Abstract
Metabolic syndrome (MetS) and thyroid dysfunction are common in clinical practice. The objectives of this review are to discuss some proposed mechanisms by which thyroid dysfunctions may lead to MetS, to describe the bidirectional relationship between thyroid hormones (THs) and adiposity and finally, to resume a list of recent studies in humans that evaluated possible associations between thyroid hormone status and MetS or its clinical components. Not solely THs, but also its metabolites regulate metabolic rate, influencing adiposity. The mechanisms enrolled are related to its direct effect on adenosine triphosphate (ATP) utilization, uncoupling synthesis of ATP, mitochondrial biogenesis, and its inotropic and chronotropic effects. THs also act controlling core body temperature, appetite, and sympathetic activity. In a bidirectional way, thyroid function is affected by adiposity. Leptin is one of the hallmarks, but the pro-inflammatory cytokines and also insulin resistance impact thyroid function and perhaps its structure. MetS development and weight gain have been positively associated with thyroid-stimulating hormone (TSH) in several studies. Adverse glucose metabolism may be related to hyperthyroidism, but also to reduction of thyroid function or higher serum TSH, as do abnormal serum triglyceride levels. Hypo- and hyperthyroidism have been related to higher blood pressure (BP), that may be consequence of genomic or nongenomic action of THs on the vasculature and in the heart. In summary, the interaction between THs and components of MetS is complex and not fully understood. More longitudinal studies controlling each of all confounding variables that interact with endpoints or exposure factors are still necessary.
Keywords: blood pressure, hyperthyroidism, hypothyroidism, insulin resistance, lipids, obesity, thyrotropin
Introduction
Patients with both thyroid dysfunction and metabolic syndrome (MetS) are frequently observed in clinical practice. It is estimated that more than 20% of adult people fulfill criteria for MetS in different population studies.1–4
MetS is most often associated with obesity and consists of different metabolic risk factors that are associated with higher risk for cardiovascular disease, type 2 diabetes, and mortality.2–4 In clinical practice, there are different criteria to define MetS, but the two most common adopted for its diagnosis are based mainly on four main characteristics, as shown in Table 1.2–4 The two criteria are those recommended by the IDF (International Diabetes Federation) and by the National Cholesterol Education Program (NCEPT)–Adult Treatment Panel III (ATPIII; NCEPT–ATPIII).2–4 The four features present in both criteria are also usually reported in other defining criteria, irrespective of the adopted standard recommendations.2–4 Those four major components of MetS consist of different physiological characteristics: (a) body adiposity, especially central adiposity measured by waist circumference; (b) serum glucose levels that reflect diabetes diagnosis or the risk for its development; (c) lipid abnormalities related to metabolic risk [high serum triglycerides or low, high-density lipoprotein cholesterol (HDL-c)]; and (d) increased blood pressure (BP) levels. The presence of three or more abnormalities, concerning any of the described elements, is needed to define MetS. Additionally, some authors define MetS by the presence of abnormal serum levels of insulin or markers of insulin resistance (IR).2–4
Table 1.
IDF | NCEPT–ATPIII | |
---|---|---|
Waist circumference (⇧adiposity) |
>94 cm ♂ (European) >90 cm ♂ (Asiatic) >80 cm ♀ |
⩾102 cm ♂ ⩾88 cm ♀ |
Serum glucose | ⩾100 mg/dl or diabetes diagnoses |
⩾110 mg/dl |
Triglycerides | ⩾150 mg/dl | ⩾150 mg/dl |
HDL-c | <40 mg/dl ♂ <45 mg/dl ♀ |
<40 mg/dl ♂ <50 mg/dl ♀ |
Blood pressure | Systolic BP ⩾130 mmHg or diastolic BP ⩾85 mmHg or HBP treatment |
Systolic BP ⩾130 mmHg or diastolic BP ⩾85 mmHg |
Three or more elements are necessary for MetS diagnosis.
BP, blood pressure; HBP, high blood pressure, HDL-c, high-density lipoprotein cholesterol; IDF, International Diabetes Federation; NCEPT–ATPIII, National Cholesterol Education Program–Adult Treatment Panel III.
At the same time, the prevalence of hypothyroidism in different population surveys has been reported to be just around 8–15%.5–7 Additionally, this prevalence increases with age, reaching almost 20% of elderly subjects.7 The interest in studying possible associations between these two common disorders has increased. The knowledge that MetS may not necessarily be a consequence of thyroid dysfunction but also that thyroid dysfunction may arise from the effects of MetS has gained attention.8–14 Sectional studies have shown that the overlap between both diagnoses is common, justifying a high association between them, as shown in Table 2.14,15–105 However, as highly prevalent entities, the cause–consequence effect may not be established in these types of studies. We also observed that some studies applied a predefined criterion to establish the presence or absence of MetS and its associations with thyroid function,14,16,19,20,24–26,29,30,33,34,38,42,45,47–49,52,54,55,57,62,66–68,73,75–77,79,82,84,92–94,96–98,100–103,106,108 but the majority just evaluated the presence of one or more specific features related to MetS and not necessarily its diagnosis.
Table 2.
Author (region) | Study population | Sample size | Results |
---|---|---|---|
Rotondi et al.15 (Italy) | Class III obese and non-obese (EU, SCH, OH) | 466 | A = obese had higher TSH and lower FT4 and FT3 |
Alevizaki et al.16 (Greece) | EU subjects | 303 |
A = FT4 negatively correlated with SCF and SCF/PPF; TSH and T3 positively correlated with SCF and PPF (not in multivariate analysis) G = TSH positively correlated with HOMA-IR L = NA BP = NA |
Teixeira et al.17 (Brazil) | SCH, OH and controls from ambulatory setting of a tertiary hospital | 103 |
A = NA G = SCH with higher FPG then OH L = TG higher in SCH and OH BP = NE |
Volzke et al.18 (Germany) | Population survey (including EU and subclinical dysfunctions) | 2910 | BP = NA with TH or subclinical thyroid function |
Park et al.19 (Korea) | Euthyroid post-menopausal women | 2205 | MetS positively associated with TSH A = NA G = NA L = TG positively associated with TSH BP = DBP positively associated with TSH |
Kim et al.20 (Korea) | EU subjects | 44,196 |
A = BMI higher in the lowest quintiles (women); WC negatively correlated with FT4 (Men); G = FPG higher in the highest quintiles of FT4 L = HDL-c higher in the highest quintile of FT4 BP = higher SBP and DBP in the highest quintiles of FT4 |
Asvold et al.21 (Norway) | No previous known thyroid disease | 32,781 | A = low thyroid function positively associated with BMI |
Nam et al.22 (Korea) | Euthyroid obese and overweight pre-menopausal women | 177 |
A = T3 positively correlated with VAT, SCF and total fat, WC and BMI G = T4L positively correlated with glucose and HOMA-IR (women) L = T4L negatively correlated with HDL BP = T4L positively correlated with DBP (men) |
Friedrich et al.23 (Pomerania) | Population survey (excluding those with known thyroid diseases) | 3348 | A = TSH positively associated with BMI and WC in women (not necessarily only euthyroid subjects) |
Ambrosi et al.24 (Italy) | Obese/overweight, EU | 581 | TSH was higher and FT4 lower in MetS A = TSH increased with severity of obesity; TSH was positively correlated with BMI and WC G = TSH positively correlated with insulin and HOMA-IR and negatively with QUICKI L = dyslipidemia had higher TSH levels BP = NA |
Ruhla et al.25 (Germany) | Euthyroid volunteers | 1333 | MetS was positively associated with TSH; OR: 1.7 (1.1–2.6) A = higher BMI and more obesity in the upper range of TSH G = TSH positively correlated with HOMA-IR L = TSH positively correlated with TG BP = NE |
Garduno-Garcia et al.26 (Mexico) | Population survey (comparing EU and SCH) and correlation with serum hormone levels in the entire group and EU subjects | 3148 |
A = NA, when comparing EU and SCH, however WC positively correlated with TSH and negatively with FT4 G = HOMA-IR and insulin were positively correlated with TSH and negatively with FT4 L = TG was positively correlated with TSH and negatively with FT4; HDL was positively correlated with FT4 and negatively with TSH BP = DBP negatively correlated with FT4 |
Maratou et al.27 (Greece) | Overt and SCH hyperthyroidism in comparison with euthyroid subjects | 38 |
G = hyper and SC hyperthyroidism had higher postprandial glucose levels Hyperthyroidism had higher postprandial insulin levels HOMA-IR was increased in overt and SC hyperthyroidism |
Marzullo et al.28 (Italy) | EU, obese subjects | 952 |
A = BMI was positively correlated with TSH and negatively with FT4 G = NA L = HDL positively correlated with FT4 BP = NE |
Lai et al.29 (China) | SHC and controls from a survey and study of correlations between serum hormone levels and endpoints in EU subgroup | 1534 | TSH higher in MetS A = TSH higher in obese/overweight; BMI positively associated with TSH; WC correlated with TSH G = neither FPG nor HOMA-IR were associated with thyroid status L = TSH higher in subjects with abnormal TG; no association between TG and TSH BP = TSH higher in HBP; no correlation with TSH |
Lee et al.30 (Korea) | EU subjects | 7270 | MetS diagnosis was associated with upper reference range of serum TSH A = BMI positively associated with TSH L = TSH correlated with TG in multivariate analysis |
Liu et al.31 (China) | Population survey (EU × SCH) | 6339 | The number of MetS components did not differ between groups A = WC was associated with SCH G = NA TG = higher in SCH BP = higher in SCH |
Diez and Iglesias32 (Spain) | Euthyroid obese, overweight and controls | 778 | A = TSH higher in obesity and positively correlated with BMI (not confirmed after excluding TPO-Ab+) |
Taneich et al.33 (Japan) | Euthyroid diabetic patients | 301 |
A = FT4 positively correlated with BMI and VFA; FT3 positively correlated with BMI and VFA G = T3 negatively correlated with HbA1c; TSH negatively correlated with FPG and HbA1c L = T3 negatively correlated with TG BP = FT4 positively correlated with DBP; T3 negatively correlated with DBP and SBP |
Park et al.34 (Korea) | EU subjects | 5998 |
A = WC was positively associated with FT4 and negatively with BMI G = NA L = TG positively associated with TSH and negatively with FT4; inverse associations for HDL BP = FT4 positively associated to DBP and SBP (for DBP, remained significant in multivariate analysis) |
Kitahara et al.35 (USA) | Euthyroid subjects from NHANES | 3114 | A = BMI and WC were positively associated with TSH and FT3 but not with FT4 |
Zhang et al.36 (China) | Euthyroid subjects from population survey | 1322 |
A = higher WC, % body fat and BMI in women, with all three parameters correlated with TSH G = NA L = NA BP = NA |
Tamez-Pérez et al.37 (Spain) | Diabetic and control subjects | 5161 | G = OR for hypothyroidism in diabetic patients was 3.45 (95% CI 2.51–4.79; p <0.0001) when comparing the rate of hypothyroidism in diabetic group and non-diabetic group |
Tarcin et al.38 (Turkey) | Obese patients without overt thyroid dysfunction | 211 | MetS had higher T3 and T4 levels; however, lower FT3/FT4; no correlation with TSH A = positive correlation between WC and T3 G = T3 positively correlated with FPG and HOMA-IR; however, lower FT3/FT4 L = lower HDL-c in TSH >2.5 BP = positive correlation between FT4 and DBP and SBP |
Aljohani et al.39 (Saudi Arabia) | SCH × controls from an endocrinology unit | 94 |
A = BMI higher in SCH G = NE L = TG higher in SCH BP = NE |
Kwarkernaak et al.40 (Europe) | Obese subjects and controls | 74 |
A = BMI positively associated with TSH in obese G = NA L = NA BP = NE |
Solanki et al.41 (India) | Volunteers with TSH between 0.4 and 10.0 | 417 | A = TSH increases with BMI |
Oh et al.42 (Korea) | Euthyroid young females (18–39 years) | 2760 | MetS was more frequent in TSH >2.5 A = WC positively associated with TSH G = NA in multivariate analysis L = TG positively associated with TSH BP = SBP and DBP positively associated with TSH |
Kouidhi et al.43 (Tunisia) | Overweight, obese and controls with TSH in the normal range | 108 |
A = TSH higher in overweight and obese and FT4 lower; BMI WC positively correlated with TSH; WC negatively with FT4 G = insulin and HOMA-IR positively correlated with TSH |
Karthlich et al.44 (India) | Women with SCH and euthyroid controls | 60 |
A = NA G = NA L = HDL lower and TG higher in SCH BP = SBP lower in SCH |
Muscogiuri et al.45 (Italy) | EU without DM | 60 |
A = overweight and obesity were associated with higher TSH; TSH was correlated with VAT G = positive correlation between TSH and glucose uptake: not confirmed in multivariate analysis L = NA BP = NE |
Vyakaranam et al.46 (India) | Euthyroid subjects and SCH | 2037 |
A = NA G = TSH positively and FT3 negatively correlated with insulin; FPG higher in SCH L = NE BP = NE |
Roef et al.47 (Italy) | Diabetic patients | 490 |
A = BMI and WC were positively associated with FT3, TT3, FT3/FT4 and negatively with FT4 G = FPG positively associated with FT3, TT3 and FT3/FT4 L = TG positively associated with TSH, FT3, TT3, FT3/FT4 and negatively with FT4; HDL-c negatively with FT3 and TT3 BP = positively associated with TSH, FT3, TT3 and FT3/FT4 |
Bakiner et al.48 (Turkey) | Obese, overweight and controls with serum TSH between 0.4 and 10.0 | 1097 | No association with MetS A = NA G = NA L = NA BP = NA |
Mamtani et al.49 (Mexico and USA) | Population study from Mexico and NHANES | 2540 |
A = thyroid function index was positively associated with BMI, WC, and central obesity G = diabetes diagnosis positively associated with thyroid function index (not confirmed in multivariate analysis) L = TG and HDL were not significantly associated BP = NA |
Ren et al.50 (China) | Population survey (euthyroidism) | 1180 |
A = BMI, fat mass and WC positively associated with FT3 G = FPG and HOMA positively associated with FT3 L = HDL negatively associated with FT3 BP = NE |
Giandalia et al.51 (Italy) | DM2 with euthyroidism | 490 |
A = BMI, high WC and visceral adiposity was more prevalent in the highest quartiles of TSH G = NA L = high TG more prevalent in the highest quartiles of TSH BP = HBP more prevalent in the highest quartiles of TSH |
Sakurai et al.52 (Japan) | Euthyroid employers | 2037 | A = positive association between TSH and BMI |
Shin et al.53 (Korea) | EU, non-diabetics | 6241 | IR was associated with highest quartiles of FT4 A = BMI and WC was negatively correlated with FT4 (not in multivariate) G = HOMA-IR was negatively correlated with FT4 that was also slightly correlated with FPG (not in multivariate analysis) L = TSH was slightly and positively correlated with HDL-c and negatively with FT4 (in multivariate analysis, a slightly positive association was found between FT4 and TSH in men) BP = NA |
Udenze et al.54 (Nigeria) | Staff from college of medicine | 150 | Sick euthyroid syndrome was more common in patients with MetS |
Shinkov et al.55 (Bulgaria) | Population survey (euthyroid) | 2401 | More MetS in the highest quartile A = NA G = NA L = low HDL-c and high TG more frequent in the highest quartile BP = NA |
Gierach and Junik56 (Poland) | Patients with MetS (comparing hypothyroid × EU) | 441 |
A = WC did not differ between hypothyroid and euthyroid G = FPG did not differ L = HDL higher in Hypothyroid and TG higher (only in women’s subgroups) BP = NA |
Aras et al.57 (Turkey) | Obese and controls | 70 |
A = FT3/FT4 positively associated with WC; TSH higher in higher BMI G = FT3/FT4 positively correlated with FPG L = FT3/FT4 positively correlated with TG and tendency for negative association with HDL BP = FT3/FT4 tended to be positively correlated with SBP |
Sieminska et al.58 (Poland) | Post-menopausal women (EU × SCH) | 372 |
A = higher WC in SCH G = NA L = higher TG BP = higher SBP and DBP in SCH |
Ozdemir et al.59 (Turkey) | Hypo-, hyperthyroid and control subjects | 63 |
A = low BMI in Hyperthyroidism G = HOMA β higher in hypothyroidism; FPG higher in hyperthyroidism L = higher TG in hypothyroidism BP = NE |
Lambrinoudak et al.60 (Greece) | Healthy women, post-menopausal | 194 | A = FT4 was lower in high-fat mass, FT3 was higher; Fat mass increased in the highest quartiles of FT3; TSH was positively correlated with BMI |
Betry et al.61 (France) | Hospitalized obese patients for check-up | 800 | A = TSH positively associated with BMI |
Petrosyan62 (Armenia) | All with MetS | 120 |
A = BMI higher in TSH >2.5 G = HbA1c higher in TSH >2.5 L = TG higher in TSH >2.5 BP = DBP higher in TSH >2.5 |
Meng et al.63 (China) | Community-based health-check investigation (without known thyroid disease) | 13,855 |
A = BMI and WC negatively correlated with FT4 (women) and TSH (men), also positively with FT3 (men) G = FPG positively correlated with FT4 and negatively with FT3 (women) L = HDL-c negative correlation with FT3 and positive with FT4 BP = positive correlation with TSH and FT4 and negative with FT3 (women) |
Aksoy et al.64 (Turkey) | SCH in LT4 use | 104 |
A = BMI was not associated with TSH G = HOMA-IR was not associated with TSH L = NA BP = NA |
Maskey et al.65 (India) | Diabetic patients | 271 |
A = BMI higher in diabetic patients with hypo G = insulin use and inadequate diabetic control was more frequent among hypothyroid patients L = HDL-c and TG higher in hypothyroidism BP = did not differ |
Bensenor et al.66 (Brazil) | Civil servants recruited in a survey (TSH evaluated in quintiles in the whole group and only in euthyroid subjects) | 10,935 | High TSH quintile was associated with IR/MetS A = higher WC and BMI G = FPG higher in low quintile with opposite effect on HOMA-IR L = higher TG in high quintile BP = NA |
Nozarian et alk.67 (Tehran) | Euthyroid patients with MetS and controls (ATPIII) | 82 | TSH, FT3 and FT4 did not differ between groups with or without MetS TSH in the upper range was associated with higher risk of MetS in multivariate analysis. A = TSH not related to TSH L = HDL not related to TSH in regression however associated with TSH >2.5–5.0 mIU/l |
Lee et al.68 (USA) | Framingham cohort: euthyroid subjects | 3483 |
A = TSH positively associated with BMI and SCF; FT4 was negatively associated with obesity and VAT G = NE L = TSH positively and FT4 negatively associated with TG BP = not associated in multivariate analysis |
Peixoto de Miranda et al.69 (Brazil) | Civil servants recruited in a survey (TSH evaluated in quintiles considering the whole group) | 12,284 | MetS did not differed A = BMI higher in the 5th quintile of TSH (including OH diagnosis) G = IR more frequent in 5th quintile of TSH L = high TG more frequent among subjects in the upper quintile BP = NA |
Kim et al.70 [South Korea] | Euthyroid middle-aged subjects | 13,496 | Higher risk for MetS in highest quartile of T3; no association with T4 or TSH A = TSH was lower; T4 and T3 was higher in obesity and overweight G = TSH was negatively associated with FPG and HbA1c; T3 was positively associated with glycemia L = HDL was negatively associated with TSH BP = T3 and T4 positively associated with SBP |
Wang et al.71 (Taiwan) | Non-obese, euthyroid, young women | 229 | TSH higher in the presence of IR |
Temizkan ert al.72 (Turkey) | Obese euthyroid patients | 5300 |
A = NA G = FPI and HOMA-IR higher in the highest quartile TSH L = NA BP = NE |
Kathiwada et al.73 (Nepal) | Patients with MetS (SCH × EU) | 169 |
A = WC was lower in EU (comparing to SC and overt hyperthyroidism); weak positive correlation between TSH and BMI and negative between BMI and FT3 and FT4 G = NA L = TSH negatively correlated with HDL BP = NA |
Tiller et al.74 (Europe) | Population surveys | 16,902 | A = TSH positively associated with BMI, WC and WC/height |
Xu et al.75 (China) | Population survey, EU | 2356 |
A = higher BMI in the upper-half serum TSH G = FPG higher in the upper-half serum TSH L = NA BP = NA |
Mehran et al.76 (Iran) | Community-based study | 5422 | Highest prevalence of MetS in hypothyroidism A = higher BMI in overt hypothyroidism G = SC hyper had higher FSG and frequency of hyperglycemia. FT4 negatively associated with FSI L = HDL-c lower in SC hyper, and TG higher in OH BP = NA |
Jayanthi et al.77 (India) | Tertiary care hospital: obese, OW and diabetic patients | 92 |
A = NE G = HOMA-IR negativelly correlated with TSH and positively with FT4; HbA1c negatively with FT4 and positive with TSH L = T3 was positively associated with HDL-c in obese diabetic patients BP = NE |
Wolffenbuttel et al.14 (Netherlands) | Population survey (EU subjects) | 26,719 |
A = WC positively associated with FT3 and FT3/FT4 and negatively with FT4 in multivariate analysis G = FPG positively associated with FT3 and FT3/FT4 and negatively with FT4 in multivariate analysis L = HDL-c positively associated with FT4 and negatively with FT3 and FT3/FT4 in multivariate analysis; TG positively associated with TSH and negatively with FT4 and positively with FT3/FT4 BP = DBP and SBP positively associated with FT3 and FT3/FT4 |
Al-Musa78 (Saudi Arabia) | Primary healthcare | 278 | A = TSH higher in obese (FT3 and FT4 did not differ) |
Lozanov et al.79 (Bulgaria) | Hospitalized | 118 | TSH in upper reference had more MetS diagnosis A = BMI was associated with higher TSH G = hypothyroid patients had higher insulin levels at 120 min of OGTT |
Kar and Sinha80 (India) | Hypothyroid patients and controls | 80 | HOMA-IR higher in hypothyroidism |
Gutsh et al.81 2017 (India) | Hospital-based cross-sectional study | 200 | TSH was higher and FT4 lower in MetS |
Ferrannini et al.82 (Italy) | Multicenter cohort with clinically healthy participants (sub-analysis of euthyroid participants) | 1018 | Insulin resistance was independently associated with higher FT3 A = BMI and WHR higher in the highest FT3 quartiles G = NA; higher insulin levels in highest quartiles L = TG higher and HDL-c lower in the highest FT3 quartiles BP = increase in higher quartiles |
Witte et al.83 (Germany) | Patients attending specialist consultations (87.9% euthyroid) | 1719 | A = NA between VAT and TSH |
Racaitaianu et al.84 (Romania) | Obese non-diabetic participants | 82 | G = TSH was higher when HOMA-IR >2.5; FT4 did not differ |
Rahbar et al.85 (Iran) | Euthyroid | 140 |
A = higher BMI in highest TSH levels G = NE L = NA |
Valdes et al.86 (Spain) | Population survey | 3928 | Higher TSH levels in morbidly obese patients |
Sami et al.87 (Pakistan) | Obese | 127 | A = high frequency of SCH in obesity |
Jang et al.88 (Korea) | Population survey without known thyroid disease (sub-analysis of euthyroid participants) | 1423 |
A = WC tended to be negatively associated with FT4 G = FPG positively associated with FT4 L = TG positively with TSH and negatively with FT4 BP = not associated |
Liu et al.89 (China) | Non-obese EU patients from endocrinology department of a university hospital | 5608 |
A = BMI positively correlated with FT3 and negatively with FT4 G = FPG and HOMA-IR positively correlated with FT3 and FT4 L = HDL negatively with FT3 and FT4 BP = NE |
Liu et al.90 (China) | Community-based health-check program | 13,505 | A = NA |
Zhou et al.91 (Taiwan) | Patients from annual examination of a health examination center at hospital | 12463 | In multivariate analysis TSH was positively associated with MetS diagnosis G = diabetes or pre-diabetes Dx was not associated BP = HBP Dx was associated with higher TSH |
Liu et al.92 (Taiwan) | Patients from annual examination of a health examination center at hospital (EU versus SCH) | 15,943 | SCH positively associated with MetS and number of its components A = WC higher in SCH (men) G = NA L = TG higher in SCH (women) BP = SBP higher in SCH and DBP also higher (women) |
Bermúdez et al.93
(Venezuela) |
Participants without thyroid diseases from a sectional study for MetS screening | 391 | Elements of MetS was more frequent in SCH A = WC did not differ between SCH and EU G = diabetes Dx as hyperglycemia was more common in SCH L = NA BP = NA |
Mousa et al.94 (Turkey) | Euthyroid under LT4 | 301 |
A = TSH correlated positively with BMI and FT3 with VAT G = TSH, FT4 and FT3 positively correlated with FPG and HOMA L = NA BP = NE |
Amouzegar et al.95 (Iran) | Population survey with euthyroid participants | 1938 | FT4 negatively associated with metabolic obese subjects |
Wang et al.96 (USA) | Population survey (NHANES) | 1560 | IR was positively associated with low FT4 and negatively with low FT3 and TT3 |
Hamlaoui et al.97 (Algeria) | Patients attending specialist consultations (hypo, hyper and EU) | <100 |
A = hypothyroidism had higher BMI and WC; more abdominal obesity G = NA L = lower HDL in hyperthyroidism BP = more hypertension in Hyper and higher SBP |
Delitala et al.98 (Italy) | Population survey (sub-analysis of euthyroid subjects) | 6148 | Positive association between components of MetS with TSH in euthyroid males and women without known thyroid disease A = FT4 negatively associated with WC G = FPG positively associated with FT4 L = TSH positively associated with TG; FT4 positively associated with HDL-c BP = DBP positively associated with FT4 |
De Vries et al.99 (Netherlands) | Euthyroid subjects with high risk for CV disease | 5542 | G = NA between TSH and DM diagnosis |
Chang et al.100 (China) | From a self-paying health examination program | 24,765 | Metabolic syndrome positively associated with TSH A = BMI, BF and WC associated with higher TSH G = TSH positively correlated with HbA1c, fasting insulin, HOMA-IR and HOMA- β; high HbA1c, hyperinsulinemia, high HOMA-β, increased HOMA-IR occurred more when TSH >2.9 L = high TG and low HDL-c associated with higher TSH BP = positively associated with TSH |
Xu et al.101 (China) | Euthyroid subjects from check-up evaluations | 16,975 |
A = overweight and obese had high serum FT3, high FT3/FT4 and low FT4. G = FBG negatively associated with FT4 and positively with TSH L = HDL positively associated with TSH and negatively with FT3; TG positively associated with FT3 and negatively with FT4 BP = SBP and DBP positively associated with FT3 |
Kim et al.102 (Korea) | Community survey (TSH = 0.6–6.68) | 13,873 | Non-obese subjects without MetS had lower TSH and higher FT4 |
Zhang et al.103 (China) | Community survey (euthyroidism) | 3590 | A = BMI increased with higher TSH |
Lertrit et al.104 (Thai) | Population survey | 2242 |
A = BMI positively associated with TSH and negatively with FT4 in multivariate analysis G = FPG positively associated with FT4 in multivariate analysis |
Raposo et al.105 (Portugal) | Population survey | 486 | MetS diagnosis was positively associated with FT3 A = NA G = FPG not associated; however, HOMA = IR and serum insulin were positively associated with FT3 L = TG positively associated with FT3 BP = NA |
A, adiposity; ATPIII, Adult Treatment Panel III; BMI, body mass index; BP, blood pressure; CI, confidence interval; DBP, diastolic blood pressure; DM, diabetes mellitus; Dx, diagnosis; EU, euthyroid; FPG, fasting plasmatic glycaemia; FPI, fasting plasmatic insulin; FSG, fasting serum glucose; FSI, fasting serum insulin; FT3, free triiodothyronine; FT4, free thyroxine; G, glucose metabolism; HbA1c, glycosylated hemoglobin; HBP, high blood pressure; HDL-c, high-density-lipoprotein cholesterol; HOMA-IR, Homeostatic Model Assessment of Insulin Resistance index; IR, insulin resistance; L, lipid profile; MetS, metabolic syndrome; NA, no association; NE, not evaluated; NHANES, National Health and Nutrition Examination Survey; OGTT, overload glucose tolerance test; OH, overt hypothyroidism; OR, odds ratio; PPF, preperitoneal fat; SBP, systolic blood pressure; SCF, subcutaneous fat; SCH, sub-clinical hypothyroidism; SC hyper, sub-clinical hyperthyroidism; T3, triiodothyronine; TG, triglycerides; TH, thyroid hormone; TSH, thyrotropin; TT3, total triiodothyronine; VAT, visceral adipose tissue; WC, waist circumference; WHR, waist-to-hip ratio; QUICKI, quantitative insulin sensitivity check index; TPO-Ab+, positive antibodies against thyroperoxidasis on serum; VFA, visceral fat area; HSC, is the same as SCH (subclinical hypothyroidism); T4L, is the same as FT4 (Free Thyroxine); LT4, levothyroxine; OW: overweight.
Cohort studies also do not seem to be capable of showing that a unidirectional pathway justifies this association,17,18,28,34,68,74,76,82,95,99,106–117 and the hypotheses that both thyroid dysfunction leads to MetS and that this condition also influences thyroid function has gained credibility.9–13 THs, and also some of their metabolites, regulate metabolic rate, leading to variations in weight gain and adiposity.9–11,13 Additionally, THs also act on central regulation of appetite control and sympathetic activity. In the opposite direction, thyroid function is affected by adiposity, with leptin having important modulatory effects.9–11,13 Also, pro-inflammatory cytokines related to obesity and IR may impact thyroid function and perhaps its structure.9,11–13 Table 3 summarizes the results of longitudinal studies done over the past decade regarding the association between thyroid function and MetS diagnosis, or even different MetS components. For this purpose, we did not include a detailed analysis of studies focusing on the effect of bariatric surgery on thyroid, even though a recent meta-analysis found that patients who underwent bariatric surgery exhibited a reduction of TSH, free triidothyronine (FT3) and triidothyronine (T3) levels after surgery.12
Table 3.
Author (region) | Follow-up | n | Population | Main results |
---|---|---|---|---|
Marzullo et al.28 (Italy) | 4 months | 100 | Obese submitted to diet | A = weight loss was associated with reduction in TSH and FT3; also, with increase in FT4 levels |
Ferrannini et al.82 (Italy) | 3 years | 940 | Euthyroid subjects | G = baseline FT3 and FT4 were positively associated with increases in FPG and decrease in insulin sensitivity measured by euglycemic clamp (CLAMP) |
Nada106 (Saudi Arabia) | Post-normalization | 42 | Women with OH | G = after LT4 replacement, there was no significant change in FBG or HOMA-IR as compared with before starting treatment, while fasting insulin significantly increased |
Amouzegar et al.95 (Iran) | 9 years | 1938 | Population-based cohort study |
A = increment in FT4 levels was accompanied by decreased risk of metabolically healthy obesity and metabolically healthy, normal-weight phenotypic development TSH increment was positively associated with metabolically unhealthy, normal-weight phenotypic development |
Mehran et al.76 (Iran) | 3 years | 2393 | Frameworks of a community-based study | BP = FT4 was associated with higher odds of high BP after adjusting for age, sex, smoking, BMI, and HOMA-IR; no significant associations between TSH and BP |
Langén et al.107 (Finland) | 11 years | 2486 | Population-based cohort | L = no association with TSH |
Langén et al.108 (Finland) | 11 years | 3453 | Population-based cohort | BP = TSH did not predict incident hypertension and was inversely associated with change in SBP and DBP in men |
Volzke et al.18 (Germany) | 5 × years | 2910 | Population-based cohort |
A = NE G = NE L = NE BP = SC hyper was not associated with changes in BP or incident hypertension in multivariate analysis |
De Vries et al.99 (Europe) | 7.6–5.9 years | 5542 | Metanalysis of population surveys | G = no more risk for incident DM |
Itterman et al.109 (Europe) | 5 years | 10,048 | Population survey | BP = High TSH was not associated with incident HBP |
Liu et al.110 (USA) | 2 years | 811 | Obese and overweight submitted to diet protocols | A = Baseline FT3 and FT4 predicted weight loss; FT3 and TT3 were positively associated with changes in body weight, BP, G, insulin, and TG; without associations with FT4 or TSH |
Eray et al.111 (Turkey) | 6 months | 129 | Obese before and after pharmacological treatment | No effects on TSH, FT3 and FT4 |
Teixeira et al.17 (Brazil) | 1 year | 103 | Ambulatory from a tertiary hospital (EU, SCH, OH) |
A = no significant changes in BMI and BF% G = no significant changes in HOMA-IR L = reduction in TG with OH treatment BP = NE |
Park et al.34 (Korea) | 3 years | 5998 | EU, SCH, SC hyper | Changes in TSH was positively associated with MetS development A = WC was not associated with changes in TSH or FT4 G = glucose and HOMA-IR were positively associated with changes in TSH L = TG was positively associated with changes in TSH and negative with FT4 BP = positively associated with changes on FT4 and TSH |
Chen et al.112 (Taiwan) | 11 years | 38,200 | Hypo-, hyperthyroid participants and controls | G = there was significantly higher occurrence of T2D in the hypothyroidism and also hyperthyroidism groups than in the control group |
Lee et al.68 (USA) | 6.1 | 2912 | EU participants |
A = NA with TSH or FT4 G = NA with TSH or FT4 L = NA with TSH or FT4 BP = NA with TSH or FT4 |
Tiller et al.74 (Europe) | 5 years | 2912 (713 for body composition) | Population-based cohort studies |
A = serum TSH at baseline was inversely associated with anthropometric changes (WC, BMI); however, with a positive association with TSH changes G = NE L = NE BP = NE |
Chang et al.113 (Taiwan) | 4.2 years | 66,822 | EU at baseline | Higher risk for SCH development in MetS (HR = 1.12) A = NA G = NA L = higher risk for SCH development when high TG BP = an increased risk of SCH was associated with high BP |
Caixàs et al.114 (Spain) | Post-normalization | 51 | Hyper- and hypothyroid patients (pre- and post-treatment) | G = Patients with hyperthyroidism showed higher glucose, insulin concentrations and HOMA-IR than their controls; after normalization of thyroid function, glucose and HOMA-IR decreased to the normal range |
Chaker et al.115 (Netherlands) | 7.9 years | 8452 | Population survey |
G = risk for developing diabetes 1.09 times higher for every doubling of TSH levels; higher FT4 levels within the normal range were associated with a decreased risk of diabetes; In participants with pre-diabetes, the associated risk of developing diabetes was 1.13 times higher for every doubling of TSH levels The risk of progression from pre-diabetes to diabetes was higher with low–normal thyroid function (HR 1.32; 95% CI, 1.06–1.64 for TSH and HR 0.91; 95% CI, 0.86–0.97 for FT4) Absolute risk of developing T2D in participants with pre-diabetes decreased from 35% to almost 15% with higher FT4 levels within the normal range |
Bjergved et al.116 (Denmark) | 11 years | 1577 | Population survey | A = positive association between BMI changes and TSH changes |
Soriguer et al.117 (Spain) | 6 years | 479 | 784 | A = obesity development was related to higher concentrations of FT3 and FT4; weight gain with FT3 |
A, adiposity; BMI, body mass index; BP, blood pressure; BF, body fat; CI, confidence interval; DBP, diastolic blood pressure; DM, diabetes mellitus; EU, euthyroid; FPG, fasting plasmatic glycaemia; FT3, free triiodothyronine; FT4, free thyroxine; G, glucose metabolism; HBP, high blood pressure; HDL-c, high-density-lipoprotein cholesterol; HOMA-IR, Homeostatic Model Assessment of Insulin Resistance index; HR, hazard ratio; IR, insulin resistance; L, lipid profile; MetS, metabolic syndrome; NA, no association; NE, not evaluated; OH, overt hypothyroidism; SBP, systolic blood pressure; SCH, sub-clinical hypothyroidism; SC hyper, sub-clinical hyperthyroidism; T2D, type 2 diabetes; TG, triglycerides; TSH, thyrotropin; TT3, total triiodothyronine; WC, waist circumference; QUICKI, quantitative insulin sensitivity check index; TPO-Ab+, positive antibodies against thyroperoxidasis on serum; VFA, visceral fat area; HSC, is the same as SCH (subclinical hypothyroidism); T4L, is the same as FT4 (Free Thyroxine); LT4: levothyroxine.
In this review, we will discuss some proposed mechanisms by which thyroid dysfunctions may lead to MetS development, and not solely focus on the diagnosis of its complete presentation but also the way in which TH may influence each one of the four main features (or components) of this important syndrome. The consequences of augmenting adiposity, which is a highly prevalent marker of MetS, may also interfere with thyroid function will also be described. Finally, a list of recent studies enrolling humans and intending to evaluate possible associations between thyroid function and MetS will be present. For this purpose, we will focus on research excluding specific populations, like pediatric or elderly subjects, and also patients with other diagnoses, such as polycystic ovary syndrome. Additionally, we do not intend to review data on patients that underwent bariatric surgery.
Molecular mechanism of action of thyroid hormones: general overview
THs act on several target peripheral tissues via several mechanisms. Briefly, thyroxine (T4), which is the main product of the thyroid gland, is converted to the active hormone, T3, an enzymatic reaction catalyzed by type 1 (D1) or type 2 5′deiodinases (D2). T4 and T3 can be inactivated by type 3 5-deiodinase (D3). T4 and T3 enter cells through specific membrane transporters, and T3, originating from the circulation or from intracellular conversion of T4 to T3, binds to TH receptors, subtypes 1, β1 or β2, located at the nucleus to regulate the transcriptional activity of target genes.118 This is the canonical pathway; however, recently, other non-classical pathways have been reported. TH actions may be mediated by cytoplasmic or mitochondrial TH receptors (TR), or through binding to unspecific membrane proteins that activate intracellular signaling cascades.118–121 These non-canonical signaling pathways have been reported to be especially important to the cardiometabolic effects of thyroid hormones.121 In that elegant study, the authors employed genetically manipulated mice to differentiate between T3 effects mediated by the canonical and non-canonical pathways. They showed that the acute hypoglycemic effect of T3 is dependent on TRβ but does not require deoxyribonucleic acid binding. Its action involves activation of the phosphatidylinositol 3-kinase (PI3K) signaling cascade. The same non-canonical signaling pathway is involved in a T3-lowering effect in serum and hepatic triglycerides. In addition, T3 actions in metabolic rate and energy expenditure, as well as in the exogenous control of heart rate have important contributions of the non-canonical signaling pathways.121
It is also important to mention that tissue responsiveness to TH may vary with age and sex, which may be related to tissue-specific alterations in T4 to T3 conversion.122–124 The interplay between age and sex are particularly interesting in TH-induced changes in body weight and energy expenditure in mice, with sex modifying the response of TH differently in old males compared with old females.122–124
Mechanisms by which thyroid function may interact with components of metabolic syndrome
TH may be involved in each one of the four major components of MetS via several mechanisms. This involvement is not necessarily unidirectional, since target tissues of TH may also be involved with thyroid function. TH actions lead to specific effects that influence endpoints regarding body adiposity, glucose or lipid levels, and BP.11,120,125–127 In this way, all four features of MetS may be influenced by TH levels as separately described in specific following sections.
In summary, adiposity may be the consequence of the role of THs (or its metabolites) on the regulation of metabolic rate, appetite control or even sympathetic activity.9,11,13 This sympathetic stimulus by THs also influences glucose and lipid metabolism as it impacts cardiovascular system regulation.9,11–13 Hyperglycemia may be the consequence of reduced glucose uptake in hypothyroidism or the consequence of increased glucose liver production in hyperthyroidism.128 Glucose-stimulated insulin secretion and insulin degradation are also regulated by THs.128 Dyslipidemia may be related to thyroid function, since THs also act stimulating both lipid synthesis and degradation.129 Finally, high BP (HBP) may be the consequence of TH action on the vasculature and in the heart by TR-mediated gene regulation at the nucleus or via other non-classical pathways at the cytoplasmatic and cellular membrane levels.130
However, it is notable that the augmentation in adiposity, especially central adiposity, which is one of the hallmarks of MetS, appears to generate an increase in several hormones, cytokines, and other compounds that influence thyroid function via different pathways.131,132 The proposed mechanisms involved in these actions will be summarized in the next sections.
Thyroid hormones influencing adiposity
Adiposity gain or loss depends primarily on the balance between energy expenditure (EE) and energy intake (EI). Resting EE (REE) is solely used in the cellular process to maintain life.133 EE can be stimulated by physical activity or acceleration of different metabolic processes, resulting in heat production (facultative thermogenesis).134 The balance between EE and EI depends mainly on satiety control, sympathetic nervous system (SNS) activity, and the endocrine system. THs are strong regulators of the metabolic rate with consequent effects on different outcomes, including adiposity.135 However, as previously described, the relationship between TH and adiposity is bidirectional, since TH and also thyroid-stimulating hormone (TSH) levels have effects on adiposity, which in turn may act on thyroid function and perhaps on the structure of this gland.136,137 Adiposity leads to production of several hormones, cytokines, and other compounds that influence thyroid function, as described in the next sections.
THs, especially T3 produced by enzymatic reaction catalyzed by type 1 (D1) or type 2 5′deiodinases (D2), are enrolled in controlling metabolic rate by several mechanisms, as explained in the following sections of this manuscript. In summary, they exert direct effects on adenosine triphosphate (ATP) utilization, uncoupling synthesis of ATP, mitochondrial biogenesis and have inotropic and chronotropic effects on body. THs also act controlling core body temperature, appetite, and sympathetic activity. Additionally to T4 and T3, other TH metabolites exert similar effects.138,139 It has been demonstrated that 3,5 diiodo-L-thyronine (T2) prevents high-fat-diet-induced adiposity by means of increasing EE and promoting anti-adipogenic and anti-lipogenic pathways in white adipose tissue (WAT).138,139 Also, studies have demonstrated that decarboxylated TH molecules, termed thyronamines, when given to animals, lead to metabolic effects that generally oppose the direction of T3. Thyronamines are primarily produced in the thyroid, but there is evidence that they may be produced in other tissues.139–141 The physiological and clinical relevance of TH metabolites is under intense investigation.139–141
The thermogenic effects of TH, especially T3, are well known, and hyperthyroid patients have an increase in heat production and are heat intolerant. Hyperthyroid patients are opposite to hypothyroid patients, who produce less heat and are cold intolerant.142 After thyroid hormone administration there is an increase in oxygen consumption in most tissues.142 THs cause a direct increase in adenosine triphosphate (ATP) utilization leading to acceleration of anabolic and catabolic pathways in the macronutrient metabolism, such as lipolysis/fatty-acid oxidation and increased protein turnover.143 In addition, THs stimulate the sodium/potassium (Na+/K+) ATPase and the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) that mediate ion transport through membranes, processes that require ATP utilization, leading to increasing of it consumption and contributing to thermogenesis.144 Therefore, thyroid hormone increased the utilization of energy reserves, such as lipids from the adipose tissue.
Another mechanism by which TH may increase the REE is related to the hormones’ inotropic and chronotropic effects, exerted in conjunction with the SNS, since it is well known that part of REE is related to cardiac function.145
TH actions at the mitochondria are very important in thermogenesis. In addition to promoting mitochondrial biogenesis, THs act to uncouple the synthesis of ATP from heat production in the mitochondria.142 This uncoupling is mediated by their action on mitochondrial uncoupling proteins (UCP) that lead to non-shivering thermogenesis via conversion of chemical energy to heat without an increase in ATP production. The presence of this mechanism, in which promoting uncoupling phosphorylation in brown adipose tissue (BAT) is promoted, is one of the markers of evolutionary process of mammals; however, for many years it was thought that BAT was not present in adults. Nevertheless, in the past decade, the presence of active BAT in adult humans has been demonstrated and its amounts are inversely associated with body weight and serum glucose levels.146,147–152 The action of TH in this tissue gains attention as additional mechanisms enrolled in MetS.
In BAT, type 1 UCP (UCP1) is the hallmark of thermogenesis. This UCP expression is stimulated by T3, which is locally generated from T4 by intracellular D2. This D2 is positively regulated by beta-adrenergic activity.152 THs cause an upregulation of adrenergic receptor expression, leading to an amplified effect on UCP1 expression, which is also activated by the SNS.152 Studies have shown that D2 is very important to TH-induced adaptive type of thermogenesis in BAT.152 D2 also responds to other thermogenic inductors, as highlighted by a recent study showing that the adipokine, adipocyte fatty-acid-binding protein (A-FABP), requires BAT D2 activity to exert its thermogenic effects.153
Another postulated effect of THs in BAT is the stimulation of WAT ‘browning,’ which consists of the acquisition of brown-fat characteristics by a certain group of WAT cells, termed beige cells.154 Although it would be an attractive tool in obesity treatment, evidence in humans is still scarce,152 and a recent experimental study does not support that TH-induced browning is accompanied by an increase in thermogenesis.155 TH also stimulates the expression of other UCPs, such as UCP2 and 3, and the latter is very important to thermogenesis and fatty oxidation in muscle.156
In addition to acting on peripheral tissues, THs also have relevant modulatory actions in the central nervous system with respect to core body temperature, satiety control, and activity of the SNS.157 The action of T3 on the hypothalamus, more specifically on the ventromedial hypothalamus (VMH), stimulates the SNS that not only stimulates TH production but also acts in combination with THs in those same peripheral tissues that affect the MetS components.125–127
Central T3 administration results in increased body temperature, concomitant with reduction of levels of hypothalamic AMP-activated protein kinase (AMPK), increased tone in the sympathetic nerves innervating BAT.158,159 Hypothalamic AMPK and fatty-acid metabolism mediate thyroid regulation of energy balance.158–160 Those responses involve UCP1, since they were abrogated in UCP1 knockout mice.161
Hyperthyroid individuals frequently have hyperphagia even in the presence of weight lost,157 which is related in great part to the direct effect of THs on appetite stimulation. In the hypothalamic nucleus arcuate, T3, produced locally by D2, increases the expression of the orexigenic peptides neuropeptide Y (NPY) and agouti-related peptide (AgRP), and decreases the anorexigenic peptide, pro-opiomelanocortin (POMC),160 and the reverse events occur in hypothyroid rats.162 Acting at the VMH, T3, in low doses, was shown to induce an increase in food intake and potently stimulate the sympathetic activity and BAT thermogenesis.126,163,164 In contrast, Hameed and colleagues demonstrated that ablation of the β isoform of the TR only at the VMH of adult rats led to increase in AgRP/NPY and reduction in POMC pathways, with a concurrent augmentation in food intake and weight gain.165 This effect was not observed when both isoforms of TR had downregulated functions in the VMH.160 Therefore, not only the availability of T3, but also the specific TR isoform, determines the final effect of THs in control of hypothalamic circuits controlling energy homeostasis.
The action of TH in the regulation of EE may be indirect via controlling the action with or without expression of other circulating or local factors. Recently, it has been reported that irisin, a hormone produced in striate muscle after exercise,166 induces browning of WAT and shows a possible relation with thyroid function.167 However, human studies present conflicting results regarding the association between thyroid function and irisin levels, with some studies demonstrating higher levels in hyperthyroidism168,169 and low levels in hypothyroid patients.170–172 However, these results were not confirmed in all studies.173–175
Altered thyroid function can modify circulating levels of fibroblast growth factor 21 (FGF21), fetuin A, and neuregulin 4 (NgL-4), among others, which modulate EE.27,48 NgL-4 is an epidermal growth factor (EGF) family member that is secreted by BAT and promotes augmentation in EE, inhibition of hepatic lipogenesis, and reduction of fat-mass storage.176 A study with 129 hyperthyroid patients demonstrated that they had higher levels of NgL-4 than controls, which showed a reduction in these levels after restoring euthyroidism with treatment.177 Studies evaluating possible opposite effects, leading to reduction of NgL-4 in hypothyroidism, are still lacking.
In addition to TH, TSH has been shown to act directly in adipose tissue that expresses TSH receptors. In differentiated human adipocytes, TSH induces lipolysis and inhibits insulin signaling through protein kinase B (Akt) phosphorylation,178 which might contribute to IR. However, Ma and coworkers showed that TSH appears to stimulate adipocyte differentiation and lipogenesis in the pre-adipocyte cell lineage 3T3-L1 through a mechanism involving peroxisome-proliferated-activator–receptor (PPAR) gamma.179 In agreement with a role of TSH as an adipogenic factor, mice that did not express the TSH receptor and were under TH supplementation, exhibited resistance to high-fat-diet-induced obesity.179
Adiposity influencing thyroid function
Leptin is a hormone produced by adipose tissue in direct proportion to the quantity of adipose tissue mass. Leptin acts mainly at hypothalamic neurons to induce satiety and increase EE. Patients with genetic mutations in the leptin gene or leptin receptor are obese, and chronic reposition of leptin caused normalization of their body weight. However, most obese patients have hyperleptinemia but are resistant to the anorexigenic central action of leptin.180,181
In addition, leptin was shown to regulate the production of neurohormones in the medio-basal hypothalamus, among them, thyrotropin-releasing hormone (TRH) neurons of the periventricular nucleus.181,182 In another study, leptin activated TRH neurons both directly and indirectly, acting through the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway.182,183 The increase in TRH release was shown to lead to higher pituitary secretion of TSH,182–184 which in turn, stimulates thyroid function and proliferation.
Besides acting as a stimulatory agent for TRH secretion, the overall response of the thyroid axis to leptin is controversial among species and depends on nutritional status.185 Both rodents and humans subject to fasting show suppression of TH function, with concomitant decreases in serum levels of leptin, and replacement of leptin partially restored normal concentrations of thyroid hormones.186–189 Therefore, during caloric deprivation, the reduction in leptin seems to contribute to an integrated response to fasting, including thyroid-function suppression. However, in conditions with hyperleptinemia or at physiological levels, the role of leptin in thyroid function is less clear and may also reflect other leptin actions in the pituitary, thyroid, and peripheral tissues. Leptin receptors have been found in the anterior pituitary and thyroid gland, and direct inhibitory actions on TSH secretion and on the expressions of the Na+/I– symporter (NIS) and thyroglobulin messenger ribonucleic acid (mRNA) in thyroid cell lines have been reported.184,190 Additionally, there is experimental evidence from rodent studies that thyroid hormone metabolism may be modulated by leptin. Exogenous leptin administration caused an increase in D1 activity in the liver and pituitary, while causing a reduction in D2 activity at the hypothalamus and in BAT. Therefore, leptin may modulate thyroid hormone actions in target tissues, but collectively, these studies indicate that nutritional status and thyroid state clearly modify the responses to leptin.191–194
Another postulated mechanism of the way in which obesity is related to thyroid disfunction concerns chronic low-grade inflammation in adipose tissue that secretes cytokines and may affect thyroid function. It has been demonstrated that tumor necrosis factor alpha (TNF-α) and interleukins 1 and 6 (IL-1 and -6) inhibit the mRNA expression of the NIS.195 Additionally, pro-inflammatory cytokines have been associated with inhibition of D1 in HepG2 hepatocarcinoma cells196 and induction of D3,197 resulting in a decrease in serum T3, one feature of the low T3 syndrome associated with chronic diseases.198
Finally, IR, in conjunction with leptin levels, appears to be related to obesity and leads to augmentation of serum TSH levels.199,200 Recent studies give support to this hypothesis, showing that metformin, a drug used to improve insulin sensitivity, may cause a reduction in serum TSH levels.201,202 Different mechanisms have been proposed and the activation of the AMP-activated protein kinase (AMPK) pathway may be enrolled.158,159,203,204
Thyroid function acting on glucose metabolism
Hypothyroidism is associated with peripheral IR due to a reduction in glucose uptake, and on the other hand, hyperthyroidism increases glycemia due to an increase in liver production.205–207 T3 acts directly on the liver through TRβ, regulating genes involved in hepatic gluconeogenesis, glycogen metabolism, and insulin signaling.205,206 In addition, TH also acts centrally on the hypothalamus to increase sympathetic flow to the liver.126 As a consequence, in the liver, there is a decrease in glycogen synthesis and increase in gluconeogenesis and glucogenolysis,126,207 leading to an increase in glucose output.208 T3 increases the translocation of the glucose transport 4 (GLUT 4) to the plasma membrane in skeletal muscle and adipose tissue, which is associated with better glucose tolerance.208–215 T2 administration has also been associated with better glucose tolerance in animal models. It induces inhibition of hepatic gluconeogenesis gene expression216–218 by means of modulation of microRNA,217 and regulation of the activity of the protein kinase mammalian target of rapamycin complexes 1 (mTORC1) and 2 (mTORC2).218
Although THs play a role in islet trophic state maintenance,219 hyperthyroidism impairs glucose-stimulated insulin secretion and accelerates insulin degradation.220 In the insulin-producing cell line, INS-1 cells, at high concentrations, T3 induced B-cell apoptosis and death.221 Also, T2, at high concentrations, is able to decrease the glucose-induced insulin secretion, even though both T2 and T3 have a stimulatory effect at low concentrations.222 The importance of maintaining low levels of T3 in pancreatic β cells was shown in mice with specific β-cell pancreatic deletion of D3 that showed a decrease in pancreatic islet area, insulin-gene expression, and glucose-stimulated insulin secretion, even though the mice were euthyroid.223
Thyroid function acting on lipid metabolism related to metabolic syndrome
The lipid abnormalities related to MetS are hypertriglyceridemia and low serum HDL-c levels. These abnormalities will be the focus of the present revision despite a high number of studies evaluating several other alterations in lipid profile associated with thyroid function.224,225
THs have effects throughout the whole body, stimulating both lipid synthesis and degradation, but in the hyperthyroid condition, there is a predominant increase in lipolysis from fat stores.142 In the liver, THs stimulate the re-esterification of free fatty acids into triacylglycerol and also induce de novo lipogenesis from glucose metabolism.226 However, THs also concurrently stimulate fatty-acid oxidation, and, under physiological conditions, the result is a balance that does not increase hepatic triacylglycerol levels.226 The mechanisms of TH action involve direct regulation of the transcription rate of specific lipogenic/oxidative genes, in addition to alterations in the concentrations of metabolites, energy state of the cells, and post-translational modifications of proteins involved in the liver lipid metabolism.117,226
TH increases cholesterol clearance because even though they stimulate endogenous cholesterol synthesis, they potently increase hepatic cholesterol uptake and excretion as bile acids.227 Low-density lipoprotein (LDL)-c accumulates in the serum of hypothyroid patients since the LDL-receptor and the sterol regulatory element-binding protein 2 (SREBP2) are under-expressed in hypothyroidism. LDL-receptors mediate liver uptake of cholesterol that comes from peripheral tissues. SREBP2 is a key transcription factor that induces the expression of lipogenic-related genes, including Ldlr.227 Levels of very-low-density lipoprotein (VLDL) in the liver and in serum are influenced by lipoprotein lipases that are up-regulated by thyroid hormones, a mechanism that may contribute to the high serum triglycerides in hypothyroidism.228 In addition, ApoB100 levels are reduced by THs contributing to the increase in VLDL and LDL production observed in the liver during hypothyroidism.229
An increase in serum HDL-c has been reported in hypothyroid patients; this finding appears to be related to a decrease in activity of the cholesterol ester transfer protein (CEPT).228 CEPT, which is positively regulated by THs, mediates the exchange of cholesteryl-ester between HDL-c and VLDL and also has a pro-atherogenic role. Higher expression of CEPT would lead to higher cardiovascular risk, related to augmentation of serum levels of VLDL and reduction of HDL-c. However, as serum levels of HLD-c are also influenced by several other mechanisms, and are reduced in states of IR and obesity, there are disagreements with respect to the results of human studies regarding thyroid function and serum HDL-c, as shown in Table 2. HDL-c levels in hypothyroid patients might also be reduced when obesity diagnosis is present with marked reduction of insulin sensitivity or MetS.
Additionally, administration of T2 in rodents has hypolipemic action, affecting the hepatic lipid metabolism.129 It has been demonstrated that T2 is able to increase hepatic lipid oxidation and contrary to T3, does not stimulate the lipogenic pathway in animals fed a high-fat diet,230 which potentially contributes to the important effect reported in avoiding lipid accumulation in the liver of those animals. Despite the evidence in rodents, the physiological role of T2 in human metabolism, and potential therapeutic use, need further clarification.231,232 Serum levels of 3,5-T2 have been associated with several clinical conditions, like impaired renal function, sepsis, and oral LT4 (levothyroxine) supplementation;232 however, further studies are necessary to evaluate causative effects between the found associations. These studies may benefit from a recently developed method to measure 3,5-T2 in human serum by mass spectrometry, which, interestingly, showed correlation with T2 isomer 3,3'-T2, but not with serum T3 or T4.233 Likewise, other methods to measure 3,5-T2 by mass spectrometry have been tested.234–236
Thyroid hormone acting on blood pressure
THs act on the vasculature and in the heart by TR-mediated gene regulation in the nucleus and also via other non-classical pathways at the cytoplasmatic and cellular membrane levels.130,237
In myocytes, and also in vasculature, THs, especially T3 with greater affinity, bind to TH nuclear receptors in its two isoforms, TRα and TRβ. Thereafter, the complex formed by TH response elements at the promoter regions of specific responsive genes lead to positive or negative regulation of several genes enrolled in cardiac function and vascular resistance. The sarcoplasmic reticulum calcium ATPase (SERCA2), the myosine-have chains-α (αMHC), the Na+/K+ ATPase, the voltage-gated K+ channels, the adenine nucleotide translocase (ANT1) and the β-adrenergic receptor are positively regulated by THs. In opposite, the myosine-have chains-β (βMHC), the phospholamban, the Na+/Ca2+ exchanger (NCX1), the TRα1, adenylyl cyclase (types V, VI) and TH transporters 8 and 10 are negatively regulated by THs.130,237
Additionally to genomic effects of TH on cardiac myocytes, and also on vasculature, there are important and faster non-genomic actions, like those related to direct modulation of membrane ion channels.130
THs have important inotropic and chronotropic effects on the heart and concomitantly, they cause vasodilatation in the systemic circulation, leading to a decrease in systemic vascular resistance. Hyperthyroid patients exhibit tachycardia, increased heart contractility, and decreased cardiac after-load, resulting in increased cardiac output, which leads to systolic hypertension. Hypothyroid patients may exhibit diastolic hypertension, associated with impaired endothelial-dependent vasodilatation.238 Alterations in the microcirculation of hypothyroid patients have also been reported, such as a decrease in blood-flow velocity and impaired vasodilation after a short period of ischemia.239 The mechanism involves TH stimulation of nitric oxide production and regulation of other local regulatory factors, resulting in a decrease in vascular smooth muscular tone.239–242
In addition, TH actions in the central nervous system have an influence on autonomic regulation of BP. Recently, a group of parvalbuminergic neurons at the anterior hypothalamus, which act to decrease BP, was described, and their development appears to be dependent on TRα signaling.243 This finding may explain the hypotension present in patients with TRα mutations.244 Different from peripheral systemic vasculature, the pulmonary vasculature does not respond to the vasodilator effect of TH and may explain reversible pulmonary hypertension related to hyperthyroidism.245
Studies evaluating the association between metabolic syndrome, or its components, and thyroid function in humans
Table 2 summarizes the results of different cross-sectional studies of the association between MetS and thyroid function that have been published in the last decade through July 2019. We excluded studies focusing on pediatric patients, elderly patients, and patients with a secondary diagnosis, such as polycystic ovary syndrome. Different criteria for defining MetS were adopted for these studies. However, the NCEPT/ATPIII was the most commonly applied criteria for diagnosis.14,16,19,25,26,38,48,54,68,69,73,77,92,94,97,98,102 Other authors used the IDF criteria,42,47,55,56,75,81,93 the World Health Organization or American Heart Association criteria,45,62,79 or even local/regional or pre-established criteria defined by a joint interim statement.29,45,52,76,96 Finally, some studies defined MetS by the presence of IR according to an abnormal Homeostatic Model Assessment of Insulin Resistance index (HOMA-IR) or euglycemic clamp result.24,49,57,71,82,84,96,100 As previously reported, not all studies evaluated the MetS diagnosis. However, the number of MetS components, or the presence of one or more of its features, were considered in many of the studies.
Almost all studies evaluated thyroid function through the assessment of serum TSH. Some studies combined assessments of serum TSH levels with the measurement of FT4. Serum FT3 or total T3 were also evaluated in some studies.15,16,22,27,33,35–38,45–47,49,54,57,60,73,74,77–79,81,82,85,86,89,93,96,101,105–110
When there was an observed association between serum TSH and the diagnosis of MetS, this association was commonly related to higher TSH levels.19,25,29,30,42,55,67,71,76,79,91,92,98,100,102,105 In some instances, it was detected among euthyroid subjects even in the presence of normal TSH levels.19,29,30,42,55,71,79,102 The association between serum FT4 and MetS diagnosis was not always found. However, when this association occurred, it was reported as positive (with higher serum FT4 levels) in some studies,38,53,102 while negative in others.24,54,95 Higher levels of serum FT3 related to MetS were also detected in some studies.38,82,96,105
As previously reported, obesity is commonly associated with high serum TSH level and with increment of deiodinases’ activities, converting T4 to T3. Thus, this hormonal profile (high TSH and FT3 levels and low serum FT4, even in its respective reference ranges) might be associated with MetS via mechanisms previously described that mediate the interaction between thyroid function and clinical components of metabolic syndrome.
As demonstrated in Table 2, glycemia or glycosylated hemoglobin might be positively37,46,62,75,93,94,100,153 or negatively33,66,76 associated with serum TSH levels. A positive association between TSH levels (or reduced thyroid function) and abnormal glucose metabolism may be related to the importance of the action of TH in different pathways related to glucose transport, especially those related to the expression of GLUT 4, as previously described. This hypothesis is supported by longitudinal studies that found a higher risk for diabetes mellitus (DM) development in patients with low thyroid function or higher levels of serum TSH.34,115
In fact, a positive association between fasting plasmatic insulin or HOMA-IR index and TSH levels has been described in some cross-sectional studies,16,24,25,59,66,70,82,84,94,100 which was confirmed in a cohort analysis of 5998 subjects.34 However, the increase in serum TSH levels may be an effect of weight gain based on several previously described mechanisms. Consequently, it may be solely a biomarker for MetS and not necessarily a causative effect of the studied endpoints related to MetS. Since patients diagnosed with MetS concomitant with IR may demonstrate lower levels of serum FT4 due to conversion of FT4 to T3, the absence of a correlation between glycemia or HOMA-IR and FT4 has been observed in a large number of studies, especially those examining euthyroid subjects (Table 2).
The adverse effects of glucose metabolism are not only associated with the reduction of thyroid function or higher serum TSH levels in humans, but the adverse effects are also associated with higher serum TH levels. Longitudinal studies found a higher risk for DM development correlated with higher levels of serum FT4.82,110,114 In fact, overt and subclinical (SC) hyperthyroidism were associated with fasting glycemia or abnormal glucose metabolism in different studies.27,59,76,114 However, the association between serum FT4 levels in the upper reference range and serum glucose was not consistently observed in all human studies (Table 2). Finally, a cohort analysis involving 38,200 individuals revealed a higher risk for DM development in patients with either hypothyroidism or hyperthyroidism. It seems reasonable to attribute a U-shaped pattern of risk to THs and glucose metabolism abnormalities.
Despite the lack of a consistent association between THs and HDL-c levels, a reduction in thyroid function and consequently, elevation of serum TSH levels, were shown to be associated with higher levels of serum TG in almost all human studies (Table 2). It is important to remember that a possible elevation of serum TSH levels as a consequence of obesity may be caused by both hormonal and metabolic abnormalities related to weight gain. Attributing this increase in serum TSH levels merely to reduced primary thyroid function may underestimate the effects of weight gain on thyroid function and overestimate hypothyroidism diagnostics, leading to possible overtreatment of conditions that should be first addressed by dietary modifications.
Not all human studies have demonstrated a correlation between TH levels and BP. However, a positive association between FT4 levels (even those levels in the reference range) and BP has been reported.20,22,34,38,63,76,98 However, the opposite results have also been found.26 Furthermore, associations between SC hypothyroidism58,89,90,100 or SC hyperthyroidism97 and higher BP have also been reported in some studies (Table 2).
Some longitudinal studies (Table 3) have shown that weight reduction is associated with lowering levels of serum TSH and FT3.28 Similarly, MetS development34,95,113 and weight gain74,116 have been found to be positively associated with TSH-level changes. However, these results have not been validated in other studies.17,67,69,110 Some researches only found this positive association for MetS development and not for changes in body mass index.34,113
Final considerations
The interaction between thyroid hormone levels and all components of MetS is complex. The potential role of T2 and novel factors, like irisin, FGF21, fetuin A and NgL-4, have been identified in recent studies that contribute to this multifaceted interaction. Researchers of human studies evaluating this association need to consider all confounding variables. Of note, longitudinal studies controlling each of those potential variables are still needed in order to assess this intriguing association, with special attention to age-, sex- and tissue-specific effects of THs.
Footnotes
Author’s note: PFS Teixeira and CC Pazos-Moura contributed to the conception and the design of the work; drafting the work and revising the manuscript.
PB dos Santos made substantial contributions to the content and reviewed and edited the review before submission as contributed in preparing the tables.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: FAPERJ (Fundação de Amparo à Pesquisa do Rio de Janeiro) and CNPQ (Conselho Nacional de Desenvolvimento Científico e Tecnológico).
Conflict of interest: CC Pazos-Moura and PB dos Santos do not have any conflict of interest to declare.
Despite no conflict of interest related to this work, PFS Teixeira has received, in the past, honoraria for consultancies from Merck and Sanofi.
Ethical approval: Ethical approval was not required for this review.
ORCID iD: Patrícia de Fátima dos Santos Teixeira https://orcid.org/0000-0001-8859-6387
Contributor Information
Patrícia de Fátima dos Santos Teixeira, Endocrine Clinic, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Professor Rodolpho Rocco, 255 – Cidade Universitária, Rio de Janeiro, RJ 21941-617, Brazil.
Patrícia Borges dos Santos, Research Fellow, Medicine School, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Endocrinologist, Instituto Estadual de Endocrinologia Luiz Capriglione, Rio de Janeiro, Brazil.
Carmen Cabanelas Pazos-Moura, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro-Brazil.
References
- 1. Palmer MK, Toth PP. Trends in lipids, obesity, metabolic syndrome, and diabetes mellitus in the United States: an NHANES analysis (2003-2004 to 2013-2104). Obesity 2019; 27: 309–314. [DOI] [PubMed] [Google Scholar]
- 2. Alberti KG, Zimmet P, Shaw J, et al. The metabolic syndrome – a new worldwide definition. Lancet 2005; 366: 1059–1062. [DOI] [PubMed] [Google Scholar]
- 3. Hu G, Qiao Q, Tuomilehto J, et al. Prevalence of the metabolic syndrome and its relation to all-cause and cardiovascular mortality in nondiabetic European men and women. Arch Intern Med 2004; 164: 1066–1076. [DOI] [PubMed] [Google Scholar]
- 4. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. Executive summary of the third report of the national cholesterol education program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol in adults (Adult Treatment Panel III). JAMA 2001; 285: 2486–2497. [DOI] [PubMed] [Google Scholar]
- 5. Mendes D, Alves C, Silveiro N, et al. Prevalence of undiagnosed hypothyroidism in Europe: a systematic review and meta-analysis. Eur Thyroid J 2019; 8: 130–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Hollowell JG, Staehling NW, Flanders WD, et al. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab 2002; 87: 489–499. [DOI] [PubMed] [Google Scholar]
- 7. Duntas LH, Yen PM. Diagnosis and treatment of hypothyroidism in the elderly. Endocrine 2019; 66: 63–69. [DOI] [PubMed] [Google Scholar]
- 8. Brenta G, Caballero AS, Nunes MT. Case finding for hypothyroidism should include type 2 diabetes and metabolic syndrome patients: a Latin American Thyroid Society (LATS) position statement. Endocr Pract 2019; 25: 101–105. [DOI] [PubMed] [Google Scholar]
- 9. Fontenelle LC, Feitosa MM, Severo JS, et al. Thyroid function in human obesity: underlying mechanisms. Horm Metab Res 2016; 48: 787–794. [DOI] [PubMed] [Google Scholar]
- 10. Lauberg P, Knudsen N, Andersen S, et al. Thyroid function and obesity. Eur Thyroid J 2012; 1: 159–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Iwen KA, Schröder E, Brabant G. Thyroid hormones and the metabolic syndrome. Eur Thyroid J 2013; 2: 83–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Guan B, Chen YY, Yang W, et al. Effect of bariatric surgery on thyroid function in obese patients: a systematic review and meta-analysis. Obes Surg 2017; 27: 3292–3305. [DOI] [PubMed] [Google Scholar]
- 13. McAninch EA, Bianco AC. Thyroid hormone signaling in energy homeostasis and energy metabolism. Ann N Y Acad Sci 2014; 1311: 77–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Wolffenbuttel BHR, Wouters HJCM, Slagter SN, et al. Thyroid function and metabolic syndrome in the population-based LifeLines cohort study. BMC Endocrine Disorders 2017; 17: 65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Rotondi M, Leporati P, La Manna A, et al. Raised serum TSH levels in patients with morbid obesity: is it enough to diagnose subclinical hypothyroidism? Eur J Endocrinol 2009; 160: 403–408. [DOI] [PubMed] [Google Scholar]
- 16. Alevizaki M, Saltiki K, Voidonikola P, et al. Free thyroxine is an independent predictor of subcutaneous fat in euthyroid individuals. Eur J Endocrinol 2009; 161: 459–465. [DOI] [PubMed] [Google Scholar]
- 17. Teixeira PF, Cabral MD, Silva NA, et al. Effect of levothyroxine treatment and relationship to menopausal status and body composition. Thyroid 2009; 19: 443–450. [DOI] [PubMed] [Google Scholar]
- 18. Volzke H, Ittermann T, Schmidt CO, et al. Subclinical hyperthyroidism and blood pressure in a population-based prospective cohort study. Eur J Endocrinol 2009; 161: 615–621. [DOI] [PubMed] [Google Scholar]
- 19. Park HT, Cho GJ, Ahn KH, et al. Thyroid stimulating hormone is associated with metabolic syndrome in euthyroid postmenopausal women. Maturitas 2009; 62: 301–305. [DOI] [PubMed] [Google Scholar]
- 20. Kim BJ, Kim TY, Koh JM, et al. Relationship between serum free T4 (FT4) levels and metabolic syndrome (MS) and its components in healthy euthyroid subjects. Clin Endocrinol (Oxf) 2009; 70: 152–160. [DOI] [PubMed] [Google Scholar]
- 21. Asvold BO, Bjoro T, Vatten LJ. Association of serum TSH with high body mass differs between smokers and never-smokers. J Clin Endocrinol Metab 2009; 94: 5023–5027. [DOI] [PubMed] [Google Scholar]
- 22. Nam JS, Cho M, Park JS, et al. Triiodothyronine level predicts visceral obesity and atherosclerosis in euthyroid, overweight and obese subjects: T3 and visceral obesity. Obes Res Clin Pract 2010; 4: e315–e323. [DOI] [PubMed] [Google Scholar]
- 23. Friedrich N, Rosskopf D, Brabant G, et al. Associations of anthropometric parameters with serum TSH, prolactin, IGF-I, and testosterone levels: results of the study of health in Pomerania (SHIP). Exp Clin Endocrinol Diabetes 2010; 118: 266–273. [DOI] [PubMed] [Google Scholar]
- 24. Ambrosi B, Masserini B, Iorio L, et al. Relationship of thyroid function with body mass index and insulin-resistance in euthyroid obese subjects. J Endocrinol Invest 2010; 33: 640–643. [DOI] [PubMed] [Google Scholar]
- 25. Ruhla S, Weickert MO, Arafat AM, et al. A high normal TSH is associated with the metabolic syndrome. Clin Endocrinol (Oxf) 2010; 72: 696–701. [DOI] [PubMed] [Google Scholar]
- 26. Garduno-Garcia de J, Alvirde-Garcia U, Lopez-Carrasco G, et al. TSH and free thyroxine concentrations are associated with differing metabolic markers in euthyroid subjects. Eur J Endocrinol 2010; 163: 273–278. [DOI] [PubMed] [Google Scholar]
- 27. Maratou E, Hadjidakis DJ, Kollias A, et al. Studies of insulin resistance in patients with clinical and subclinical hypothyroidism. Eur J Endocrinol 2009; 160: 785–790. [DOI] [PubMed] [Google Scholar]
- 28. Marzullo P, Minocci A, Tagliaferri MA, et al. Investigations of thyroid hormones and antibodies in obesity: leptin levels are associated with thyroid autoimmunity independent of bioanthropometric, hormonal, and weight-related determinants. J Clin Endocrinol Metab 2010; 95: 3965–3972. [DOI] [PubMed] [Google Scholar]
- 29. Lai Y, Wang J, Jiang F, et al. The relationship between serum thyrotropin and components of metabolic syndrome. Endocr J 2011; 58: 23–30. [DOI] [PubMed] [Google Scholar]
- 30. Lee YK, Kim JE, Oh HJ, et al. Serum TSH level in healthy Koreans and the association of TSH with serum lipid concentration and metabolic syndrome. Korean J Intern Med 2011; 26: 432–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Liu C, Scherbaum WA, Schott M, et al. Subclinical hypothyroidism and the prevalence of the metabolic syndrome. Horm Metab Res 2011; 43: 417–421. [DOI] [PubMed] [Google Scholar]
- 32. Diez JJ, Iglesias P. Relationship between thyrotropin and body mass index in euthyroid subjects. Exp Clin Endocrinol Diabetes 2011; 119: 144–150. [DOI] [PubMed] [Google Scholar]
- 33. Taneichi H, Sasai T, Ohara M, et al. Higher serum free triiodothyronine levels within the normal range are associated with metabolic syndrome components in type 2 diabetic subjects with euthyroidism. Tohoku J Exp Med 2011; 224: 173–178. [DOI] [PubMed] [Google Scholar]
- 34. Park SB, Choi HC, Joo NS. The relation of thyroid function to components of the metabolic syndrome in Korean men and women. J Korean Med Sci 2011; 26: 540–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Kitahara CM, Platz EA, Ladenson PW, et al. Fatness and markers of thyroid function among U.S. men and women. PLoS One 2012; 7: e34979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Zhang J, Sun H, Chen L, et al. Relationship between serum TSH level with obesity and NAFLD in euthyroid subjects. J Huazhong Univ Sci Technol Sci 2012; 32: 47–52. [DOI] [PubMed] [Google Scholar]
- 37. Tamez-Pérez HE, Martínez E, Quintanilla-Flores DL, et al. The rate of primary hypothyroidism in diabetic patients is greater than in the non-diabetic population. An observational study. Med Clin (Barc) 2012; 138: 475–477. [DOI] [PubMed] [Google Scholar]
- 38. Tarcin O, Abanonu GB, Yazici D, et al. Association of metabolic syndrome parameters with TT3 and FT3/FT4 ratio in obese Turkish population. Metab Syndr Relat Disord 2012; 10: 137–142. [DOI] [PubMed] [Google Scholar]
- 39. Aljohani NJ, Al-Daghri NM, Al-Attas OS, et al. Differences and associations of metabolic and vitamin D status among patients with and without sub-clinical hypothyroid dysfunction. BMC Endocr Disord 2013; 13: 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Kwakernaak AJ, Lambert G, Muller Kobold AC, et al. Adiposity blunts the positive relationship of thyrotropin with proprotein convertase subtilisin-kexin type 9 levels in euthyroid subjects. Thyroid 2013; 23: 166–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Solanki A, Bansal S, Jindal S, et al. Relationship of serum thyroid stimulating hormone with body mass index in healthy adults. Indian J Endocrinol Metab 2013; 17(Suppl. 1): S167–S169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Oh JY, Sung YA, Lee HJ. Elevated thyroid stimulating hormone levels are associated with metabolic syndrome in euthyroid young women. Korean J Intern Med 2013; 28: 180–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Kouidhi S, Berhouma R, Ammar M, et al. Relationship of thyroid function with obesity and type 2 diabetes in euthyroid Tunisian subjects. Endocr Res 2013; 38: 15–23. [DOI] [PubMed] [Google Scholar]
- 44. Karthick N, Dillara K, Poornima KN, et al. Dyslipidaemic changes in women with subclinical hypothyroidism. J Clin Diagn Res 2013; 7: 2122–2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Muscogiuri G, Sorice GP, Mezza T, et al. High-normal TSH values in obesity: is it insulin resistance or adipose tissue’s guilt? Obesity 2013; 21: 101–106. [DOI] [PubMed] [Google Scholar]
- 46. Vyakaranam S, Vanaparthy S, Nori S, et al. Study of insulin resistance in subclinical hypothyroidism. Int J Health Sci Res 2014; 4: 147–153. [PMC free article] [PubMed] [Google Scholar]
- 47. Roef GL, Rietzschel ER, Van Daele CM, et al. Triiodothyronine and free thyroxine levels are differentially associated with metabolic profile and adiposity-related cardiovascular risk markers in euthyroid middle-aged subjects. Thyroid 2014; 24: 223–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Bakiner O, Bozkirli E, Ertugrul D, et al. Plasma fetuin-A levels are reduced in patients with hypothyroidism. Eur J Endocrinol 2014; 170: 411–418. Erratum in: Eur J Endocrinol. 2014; 171: X3. [DOI] [PubMed] [Google Scholar]
- 49. Mamtani M, Kulkarni H, Dyer TD, et al. Increased waist circumference is independently associated with hypothyroidism in Mexican Americans: replicative evidence from two large, population-based studies. BMC Endocr Disord 2014; 14: 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Ren R, Jiang X, Zhang X, et al. Association between thyroid hormones and body fat in euthyroid subjects. Clin Endocrinol 2014; 80: 585–590. [DOI] [PubMed] [Google Scholar]
- 51. Giandalia A, Russo GT, Romeo EL, et al. Influence of high-normal serum TSH levels on major cardiovascular risk factors and visceral adiposity index in euthyroid type 2 diabetic subjects. Endocrine 2014; 47: 152–160. [DOI] [PubMed] [Google Scholar]
- 52. Sakurai M, Nakamura K, Miura K, et al. Association between a serum thyroid-stimulating hormone concentration within the normal range and indices of obesity in Japanese men and women. Intern Med 2014; 53: 669–674. [DOI] [PubMed] [Google Scholar]
- 53. Shin DY, Kim KJ, Cho Y, et al. Body mass index is associated with hypercholesterolemia following thyroid hormone withdrawal in thyroidectomized patients. Int J Endocrinol 2014; 2014: 649016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Udenze I, Nnaji I, Oshodi T. Thyroid function in adult Nigerians with metabolic syndrome. Pan Afr Med J 2014; 18: 352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Shinkov A, Borissova AM, Kovatcheva R, et al. The prevalence of the metabolic syndrome increases through the quartiles of thyroid stimulating hormone in a population-based sample of euthyroid subjects. Arq Bras Endocrinol Metab 2014; 58: 926–932. [DOI] [PubMed] [Google Scholar]
- 56. Gierach M, Junik R. The effect of hypothyroidism occurring in patients with metabolic syndrome. Endokrynol Pol 2015; 66: 288–294. [DOI] [PubMed] [Google Scholar]
- 57. Aras S, Üstünsoy S, Armutçu F. Indices of central and peripheral obesity; anthropometric measurements and laboratory parameters of metabolic syndrome and thyroid function. Balkan Med J 2015; 32: 414–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Sieminska L, Wojciechowska C, Walczak K, et al. Associations between metabolic syndrome, serum thyrotropin, and thyroid antibodies status in postmenopausal women, and the role of interleukin-6. Endokrynologia Polska 2015; 6: 394–403. [DOI] [PubMed] [Google Scholar]
- 59. Ozdemir D, Dagdelen S, Usman A. Serum adiponectin levels and changes in glucose metabolism before and after treatment for thyroid dysfunction. Intern Med 2015; 54: 1849–1857. [DOI] [PubMed] [Google Scholar]
- 60. Lambrinoudaki I, Armeni E, Rizos D, et al. Indices of adiposity and thyroid hormones in euthyroid postmenopausal women. Eur J Endocrinol 2015; 173: 237–245. [DOI] [PubMed] [Google Scholar]
- 61. Bétry C, Challan-Belval MA, Bernard A, et al. Increased TSH in obesity: evidence for a BMI-independent association with leptin. Diabetes Metab 2015; 41: 248–251. [DOI] [PubMed] [Google Scholar]
- 62. Petrosyan L. Relationship between high normal TSH levels and metabolic syndrome components in type 2 diabetic subjects with euthyroidism. J Clin Transl Endocrinol 2015; 2: 110–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Meng Z, Liu M, Zhang Q, et al. Gender and age impacts on the association between thyroid function and metabolic syndrome in Chinese. Medicine 2015; 94: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Arsoy N, Yeler MT, Ayan NN, et al. Association between thyroid hormone levels and insulin resistance and body mass index. Park J Med Sci 2015; 31: 1417–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Maskey R, Shakya DR, Baranwal JK, et al. Hypothyroidism in diabetes mellitus patients in Eastern Nepal. Indian J Endocrinol Metab 2015; 19: 411–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Benseñor IM, Goulart AC, Molina Mdel C, et al. Thyrotropin levels, insulin resistance, and metabolic syndrome: a cross-sectional analysis in the Brazilian longitudinal study of adult healthy (ELSA-Brasil). Metab Syndr Relat Disord 2015; 13: 362–369. [DOI] [PubMed] [Google Scholar]
- 67. Nozarian Z, Abdollahi A, Mehrtash V, et al. Upper normal limit of thyroid-stimulating hormone and metabolic syndrome in Iranian patients with obesity. Iran J Pathol 2017; 12: 88–93. [PMC free article] [PubMed] [Google Scholar]
- 68. Lee JJ, Pedley A, Marqusee E, et al. Thyroid function and cardiovascular disease risk factors in euthyroid adults: a cross-sectional and longitudinal study. Clin Endocrinol 2016; 85: 932–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Peixoto de Miranda ÉJ, Bittencourt MS, Santos IS, et al. Thyroid function and high-sensitivity C-reactive protein in cross-sectional results from the Brazilian longitudinal study of adult health (ELSA-Brasil): effect of adiposity and insulin resistance. Eur Thyroid J 2016; 5: 240–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Kim HJ, Bae JC, Park HK, et al. Triiodothyronine levels are independently associated with metabolic syndrome in euthyroid middle-aged subjects. Endocrinol Metab 2016; 31: 311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Wang CC, Chang CJ, Hsu MI. The clinical and biochemical characteristics associated with insulin resistance in non-obese young women. Gynecol Endocrinol 2016; 32: 767–771. [DOI] [PubMed] [Google Scholar]
- 72. Temizkan S, Balaforlou B, Ozderya A, et al. Effects of thyrotrophin, thyroid hormones and thyroid antibodies on metabolic parameters in a euthyroid population with obesity. Clin Endocrinol 2016; 85: 616–623. [DOI] [PubMed] [Google Scholar]
- 73. Khatiwada S, Sah SK, Kc R, et al. Thyroid dysfunction in metabolic syndrome patients and its relationship with components of metabolic syndrome. Clin Diabetes Endocrinol 2016; 2: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Tiller D, Ittermann T, Greiser KH, et al. Association of serum TSH with anthropometric markers of obesity in the general population. Thyroid 2016; 26: 1205–1214. [DOI] [PubMed] [Google Scholar]
- 75. Xu B, Yang H, Wang Z, et al. Elevated thyroid stimulating hormone levels are associated with metabolic syndrome in a Chinese community-based population of euthyroid people aged 40 years and older. J Biomed Res 2016; 30: 476–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Mehran L, Amouzegar A, Rahimabad PK, et al. Thyroid function and metabolic syndrome: a population-based thyroid study. Horm Metab Res 2017; 49: 192–200. [DOI] [PubMed] [Google Scholar]
- 77. Jayanthi R, Srinivasan AR, Hanifah M, et al. Associations among insulin resistance, triacylglycerol/high density lipoprotein (TAG/HDL ratio) and thyroid hormone levels-a study on type 2 diabetes mellitus in obese and overweight subjects. Diabetes Metab Syndr 2017; 11: S121–S126. [DOI] [PubMed] [Google Scholar]
- 78. Al-Musa HM. Impact of obesity on serum levels of thyroid hormones among euthyroid Saudi adults. J Thyroid Res 2017; 2017: 5739806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Lozanov LB, Gorcheva D, Lozanov BS, et al. Role of thyroid deficiency on adiponectin, leptin and metabolic status in visceral obesity: a cross-sectional study. Horm Metab Res 2017; 49: 667–672. [DOI] [PubMed] [Google Scholar]
- 80. Kar K, Sinha S. Variations of adipokines and insulin resistance in primary hypothyroidism. J Clin Diagn Res 2017; 11: BC07–BC09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Gutch M, Rungta S, Kumar S, et al. Thyroid functions and serum lipid profile in metabolic syndrome. Biomed J 2017; 40: 147–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Ferrannini E, Iervasi G, Cobb J, et al. Insulin resistance and normal thyroid hormone levels: prospective study and metabolomic analysis. Am J Physiol Endocrinol Metab 2017; 312: E429–E436. [DOI] [PubMed] [Google Scholar]
- 83. Witte T, Völzke H, Lerch MM, et al. Association between serum thyroid-stimulating hormone levels and visceral adipose tissue: a population-based study in northeast Germany. Eur Thyroid J 2017; 6: 12–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Răcătăianu N, Leach N, Bondor CI, et al. Thyroid disorders in obese patients. Does insulin resistance make a difference? Arch Endocrinol Metab 2017; 61: 575–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Rahbar AR, Kalantarhormozi M, Izadi F, et al. Relationship between body mass index, waist-to-hip ratio, and serum lipid concentrations and thyroid-stimulating hormone in the euthyroid adult population. Iran J Med Sci 2017; 42: 301–305. [PMC free article] [PubMed] [Google Scholar]
- 86. Valdés S, Maldonado-Araque C, Lago-Sampedro A, et al. Reference values for TSH may be inadequate to define hypothyroidism in persons with morbid obesity: Di@bet.es study. Obesity 2017; 25: 788–793. [DOI] [PubMed] [Google Scholar]
- 87. Sami A, Iftekhar MF, Rauf MA, et al. Subclinical hypothyroidism among local adult obese population. Park J Med Sci 2018; 34: 980–983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Jang J, Kim Y, Shin J, et al. Association between thyroid hormones and the components of metabolic syndrome. BMC Endocr Disord 2018; 18: 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Liu J, Duan Y, Fu J, et al. Association between thyroid hormones, thyroid antibodies, and cardiometabolic factors in non-obese individuals with normal thyroid function. Front Endocrinol (Lausanne) 2018; 9: 130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Liu X, Zhang C, Meng Z, Li X, et al. Waist circumference and subclinical thyroid dysfunction in a large cohort of Chinese men and women. Endocr Pract 2018; 24: 733–739. [DOI] [PubMed] [Google Scholar]
- 91. Zhou YC, Fang WH, Kao TW, et al. Exploring the association between thyroid- stimulating hormone and metabolic syndrome: a large population-based study. PLoS One 2018; 13: e0199209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Liu FH, Hwang JS, Kuo CF, et al. Subclinical hypothyroidism and metabolic risk factors association: a health examination-based study in northern Taiwan. Biomed J 2018; 41: 52–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Bermúdez V, Salazar J, Añez R, et al. Metabolic syndrome and subclinical hypothyroidism: a type 2 diabetes-dependent association. J Thyroid Res 2018; 2018: 8251076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Mousa U, Bozkus Y, Demir CC, et al. Fat distribution and metabolic profile in subjects with Hashimoto’s. Acta Endocrinologica (Buc) 2018; XIV: 105–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Amouzegar A, Kazemian E, Abdi H, et al. Association between thyroid function and development of different obesity phenotypes in euthyroid adults: a nine-year follow-up. Thyroid 2018; 28: 458–464. [DOI] [PubMed] [Google Scholar]
- 96. Wang CY, Yu TY, Shih SR, et al. Low total and free triiodothyronine levels are associated with insulin resistance in non-diabetic individuals. Sci Rep 2018; 8: 10685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Hamlaoui ML, Ayachi A, Dekaken A, et al. Relationship of metabolic syndrome and its components with thyroid dysfunction in Algerian patients. Diabetes Metab Syndr 2018; 12: 1–4. [DOI] [PubMed] [Google Scholar]
- 98. Delitala AP, Scuteri A, Fiorillo E, et al. Role of adipokines in the association between thyroid hormone and components of the metabolic syndrome. J Clin Med 2019; 8: 764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. De Vries TI, Kappelle LJ, Van der, Graaf Y, et al. Thyroid-stimulating hormone levels in the normal range and incident type 2 diabetes mellitus. Acta Diabetol 2019; 56: 431–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Chang YC, Hua SC, Chang CH, et al. High TSH level within normal range is associated with obesity, dyslipidemia, hypertension, inflammation, hypercoagulability, and the metabolic syndrome: a novel cardiometabolic marker. J Clin Med 2019; 8: E817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Xu R, Huang F, Zhang S, et al. Thyroid function, body mass index, and metabolic risk markers in euthyroid adults: a cohort study. BMC Endocrine Disorders 2019; 19: 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Kim JM, Kim BH, Lee H, et al. The relationship between thyroid function and different obesity phenotypes in Korean euthyroid adults. Diabetes Metab J 2019; 43: e26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Zhang X, Li Y, Zhou X, et al. Association between serum thyrotropin within the euthyroid range and obesity. Endocr J 2019; 66: 451–457. [DOI] [PubMed] [Google Scholar]
- 104. Lertrit A, Chailurkit LO, Ongphiphadhanakul B, et al. Thyroid function is associated with body mass index and fasting plasma glucose in Thai euthyroid population. Diabetes Metab Syndr 2019; 13: 468–473. [DOI] [PubMed] [Google Scholar]
- 105. Raposo L, Martins S, Ferreira D, et al. Metabolic syndrome, thyroid function and autoimmunity - the PORMETS study. Endocr Metab Immune Disord Drug Targets 2019; 19: 75–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Nada AM. Effect of treatment of overt hypothyroidism on insulin resistance. World J Diabetes 2013; 15: 157–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Langén VL, Niiranen TJ, Puukka P, et al. Association between thyroid-stimulating hormone and blood pressure in adults: an 11-year longitudinal study. Clin Endocrinol (Oxf) 2016; 84: 741–747. [DOI] [PubMed] [Google Scholar]
- 108. Langén VL, Niiranen TJ, Puukka P, et al. Association of thyroid-stimulating hormone with lipid concentrations: an 11-year longitudinal study. Clin Endocrinol (Oxf) 2017; 86: 120–127. [DOI] [PubMed] [Google Scholar]
- 109. Itterman T, Tiller D, Meisinger C, et al. High serum thyrotropin levels are associated with current but not with incident hypertension. Thyroid 2013; 23: 955–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Liu G, Liang L, Bray GA, et al. Thyroid hormones and changes in body weight and metabolic parameters in response to weight loss diets: the POUNDS LOST trial. Int J Obes (Lond) 2017; 41: 878–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Eray E, Sari F, Ozdem S, et al. Relationship between thyroid volume and iodine, leptin, and adiponectin in obese women before and after weight loss. Med Princ Pract 2011; 20: 43–46. [DOI] [PubMed] [Google Scholar]
- 112. Chen RH, Chen HY, Man KM, et al. Thyroid diseases increased the risk of type 2 diabetes mellitus: a nation-wide cohort study. Medicine (Baltimore) 2019; 98: e15631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Chang CH, Yeh YC, Caffrey JL, et al. Metabolic syndrome is associated with an increased incidence of subclinical hypothyroidism - A Cohort Study. Sci Rep 2017; 7: 6754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Caixàs A, Tirado R, Vendrell J, et al. Plasma visfatin concentrations increase in both hyper and hypothyroid subjects after normalization of thyroid function and are not related to insulin resistance, anthropometric or inflammatory parameters. Clin Endocrinol (Oxf) 2009; 71: 733–738. [DOI] [PubMed] [Google Scholar]
- 115. Chaker L, Ligthart S, Korevaar TI, et al. Thyroid function and risk of type 2 diabetes: a population-based prospective cohort study. BMC Med 2016; 14: 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Bjergved L, Jørgensen T, Perrild H, et al. Thyroid function and body weight: a community-based longitudinal study. PLoS One 2014; 9: e93515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Soriguer F, Valdes S, Morcillo S, et al. Thyroid hormone levels predict the change in body weight: a prospective study. Eur J Clin Invest 2011; 41: 1202–1209. [DOI] [PubMed] [Google Scholar]
- 118. Ortiga-Carvalho TM, Sidhaye AR, Wondisford FE. Thyroid hormone receptors and resistance to thyroid hormone disorders. Nat Rev Endocrinol 2014; 10: 582–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Cordeiro A, Souza LL, Einicker-Lamas M, et al. Non-classic thyroid hormone signalling involved in hepatic lipid metabolism. J Endocrinol 2013; 216: R47–R57. [DOI] [PubMed] [Google Scholar]
- 120. Bianco AC, Dumitrescu A, Gereben B, et al. Paradigms of dynamic control of thyroid hormone signaling. Endocr Rev 2019; 40: 1000–1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Hönes GS, Rakov H, Logan J, et al. Noncanonical thyroid hormone signaling mediates cardiometabolic effects in vivo. Proc Natl Acad Sci 2017; 26; 114: E11323–E11332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Engels K, Rakov H, Hönes GS, et al. Aging alters phenotypic traits of thyroid dysfunction in male mice with divergent effects on complex systems but preserved thyroid hormone action in target organs. J Gerontol A Biol Sci Med Sci 2019; 74: 1162–1169. [DOI] [PubMed] [Google Scholar]
- 123. Rakov H, De Angelis, Renko K, et al. Aging is associated with low thyroid state and organ-specific sensitivity to thyroxine. Thyroid 2019; 29: 1723–1733. [DOI] [PubMed] [Google Scholar]
- 124. Rakov H, Engels K, Hönes GS, et al. Sex-specific phenotypes of hyperthyroidism and hypothyroidism in aged mice. Biol Sex Differ 2017; 8: 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Ribeiro MO, Bianco SD, Kaneshige M, et al. Expression of uncoupling protein 1 in mouse brown adipose tissue is thyroid hormone receptor-beta isoform specific and required for adaptive thermogenesis. Endocrinology 2010; 151: 432–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Klieverik LP, Janssen SF, van Riel A, et al. Thyroid hormone modulates glucose production via a sympathetic pathway from the hypothalamic paraventricular nucleus to the liver. Proc Natl Acad Sci U S A 2009; 106: 5966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Gaur A, Pal GK, Ananthanarayanan PH, et al. Role of Ventromedial hypothalamus in high fat diet induced obesity in male rats: association with lipid profile, thyroid profile and insulin resistance. Ann Neurosci 2014; 21: 104–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Brenta G. Why can insulin resistance be a natural consequence of thyroid dysfunction? J Thyroid Res 2011; 2011: 152850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Senese R, Lasala P, Leanza C, et al. New avenues for regulation of lipid metabolism by thyroid hormones and analogs. Front Physiol 2014; 5: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Razvi S, Jabbar A, Pingitore A, et al. Thyroid hormones and cardiovascular function and diseases. J Am Coll Cardiol 2018; 71: 1781–1796. [DOI] [PubMed] [Google Scholar]
- 131. Toni R, Malaguti A, Castorina S, et al. New paradigms in neuroendocrinology: relationships between obesity, systemic inflammation and the neuroendocrine system. J Endocrinol Invest 2004; 27: 182–186. [DOI] [PubMed] [Google Scholar]
- 132. Duntas LH, Biondi B. The interconnections between obesity, thyroid function, and autoimmunity: the multifold role of leptin. Thyroid 2013; 23: 646–653. [DOI] [PubMed] [Google Scholar]
- 133. Vaitikus JA, Farrar JS, Celi FS. Thyroid hormone mediated modulation of energy expenditure. Eur J Clin Nutr 2019; 73: 166–171.30254244 [Google Scholar]
- 134. Mountain GE, Allen EV, Haines SF. The basal metabolic rate in essential hypertension. Am Heart J 1943; 26: 528–535. [Google Scholar]
- 135. Johannsen DL, Galgani JE, Johannsen NM, et al. Effect of short-term thyroxine administration on energy metabolism and mitochondrial efficiency in humans. PLoS One 2012; 7: e40837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Rotondi M, Magri F, Chiovato L. Risk of coronary heart disease and mortality for adults with subclinical hypothyroidism. JAMA 2010; 304: 2481. [DOI] [PubMed] [Google Scholar]
- 137. Kyrou I, Adesanya O, Hedley N, et al. Improved thyroid hypoechogenicity following bariartric-induced weight loss in Euthyroid adults with severe obesity- a pilot study. Front Endocrinol 2018; 9: 488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Silvestri E, Senese R, Cioffi F, et al. 3,5-diiodo-L-thyronine exerts metabolically favorable effects on visceral adipose tissue of rats receiving a high-fat diet. Nutrients 2019; 11: E278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Köhrle J. The colorful diversity of thyroid hormone metabolites. Eur Thyroid J 2019; 8: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Köhrle J, Biebermann H. 3-iodothyronamine-a thyroid hormone metabolite with distinct target profiles and mode of action. Endocr Rev 2019; 40: 602–630. [DOI] [PubMed] [Google Scholar]
- 141. Zucchi R, Rutigliano G, Saponaro F. Novel thyroid hormones. Endocrine 2019; 66: 95–104. [DOI] [PubMed] [Google Scholar]
- 142. Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, et al. Hypothalamus-pituitary-thyroid axis. Compr Physiol 2016; 6: 1387–1428. [DOI] [PubMed] [Google Scholar]
- 143. Sayre NL, Lechleiter JD. Fatty acid metabolism and thyroid hormones. Curr Trends Endocrinol 2012; 6: 65–76. [PMC free article] [PubMed] [Google Scholar]
- 144. Trivieri MG, Oudit GY, Sah R, et al. Cardiac-specific elevations in thyroid hormone enhance contractility and prevent pressure overload-induced cardiac dysfunction. Proc Natl Acad Sci 2006; 103: 6043–6048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Macdonald IA. Advances in our understanding of the role of the sympathetic nervous system in obesity. Int J Obes Relat Metab Disord 1995; 19(Suppl. 7): S2–S7. [PubMed] [Google Scholar]
- 146. Cypess AM, Lehman S, Williams G, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med 2009; 360: 1509–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, et al. Cold-activated brown adipose tissue in healthy men. N Engl J Med 2009; 360: 1500–1508. [DOI] [PubMed] [Google Scholar]
- 148. Virtanen KA, Lidell ME, Orava J, et al. Functional brown adipose tissue in healthy adults. N Engl J Med 2009; 9; 360: 1518–1525. [DOI] [PubMed] [Google Scholar]
- 149. Pfannenberg C, Werner MK, Ripkens S, et al. Thyroid functions and serum lipid profile inmetabolic syndrome. Diabetes 2010; 59: 1789–1793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Saito M, Okamatsu-Ogura Y, Matsushita M, et al. High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity. Diabetes 2009; 58: 1526–1531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Zingaretti MC, Crosta F, Vitali A, et al. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. FASEB J 2009; 23: 3113–3120. [DOI] [PubMed] [Google Scholar]
- 152. Bianco AC, McAninch EA. The role of thyroid hormone and brown adipose tissue in energy homoeostasis. Lancet Diabetes Endocrinol 2013; 1: 250–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Shu L, Hoo RL, Wu X, et al. A-FABP mediates adaptive thermogenesis by promoting intracellular activation of thyroid hormones in brown adipocytes. Nat Commun 2017; 27; 8: 14147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Johann K, Cremer AL, Fisher AW, et al. Thyroid-hormone-induced browning of white adipose tissue does not contribute to thermogenesis and glucose consumption. Cell Reports 2019; 27: 3385–3400. [DOI] [PubMed] [Google Scholar]
- 155. Weiner J, Hankir M, Heiker JT, et al. Thyroid hormones and browning of adipose tissue. Mol Cell Endocrinol 2017; 458: 156–159. [DOI] [PubMed] [Google Scholar]
- 156. Fuller-Jackson JP, Henry BA. Adipose and skeletal muscle thermogenesis: studies from large animals. J Endocrinol 2018; 237: R99–R115. [DOI] [PubMed] [Google Scholar]
- 157. Mullur R, Liu YY, Brent GA. Thyroid hormone regulation of metabolism. Physiol Rev 2014; 94: 355–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. López M, Varela L, Vázquez MJ, et al. Hypothalamic AMPK and fatty acid metabolism mediate thyroid regulation of energy balance. Nat Med 2010; 16: 1001–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Cannon B, Nedergaard J. Thyroid hormones: igniting brown fat via the brain. Nat Med 2010; 16: 965–967. [DOI] [PubMed] [Google Scholar]
- 160. Varela L, Martinez-Sanchez N, Gallego R, et al. Hypothalamic mTOR pathway mediates thyroid hormone-induced hyperphagia in hyperthyroidism. J Pathol 2012; 227: 209–222. [DOI] [PubMed] [Google Scholar]
- 161. Alvarez-Crespo M, Csikasz RI, Martínez-Sánchez N, et al. Essential role of UCP1 modulating the central effects of thyroid hormones on energy balance. Mol Metab 2016; 5: 271–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Calvino C, Império GE, Wilieman M, et al. Hypothyroidism induces hypophagia associated with alterations in protein expression of neuropeptide Y and proopiomelanocortin in the arcuate nucleus, independently of hypothalamic nuclei-specific changes in leptin signaling. Thyroid 2016; 26: 134–143. [DOI] [PubMed] [Google Scholar]
- 163. Coppola M, Cioffi F, Moreno M, et al. 3,5-diiodo-L-thyronine: a possible pharmacological agent? Curr Drug Deliv 2016; 13: 330–338. [DOI] [PubMed] [Google Scholar]
- 164. Fliers E, Klieverik LP, Kalsbeek A. Novel neural pathways for metabolic effects of thyroid hormone. Trends Endocrinol Metab 2010; 21: 230–236. [DOI] [PubMed] [Google Scholar]
- 165. Hameed S, Patterson M, Dhillo WS, et al. Thyroid hormone receptor beta in the ventromedial hypothalamus is essential for the physiological regulation of food intake and body weight. Cell Rep 2017; 19: 2202–2209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Boström P, Wu J, Jedrychowski MP, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012; 481: 463–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Tekin S, Erden Y, Ozyalin F, et al. Central irisin administration suppresses thyroid hormone production but increases energy consumption in rats. Neurosci Lett 2018; 674: 136–141. [DOI] [PubMed] [Google Scholar]
- 168. Şahin M, Canpolat AG, Çorapçioğlu D, et al. Association between circulating irisin levels and epicardial fat in patients with treatment-naïve overt hyperthyroidism. Biomarkers 2018; 23: 742–747. [DOI] [PubMed] [Google Scholar]
- 169. Yalcin MM, Akturk M, Tohma Y, et al. Irisin and myostatin levels in patients with graves’ disease. Arch Med Res 2016; 47: 471–475. [DOI] [PubMed] [Google Scholar]
- 170. Yang N, Zhang H, Gao X, et al. Role of irisin in Chinese patients with hypothyroidism: an interventional study. J Int Med Res 2019; 47: 1592–1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Uc ZA, Gorar S, Mizrak S, et al. Irisin levels increase after treatment in patients with newly diagnosed Hashimoto thyroiditis. J Endocrinol Invest 2019; 42: 175–181. [DOI] [PubMed] [Google Scholar]
- 172. Zybek-Kocik A, Sawicka-Gutaj N, Szczepanek-Parulska, et al. The association between irisin and muscle metabolism in different thyroid disorders. Clin Endocrinol 2018; 88: 460–467. [DOI] [PubMed] [Google Scholar]
- 173. Panagiotou G, Pazaitou-Panayiotou K, Paschou SA, et al. Changes in thyroid hormone levels within the normal and/or subclinical hyper- or hypothyroid range do not affect circulating irisin levels in humans. Thyroid 2016; 26: 1039–1045. [DOI] [PubMed] [Google Scholar]
- 174. Yasar HY, Demirpence M, Colak A, et al. Serum irisin and apelin levels and markers of atherosclerosis in patients with subclinical hypothyroidism. Arch Endocrinol Metab 2019; 63: 16–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Stratigou T, Dalamaga M, Antonakos G, et al. Hyperirisinemia is independently associated with subclinical hypothyroidism: correlations with cardiometabolic biomarkers and risk factors. Endocrice 2018; 61: 83–93. [DOI] [PubMed] [Google Scholar]
- 176. Chen Z, Wang GX, Ma SL, et al. Nrg4 promotes fuel oxidation and a healthy adipokine profile to ameliorate diet-induced metabolic disorders. Mol Metab 2017; 6: 863–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Li M, Chen Y, Jiang J, et al. Elevated serum neuregulin 4 levels in patients with hyperthyroidism. Endocr Connect 2019; 8: 728–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Felske D, Gagnon A, Sorisky A. Interacting effects of TSH and insulin on human differentiated adipocytes. Horm Metab Res 2015; 47: 681–685. [DOI] [PubMed] [Google Scholar]
- 179. Ma S, Jing F, Xu C, et al. Thyrotropin and obesity: increased adipose triglyceride content through glycerol-3-phosphate acyltransferase 3. Sci Rep 2015; 5: 7633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Münzberg H, Flier JS, Bjørbaek C. Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology 2004; 145: 4880–4899. [DOI] [PubMed] [Google Scholar]
- 181. Park HK, Ahima RS. Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism 2015; 64: 24–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Hollenberg AN. The role of the thyrotropin-releasing hormone (TRH) neuron as a metabolic sensor. Thyroid 2008; 18: 131–139. [DOI] [PubMed] [Google Scholar]
- 183. Paz-Filho G, Mastronardi C, Franco CB, et al. Leptin: molecular mechanisms, systemic pro-inflammatory effects, and clinical implications. Arq Bras Endocrinol Metabol 2012; 56: 597–607. [DOI] [PubMed] [Google Scholar]
- 184. Ortiga-Carvalho TM, Oliveira KJ, Soares BA, et al. The role of leptin in the regulation of TSH secretion in the fed state: in vivo and in vitro studies. J Endocrinol 2002; 174: 121–125. [DOI] [PubMed] [Google Scholar]
- 185. Santini F, Marzullo P, Rotondi M, et al. Mechanisms in endocrinology: the crosstalk between thyroid gland and adipose tissue: signal integration in health and disease. Eur J Endocrinol 2014; 171: R137–R152. [DOI] [PubMed] [Google Scholar]
- 186. Ahima RS, Prabakaran D, Mantzoros C, et al. Role of leptin in the neuroendocrine response to fasting. Nature 1996; 382: 250–252. [DOI] [PubMed] [Google Scholar]
- 187. Mantzoros CS, Magkos F, Brinkoetter M, et al. Leptin in human physiology and pathophysiology. Am J Physiol Endocrinol Metab 2011; 301: E567–E584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Rosenbaum M, Murphy EM, Heymsfield SB, et al. Low dose leptin administration reverses effects of sustained weight-reduction on energy expenditure and circulating concentrations of thyroid hormones. J Clin Endocrinol Metab 2002; 87: 2391–2394. [DOI] [PubMed] [Google Scholar]
- 189. Boelen A, Kwakkel J, Vos XG, et al. Differential effects of leptin and refeeding on the fasting-induced decrease of pituitary type 2 deiodinase and thyroid hormone receptor beta2 mRNA expression in mice. J Endocrinol 2006; 190: 537–544. [DOI] [PubMed] [Google Scholar]
- 190. Isozaki O, Tsushima T, Nozoe Y, et al. Leptin regulation of the thyroids: negative regulation on thyroid hormone levels in euthyroid subjects and inhibitory effects on iodine uptake and Na+/I– symporter mRNA expression in rat FRTL-5 cells. Endocr J 2004; 51: 415–423. [DOI] [PubMed] [Google Scholar]
- 191. Cabanelas A, Ortiga-Carvalho TM, Pazos-Moura CC. Acute cold exposure, leptin, and somatostatin analog (octreotide) modulate thyroid 5-deiodinase activity. Am J Physiol Endocrinol Metab 2003; 284: E1172–E1176. [DOI] [PubMed] [Google Scholar]
- 192. Cabanelas A, Lisboa PC, Moura EG, et al. Leptin acute modulation of the 5'-deiodinase activities in hypothalamus, pituitary and brown adipose tissue of fed rats. Horm Metab Res 2006; 38: 481–485. [DOI] [PubMed] [Google Scholar]
- 193. Araujo RL, De Andrade BM, De Figueiredo AS, et al. Low replacement doses of thyroxine during food restriction restores type 1 deiodinase activity in rats and promotes body protein loss. J Endocrinol 2008; 198: 119–125. [DOI] [PubMed] [Google Scholar]
- 194. Araujo RL, Andrade BM, Da Silva ML, et al. Tissue-specific deiodinase regulation during food restriction and low replacement dose of leptin in rats. Am J Physiol Endocrinol Metab 2009; 296: E1157–E1163. [DOI] [PubMed] [Google Scholar]
- 195. Ajjan RA, Watson PF, Findlay C, et al. The sodium iodide symporter gene and its regulation by cytokines found in autoimmunity. J Endoncrinol 1998; 158: 351–308. [DOI] [PubMed] [Google Scholar]
- 196. Jakobs TC, Mentrup B, Schmutzler C, et al. Proinflammatory cytokines inhibit the expression and function of human type I 5'-deiodinase in HepG2 hepatocarcinoma cells. Eur J Endocrinol 2002; 146: 559–566. [DOI] [PubMed] [Google Scholar]
- 197. Boelen A, Kwakkel J, Alkemade A, et al. Induction of type 3 deiodinase activity in inflammatory cells of mice with chronic local inflammation. Endocrinology 2005; 146: 5128–5134. [DOI] [PubMed] [Google Scholar]
- 198. De Vries EM, Fliers E, Boelen A. The molecular basis of the non-thyroidal illness syndrome. J Endocrinol 2015; 225: R67–R81. [DOI] [PubMed] [Google Scholar]
- 199. Galofré JC, Pujante P, Abreu C, et al. Relationship between thyroid-stimulating hormone and insulin in euthyroid obese men. Ann Nutr Metab 2008; 53: 188–194. [DOI] [PubMed] [Google Scholar]
- 200. Michalaki MA, Vagenakis AG, Leonardou AS, et al. Thyroid function in humans with morbid obesity. Thyroid 2006; 16: 73–78. [DOI] [PubMed] [Google Scholar]
- 201. Lupoli R, Di Minno A, Tortora A, et al. Effects of treatment with metformin on TSH levels: a meta-analysis of literature studies. J Clin Endocrinol Metab 2014; 99: E143–E148. [DOI] [PubMed] [Google Scholar]
- 202. Dos Santos PB, Gertrudes LN, Conceição FL, et al. Effects of metformin on TSH levels and benign nodular goiter volume in patients without insulin resistance or iodine insufficiency. Front Endocrinol (Lausanne) 2019; 17; 10: 465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Wang J, Gao J, Fan Q, et al. The effect of metformin on thyroid-associated serum hormone level and physiological indexes: a meta-analysis. Curr Pharm Des 2019; 25: 3257–3265. [DOI] [PubMed] [Google Scholar]
- 204. Rena G, Hardie G, Pearson ER. The mechanisms of action of metformin. Diabetologia 2017; 60: 1577–1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Feng X, Jiang Y, Meltzer P, et al. Thyroid hormone regulation of hepatic genes in vivo detected by complementary DNA microarray. Mol Endocrinol 2000; 14: 947–955. [DOI] [PubMed] [Google Scholar]
- 206. Santiago LA, Santiago DA, Faustino LC, et al. The ∆337T mutation on the TRβ causes alterations in growth, adiposity and hepatic glucose homeostasis in mice. J Endocrinol 2011; 211: 39–46. [DOI] [PubMed] [Google Scholar]
- 207. Jornayvaz FR, Lee HY, Jurczak MJ, et al. Thyroid hormone receptor-α gene knockout mice are protected from diet-induced hepatic insulin resistance. Endocrinology 2012; 153: 583–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Weinstein SP, O’Boyle E, Fisher M, et al. Regulation of GLUT2 glucose transporter expression in liver by thyroid hormone: evidence for hormonal regulation of the hepatic glucose transport system. Endocrinology 1994; 135: 649–654. [DOI] [PubMed] [Google Scholar]
- 209. Brunetto EL, Teixeira Sda S, Giannocco G, et al. T3 rapidly increases SLC2A4 gene expression and GLUT4 trafficking to the plasma membrane in skeletal muscle of rat and improves glucose homeostasis. Thyroid 2012; 22: 70–79. [DOI] [PubMed] [Google Scholar]
- 210. Weinstein SP, O’Boyle E, Haber RS. Thyroid hormone increases basal and insulin-stimulated glucose transport in skeletal muscle. The role of GLUT4 glucose transporter expression. Diabetes 1994; 43: 1185–1189. [DOI] [PubMed] [Google Scholar]
- 211. Dimitriadis GD, Raptis SA. Thyroid hormone excess and glucose intolerance. Exp Clin Endocrinol Diabetes 2001; 109(Suppl. 2): S225–S239. [DOI] [PubMed] [Google Scholar]
- 212. Diamant S, Gorin E, Shafrir E. Enzyme activities related to fatty-acid synthesis in liver and adipose tissue of rats treated with triiodothyronine. Eur J Biochem 1972; 26: 553–559. [DOI] [PubMed] [Google Scholar]
- 213. Loose DS, Cameron DK, Short HP, et al. Thyroid hormone regulates transcription of the gene for cytosolic phosphoenolpyruvate carboxykinase (GTP) in rat liver. Biochemistry 1985; 24: 4509–4512. [DOI] [PubMed] [Google Scholar]
- 214. Song MK, Dozin B, Grieco D, et al. Transcriptional activation and stabilization of malic enzyme mRNA precursor by thyroid hormone. J Biol Chem 1988; 263:17970–17974. [PubMed] [Google Scholar]
- 215. Carvalho SD, Negrao N, Bianco AC. Hormonal regulation of malic enzyme and glucose-6-phosphate dehydrogenase in brown adipose tissue. Am J Physiol 1993; 264: E874–E881. [DOI] [PubMed] [Google Scholar]
- 216. Da Silva Teixeira S, Filgueira C, Sieglaff DH, et al. 3,5-diiodothyronine (3,5-T2) reduces blood glucose independently of insulin sensitization in obese mice. Acta Physiol (Oxf) 2017; 220: 238–250. [DOI] [PubMed] [Google Scholar]
- 217. Senese R, Cioffi F, De Lange P, et al. miR-22-3p is involved in gluconeogenic pathway modulated by 3,5-diiodo-L-thyronine (T2). Sci Rep 2019; 9: 16645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Gnocchi D, Ellis ECS, Johansson H, et al. Diiodothyronines regulate metabolic homeostasis in primary human hepatocytes by modulating mTORC1 and mTORC2 activity. Mol Cell Endocrinol 2020; 499: 110604. [DOI] [PubMed] [Google Scholar]
- 219. Verga Falzacappa C, Mangialardo C, Madaro L, et al. Thyroid hormone T3 counteracts STZ induced diabetes in mouse. PLoS One 2011; 6: e19839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Fukuchi M, Shimabukuro M, Shimajiri Y, et al. Evidence for a deficient pancreatic beta-cell response in a rat model of hyperthyroidism. Life Sci 2002; 71: 1059–1070. [DOI] [PubMed] [Google Scholar]
- 221. Ximenes HM, Lortz S, Jörns A, et al. Triiodothyronine (T3)-mediated toxicity and induction of apoptosis in insulin-producing INS-1 cells. Life Sci 2007; 80: 2045–2050. [DOI] [PubMed] [Google Scholar]
- 222. Fallahi P, Ferrari SM, Santini E, et al. Both 3,5-diiodo-L-thyronine (T2) and T3 modulate glucose-induced insulin secretion. J Biol Regul Homeost Agents 2017; 31: 503–508. [PubMed] [Google Scholar]
- 223. Medina MC, Fonesca TL, Molina J, et al. Maternal inheritance of an inactive type III deiodinase gene allele affects mouse pancreatic β-cells and disrupts glucose homeostasis. Endocrinology 2014; 155: 3160–3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Duntas LH, Brenta G. A renewed focus on the association between thyroid hormones and lipid metabolism. Front Endocrinol (Lausanne) 2018; 9: 511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Liu XL, He S, Zhang SF, et al. Alteration of lipid profile in subclinical hypothyroidism: a meta-analysis. Med Sci Monit 2014; 20: 1432–1441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Sinha RA, Singh BK, Yen PM. Direct effects of thyroid hormones on hepatic lipid metabolism. Nat Rev Endocrinol 2018; 14: 259–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Shin DJ, Osborne TF. Thyroid hormone regulation and cholesterol metabolism are connected through sterol regulatory element-binding protein-2 (SREBP-2). J Biol Chem 2003; 278: 34114–34118. [DOI] [PubMed] [Google Scholar]
- 228. Duntas LH. Thyroid disease and lipids. Thyroid 2002; 12: 287–293. [DOI] [PubMed] [Google Scholar]
- 229. Davidson NO, Powell LM, Wallis SC, et al. Thyroid hormone modulates the introduction of a stop codon in rat liver apolipoprotein B messenger RNA. J Biol Chem 1988, 263: 13482–13485. [PubMed] [Google Scholar]
- 230. Senese R, Cioffi F, de Lange P, et al. Both 3,5-diiodo-L-thyronine and 3,5,3'-triiodo-L-thyronine prevent short-term hepatic lipid accumulation via distinct mechanisms in rats being fed a high-fat diet. Front Physiol 2017; 8: 706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Van der Valk F, Hassing C, Visser M, et al. The effect of a diiodothyronine mimetic on insulin sensitivity in male cardiometabolic patients: a double-blind randomized controlled trial. PLoS One 2014; 9: e86890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Köhrle J, Lehmphul I, Pietzner M, et al. 3,5-T2—a Janus-faced thyroid hormone metabolite exerts both canonical T3-mimetic endocrine and intracrine hepatic action. Front Endocrinol 2020; 10: 787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Lorenzini L, Nguyen NM, Sacripanti G, et al. Assay of endogenous 3,5-diiodo-L-thyronine (3,5-T2) and 3,3′-diiodo-L-thyronine (3,3′-T2) in human serum: a feasibility study. Front Endocrinol 2019; 10: 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Richards KH, Monk R, Renko K, et al. A combined LC-MS/MS and LC-MS3 multi-method for the quantification of iodothyronines in human blood serum. Anal Bioanal Chem 2019; 411: 5605–5616. [DOI] [PubMed] [Google Scholar]
- 235. Gu J, Soldin OP, Soldin SJ. Development and validation of an isotope dilution tandem mass spectrometry method for the simultaneous quantification of 3-iodothyronamine, thyroxine, triiodothyronine, and 3,3'-diiodo-L-thyronine in humans. Clin Chem 2010; 56: A60. [Google Scholar]
- 236. Jonklaas J, Sathasivam A, Wang H, et al. 3,3'-diiodothyronine concentrations in hospitalized or thyroidectomized patients: results from a pilot study. Endocr Pract 2014; 20: 797–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Berta E, Lengyel I, Halmi S, et al. Hypertension in thyroid diseases. Front Endocrinol 2019; 10: 482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Lekakis J, Papamichael C, Alevizaki M, et al. Flow-mediated, endothelium-dependent vasodilation is impaired in subjects with hypothyroidism, borderline hypothyroidism, and high-normal serum thyrotropin (TSH) values. Thyroid 1997; 7: 411–414. [DOI] [PubMed] [Google Scholar]
- 239. Pazos-Moura CC, Moura EG, Breitenbach MM, et al. Nailfold capillaroscopy in hypothyroidism and hyperthyroidism: blood flow velocity during rest and postocclusive reactive hyperemia. Angiology 1998; 49: 471–476. [DOI] [PubMed] [Google Scholar]
- 240. Cai Y, Manio MM, Leung GP, et al. Thyroid hormone affects both endothelial and vascular smooth muscle cells in rat arteries. Eur J Pharmacol 2015; 747: 18–28. [DOI] [PubMed] [Google Scholar]
- 241. Tamajusuku AS, Carrillo-Sepúlveda MA, Braganhol E, et al. Activity and expression of ecto-5-nucleotidase/CD73 are increased by thyroid hormones in vascular smooth muscle cells. Mol Cell Biochem 2006; 289: 65–72. [DOI] [PubMed] [Google Scholar]
- 242. Samuel S, Zhang K, Tang YD, et al. Triiodothyronine potentiates vasorelaxation via PKG/VASP signaling in vascular smooth muscle cells. Cell Physiol Biochem 2017; 41: 1894–1904. [DOI] [PubMed] [Google Scholar]
- 243. Mittag J, Lyons DJ, Sällström J, et al. Thyroid hormone is required for hypothalamic neurons regulating cardiovascular functions. J Clin Invest 2013; 123: 509–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Bochukova E, Schoenmakers N, Agostini M, et al. A mutation in the thyroid hormone receptor alpha gene. N Engl J Med 2012; 366: 243–249. Erratum in: N Engl J Med 2012; 367: 1474. [DOI] [PubMed] [Google Scholar]
- 245. Marvisi M, Zambrelli P, Brianti M, et al. Pulmonary hypertension is frequent in hyperthyroidism and normalizes after therapy. Eur J Intern Med 2006; 17: 267–271. [DOI] [PubMed] [Google Scholar]