N. Humoral immune response and delayed type hypersensitivity to influenza vaccine in patients with diabetes mellitus. Diabetologia. 1987;30(6):397-401.
  31. Moutschen MP, Scheen AJ, Lefebvre PJ. Impaired immune responses in diabetes mellitus: analysis of the factors and mechanisms involved. Relevance to the increased susceptibility of diabetic patients to specific infections. Diabete Metab. 1992 May-Jun;18(3):187-201.
  32. Martinez PJ, Mathews C, Actor JK, Hwang SA, Brown EL, De Santiago HK, Fisher Hoch SP, McCormick JB, Mirza S. Impaired CD4+ and T-helper 17 cell memory response to Streptococcus pneumoniae is associated with elevated glucose and percent glycated hemoglobin A1c in Mexican Americans with type 2 diabetes mellitus. Transl Res. 2014 Jan;163(1):53-63.
  33. Geerlings SE, Hoepelman AI. Immune dysfunction in patients with diabetes mellitus (DM). FEMS Immunol Med Microbiol. 1999 Dec;26(3-4):259-65.
  34. Mooradian AD, Reed RL, Meredith KE, Scuderi P. Serum levels of tumor necrosis factor and IL-1 alpha and IL-1 beta in diabetic patients. Diabetes Care. 1991;14(1):63-5. doi: 10.2337/diacare.14.1.63. PMID: 1991438.
  35. Ohno Y, Aoki N, Nishimura A. In vitro production of interleukin-1, interleukin-6, and tumor necrosis factor-alpha in insulin-dependent diabetes mellitus. J Clin Endocrinol Metab. 1993;77(4):1072-7.
  36. Reinhold D, Ansorge S, Schleicher ED. Elevated glucose levels stimulate transforming growth factor-beta 1 (TGF-beta 1), suppress interleukin IL-2, IL-6 and IL-10 production and DNA synthesis in peripheral blood mononuclear cells. Horm Metab Res. 1996;28(6):267-70.
  37. Wang X, Ota N, Manzanillo P, Kates L, Zavala-Solorio J, Eidenschenk C, Zhang J, Lesch J, Lee WP, Ross J, Diehl L, van Bruggen N, Kolumam G, Ouyang W. Interleukin-22 alleviates metabolic disorders and restores mucosal immunity in diabetes. Nature. 2014;514(7521):237-41.
  38. Price CL, Hassi HO, English NR, Blakemore AI, Stagg AJ, Knight SC. Methylglyoxal modulates immune responses: relevance to diabetes. J Cell Mol Med. 2010;14(6B):1806-15.
  39. Akash MSH, Rehman K, Fiayyaz F, Sabir S, Khurshid M. Diabetes-associated infections: development of antimicrobial resistance and possible treatment strategies. Arch Microbiol. 2020l;202(5):953-965.
  40. Casqueiro J, Casqueiro J, Alves C. Infections in patients with diabetes mellitus: A review of pathogenesis. Indian J Endocrinol Metab. 2012;16 Suppl 1(Suppl1):S27-36.
  41. Joshi N, Caputo GM, Weitekamp MR, Karchmer AW. Infections in patients with diabetes mellitus. N Engl J Med. 1999;341(25):1906-12.
  42. Peleg AY, Weerarathna T, McCarthy JS, Davis TM. Common infections in diabetes: pathogenesis, management and relationship to glycaemic control. Diabetes Metab Res Rev. 2007;23(1):3-13.
  43. Kornum JB, Thomsen RW, Riis A, Lervang HH, Schønheyder HC, Sørensen HT. Diabetes, glycemic control, and risk of hospitalization with pneumonia: a population-based case-control study. Diabetes Care. 2008;31(8):1541-5.
  44. Martins M, Boavida JM, Raposo JF, Froes F, Nunes B, Ribeiro RT, Macedo MP, Penha-Gonçalves C. Diabetes hinders community-acquired pneumonia outcomes in hospitalized patients. BMJ Open Diabetes Res Care. 2016;4(1):e000181.
  45. Kornum JB, Thomsen RW, Riis A, Lervang HH, Schønheyder HC, Sørensen HT. Type 2 diabetes and pneumonia outcomes: a population-based cohort study. Diabetes Care. 2007;30(9):2251-7.
  46. Di Yacovo S, Garcia-Vidal C, Viasus D, Adamuz J, Oriol I, Gili F, Vilarrasa N, García-Somoza MD, Dorca J, Carratalà J. Clinical features, etiology, and outcomes of community-acquired pneumonia in patients with diabetes mellitus. Medicine (Baltimore). 2013;92(1):42-50.
  47. Metlay JP, Waterer GW, Long AC, Anzueto A, Brozek J, Crothers K, Cooley LA, Dean NC, Fine MJ, Flanders SA, Griffin MR, Metersky ML, Musher DM, Restrepo MI, Whitney CG. Diagnosis and Treatment of Adults with Community-acquired Pneumonia. An Official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200(7):e45-e67.
  48. Dooley KE, Chaisson RE. Tuberculosis and diabetes mellitus: convergence of two epidemics. Lancet Infect Dis. 2009;9(12):737-46.
  49. Suaya JA, Eisenberg DF, Fang C, Miller LG. Skin and soft tissue infections and associated complications among commercially insured patients aged 0-64 years with and without diabetes in the U.S. PLoS One. 2013;8(4):e60057.
  50. Boulton AJM, Whitehouse RW. The Diabetic Foot. In: Feingold KR, Anawalt B, Boyce A, et al., eds. Endotext (Internet). South Dartmouth, MA; 2020. Available from: https://www.ncbi.nlm.nih.gov/books/NBK409609/
  51. Stevens DL, Bisno AL, Chambers HF, Dellinger EP, Goldstein EJ, Gorbach SL, Hirschmann JV, Kaplan SL, Montoya JG, Wade JC; Infectious Diseases Society of America. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clin Infect Dis. 2014;59(2):e10-52.
  52. Cheng NC, Tai HC, Chang SC, Chang CH, Lai HS. Necrotizing fasciitis in patients with diabetes mellitus: clinical characteristics and risk factors for mortality. BMC Infect Dis. 2015;15:417.
  53. Gürlek A, Firat C, Oztürk AE, Alaybeyoğlu N, Fariz A, Aslan S. Management of necrotizing fasciitis in diabetic patients. J Diabetes Complications. 2007;21(4):265-71.
  54. Misiakos EP, Bagias G, Patapis P, Sotiropoulos D, Kanavidis P, Machairas A. Current concepts in the management of necrotizing fasciitis. Front Surg. 2014;1:36.
  55. Fadini GP, Sarangdhar M, De Ponti F, et al Pharmacovigilance assessment of the association between Fournier’s gangrene and other severe genital adverse events with SGLT-2 inhibitors BMJ Open Diabetes Research and Care 2019;7:e000725.
  56. Bersoff-Matcha SJ, Chamberlain C, Cao C, Kortepeter C, Chong WH. Fournier Gangrene Associated With Sodium-Glucose Cotransporter-2 Inhibitors: A Review of Spontaneous Postmarketing Cases. Ann Intern Med. 2019;170(11):764-769.
  57. Silverii GA, Dicembrini I, Monami M, Mannucci E. Fournier's gangrene and sodium-glucose co-transporter-2 inhibitors: A meta-analysis of randomized controlled trials. Diabetes Obes Metab. 2020;22(2):272-275.
  58. Yang JY, Wang T, Pate V, Buse JB, Stürmer T. Real-world evidence on sodium-glucose cotransporter-2 inhibitor use and risk of Fournier's gangrene. BMJ Open Diabetes Res Care. 2020 Jan;8(1):e000985.
  59. Borger MA, Rao V, Weisel RD, Ivanov J, Cohen G, Scully HE, David TE. Deep sternal wound infection: Risk factors and outcomes. Ann Thorac Surg 1998;65:1050-1056.
  60. 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. Ann Thorac Surg 1999;67:352-360.
  61. Kramer R, Groom R, Weldner D, Gallant P, Heyl B, Knapp R, Arnold A.. Glycemic control and reduction of deep sternal wound infection rates: A multidisciplinary approach. Arch Surg 2008;143:451-456.
  62. Zerr KJ, Furnary AP, Grunkemeier GL, Bookin S, Kanhere V, Starr A. Glucose control lowers the risk of wound infection in diabetics after open heart operations. Ann Thorac Surg. 1997;63(2):356-61.
  63. Hammerstad SS, Grock SF, Lee HJ, Hasham A, Sundaram N, Tomer Y. Diabetes and Hepatitis C: A Two-Way Association. Front Endocrinol (Lausanne). 2015;6:134.
  64. Ito T, Shiraki K, Sekoguchi K. Metastatic gas gangrene of the leg due to acute emphysematous cholecystitis. Dig Dis Sci. 2001;46(11):2480-2483.
  65. Safwan M, Penny SM. Emphysematous Cholecystitis: A Deadly Twist to a Common Disease. Journal of Diagnostic Medical Sonography. 2016;32(3):131-137.
  66. Hirji I, Guo Z, Andersson SW, Hammar N, Gomez-Caminero A. Incidence of urinary tract infection among patients with type 2 diabetes in the UK General Practice Research Database (GPRD). J Diabetes Complications. 2012;26(6):513–516.
  67. Nitzan O, Elias M, Chazan B, Saliba W. Urinary tract infections in patients with type 2 diabetes mellitus: review of prevalence, diagnosis, and management. Diabetes Metab Syndr Obes. 2015;8:129-36.
  68. Gorter KJ, Hak E, Zuithoff NP, Hoepelman AI, Rutten GE. Risk of recurrent acute lower urinary tract infections and prescription pattern of antibiotics in women with and without diabetes in primary care. Fam Pract. 2010;27(4):379-85.
  69. Kumar S, Ramachandran R, Mete U, Mittal T, Dutta P, Kumar V, Rathi M, Jha V, Gupta KL, Sakhuja V, Kohli HS. Acute pyelonephritis in diabetes mellitus: Single center experience. Indian J Nephrol. 2014;24(6):367-71.
  70. Ronald A, Ludwig E. Urinary tract infections in adults with diabetes. Int J Antimicrob Agents. 2001;17(4):287-92.
  71. Huang TT, Tseng FY, Liu TC, Hsu CJ, Chen YS. Deep neck infection in diabetic patients: comparison of clinical picture and outcomes with nondiabetic patients. Otolaryngol Head Neck Surg. 2005;132(6):943-7.
  72. Handzel O, Halperin D. Necrotizing (malignant) external otitis. Am Fam Physician. 2003;68(2):309-12.
  73. Rodrigues CF, Rodrigues ME, Henriques M. Candida sp. Infections in Patients with Diabetes Mellitus. J Clin Med. 2019;8(1):76.
  74. Mohammadi F, Javaheri MR, Nekoeian S, Dehghan P. Identification of Candida species in the oral cavity of diabetic patients. Curr Med Mycol. 2016;2(2):1-7.
  75. Darwazeh AM, MacFarlane TW, McCuish A, Lamey PJ. Mixed salivary glucose levels and candidal carriage in patients with diabetes mellitus. J Oral Pathol Med. 1991; 20(6):280-3.
  76. Cathcart S, Cantrell W, Elewski B. Onychomycosis and diabetes. J Eur Acad Dermatol Venereol. 2009;23(10):1119-22.
  77. Thomas L, Tracy CR. Treatment of Fungal Urinary Tract Infection. Urol Clin North Am. 2015 Nov;42(4):473-83.
  78. Nyirjesy P, Sobel JD. Genital mycotic infections in patients with diabetes. Postgrad Med. 2013 May;125(3):33-46.
  79. Grandy S, Fox KM; SHIELD Study Group. Self-reported prevalence of vaginitis and balanitis among individuals with type 2 diabetes mellitus. Presented at: 70th Annual Scientific Sessions of the American Diabetes Association; June 25–29, 2010; Orlando, FL. Abstract 2369-PO.
  80. Dave CV, Schneeweiss S, Patorno E. Comparative risk of genital infections associated with sodium-glucose co-transporter-2 inhibitors. Diabetes Obes Metab. 2019;21(2):434-438.
  81. Williams SM, Ahmed SH. Improving compliance with SGLT2 inhibitors by reducing the risk of genital mycotic infections: the outcomes of personal hygiene advice. Diabetes. 2019;68(suppl 1):1224-P.
  82. Liu J, Li L, Li S, Jia P, Deng K, Chen W, Sun X. Effects of SGLT2 inhibitors on UTIs and genital infections in type 2 diabetes mellitus: a systematic review and meta-analysis. Sci Rep. 2017;7(1):2824.
  83. Fralick M, MacFadden DR. A hypothesis for why sodium glucose co-transporter 2 inhibitors have been found to cause genital infection, but not urinary tract infection. Diabetes Obes Metab. 2020 May;22(5):755-758.
  84. Jeong W, Keighley C, Wolfe R, Lee WL, Slavin MA, Kong DCM, Chen SC. The epidemiology and clinical manifestations of mucormycosis: a systematic review and meta-analysis of case reports. Clin Microbiol Infect. 2019;25(1):26-34.
  85. Rammaert B, Lanternier F, Poirée S, Kania R, Lortholary O. Diabetes and mucormycosis: a complex interplay. Diabetes Metab. 2012;38(3):193-204.
  86. Roden MM, Zaoutis TE, Buchanan WL, Knudsen TA, Sarkisova TA, Schaufele RL, Sein M, Sein T, Chiou CC, Chu JH, Kontoyiannis DP, Walsh TJ. Epidemiology and outcome of zygomycosis: a review of 929 reported cases. Clin Infect Dis. 2005;41(5):634-53.
  87. Jenks JD, Salzer HJ, Prattes J, Krause R, Buchheidt D, Hoenigl M. Spotlight on isavuconazole in the treatment of invasive aspergillosis and mucormycosis: design, development, and place in therapy. Drug Des Devel Ther. 2018;12:1033-1044.
  88. Lim S, Bae JH, Kwon HS, Nauck MA. COVID-19 and diabetes mellitus: from pathophysiology to clinical management. Nat Rev Endocrinol. 2021;17(1):11-30.
  89. Gregory JM, Slaughter JC, Duffus SH, Smith TJ, LeStourgeon LM, Jaser SS, McCoy AB, Luther JM, Giovannetti ER, Boeder S, Pettus JH, Moore DJ. COVID-19 Severity Is Tripled in the Diabetes Community: A Prospective Analysis of the Pandemic's Impact in Type 1 and Type 2 Diabetes. Diabetes Care. 2021;44(2):526-532.
  90. Holman N, Knighton P, Kar P, O'Keefe J, Curley M, Weaver A, Barron E, Bakhai C, Khunti K, Wareham NJ, Sattar N, Young B, Valabhji J. Risk factors for COVID-19-related mortality in people with type 1 and type 2 diabetes in England: a population-based cohort study. Lancet Diabetes Endocrinol. 2020;8(10):823-833.
  91. Dennis JM, Mateen BA, Sonabend R, et al. Type 2 Diabetes and COVID-19-Related Mortality in the Critical Care Setting: A National Cohort Study in England, March-July 2020. Diabetes Care. 2021;44(1):50-57.
  92. Mor A, Dekkers OM, Nielsen JS, Beck-Nielsen H, Sørensen HT, Thomsen RW. Impact of Glycemic Control on Risk of Infections in Patients With Type 2 Diabetes: A Population-Based Cohort Study. Am J Epidemiol. 2017;186(2):227-236.
  93. Chang CH, Wang JL, Wu LC, Chuang LM, Lin HH. Diabetes, Glycemic Control, and Risk of Infection Morbidity and Mortality: A Cohort Study. Open Forum Infect Dis. 2019;6(10):ofz358.
  94. McGovern AP, Hine J, de Lusignan S. Infection risk in elderly people with reduced glycaemic control. Lancet Diabetes Endocrinol. 2016;4(4):303-4.
  95. van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R. Intensive insulin therapy in critically ill patients. N Engl J Med. 2001;345(19):1359-67.
  96. NICE-SUGAR Study Investigators, Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, Bellomo R, Cook D, Dodek P, Henderson WR, Hébert PC, Heritier S, Heyland DK, McArthur C, McDonald E, Mitchell I, Myburgh JA, Norton R, Potter J, Robinson BG, Ronco JJ. Intensive versus conventional glucose control in critically ill patients. N Engl J Med. 2009;360(13):1283-97.
  97. Hruska LA, Smith JM, Hendy MP, Fritz VL, McAdams S. Continuous insulin infusion reduces infectious complications in diabetics following coronary surgery. J Card Surg. 2005;20(5):403-7.
  98. The NICE-SUGAR Study Investigators. Hypoglycemia and risk of death in critically ill patients. N Engl J Med. 2012;367:1108.
  99. Tsai S, Clemente-Casares X, Zhou AC, Lei H, Ahn JJ, Chan YT, Choi O, Luck H, Woo M, Dunn SE, Engleman EG, Watts TH, Winer S, Winer DA. Insulin Receptor-Mediated Stimulation Boosts T Cell Immunity during Inflammation and Infection. Cell Metab. 2018;28(6):922-934.
  100. Eikawa S, Nishida M, Mizukami S, Yamazaki C, Nakayama E, Udono H. Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci USA. 2015;112(6):1809-14.
  101. Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, Jones RG, Choi Y. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009;460(7251):103-7.
  102. Bonnet F, Scheen AJ. Effects of SGLT2 inhibitors on systemic and tissue low-grade inflammation: The potential contribution to diabetes complications and cardiovascular disease. Diabetes Metab. 2018 Dec;44(6):457-464.
  103. Croasdell A, Duffney PF, Kim N, Lacy SH, Sime PJ, Phipps RP. PPARγ and the Innate   Immune System Mediate the Resolution of Inflammation. PPAR Res. 2015;2015:549691.
  104. Lee YS, Jun HS. Anti-Inflammatory Effects of GLP-1-Based Therapies beyond Glucose Control. Mediators Inflamm. 2016;2016:3094642.
  105. Yonem A, Cakir B, Guler S, Azal OO, Corakci A. Effects of granulocyte‐colony stimulating factor in the treatment of diabetic foot infection. Diabetes Obes Metab 2001;3:332–337.
  106. Cruciani M, Lipsky BA, Mengoli C, de Lalla F. Are granulocyte colony-stimulating factors beneficial in treating diabetic foot infections?: A meta-analysis. Diabetes Care. 2005;28(2):454-60.
  107. Nieman DC, Wentz LM. The compelling link between physical activity and the body's defense system. J Sport Health Sci. 2019;8(3):201-217.
  108. Crespilho DM, de Almeida Leme JA, de Mello MA, Luciano E. Effects of physical training on the immune system in diabetic rats. Int J Diabetes Dev Ctries. 2010;30(1):33-7.