Online first
Review paper
Published online: 2024-07-24

open access

Page views 18
Article views/downloads 17
Get Citation

Connect on Social Media

Connect on Social Media

Causes of difficulties with adequate levothyroxine substitution — an immunoendocrine perspective

Magdalena Łukawska-Tatarczuk1, Edward Franek12

Abstract

Hypothyroidism is one of the most common endocrinopathies worldwide, the treatment of which is based on replacement therapy with levothyroxine. However, this seemingly simple treatment method is fraught with many difficulties and frequent dissatisfaction among patients.

In fact, differences in response to levothyroxine probably depend on a complex interaction between individual, environmental, genetic, and epigenetic factors that are still not sufficiently understood. Immunological disturbances, underlying Hashimoto’s disease, the most common cause of hypothyroidism, probably play a significant role in these relationships. Indeed, a growing number of studies
indicate that autoimmunity through activation of low-grade inflammation can lead to impaired absorption, transport, metabolism, and action of thyroid hormones. This review provides an up-to-date overview of the causes responsible for both the difficulty in achieving target thyrotropin levels and persistence of nonspecific symptoms despite adequate hormone replacement from an immunoendocrine perspective. Understanding these mechanisms points to a new direction in the approach to hypothyroidism, indicating the need for new
personalized treatment strategies.

 

Article available in PDF format

View PDF Download PDF file

References

  1. Garmendia Madariaga A, Santos Palacios S, Guillén-Grima F, et al. The incidence and prevalence of thyroid dysfunction in Europe: a meta-analysis. J Clin Endocrinol Metab. 2014; 99(3): 923–931.
  2. Li J, Li Y, Shi X, et al. Thyroid Disorders, Iodine Status and Diabetes Epidemiological Survey Group. Prevalence and risk factors of hypothyroidism after universal salt iodisation: a large cross-sectional study from 31 provinces of China. BMJ Open. 2023; 13(2): e064613.
  3. Hughes K, Eastman C. Thyroid disease: Long-term management of hyperthyroidism and hypothyroidism. Aust J Gen Pract. 2021; 50(1-2): 36–42.
  4. Jonklaas J. Optimal Thyroid Hormone Replacement. Endocr Rev. 2022; 43(2): 366–404.
  5. Hennessey JV. The emergence of levothyroxine as a treatment for hypothyroidism. Endocrine. 2017; 55(1): 6–18.
  6. Bednarczuk T, Attanasio R, Hegedüs L, et al. Use of thyroid hormones in hypothyroid and euthyroid patients: a THESIS* questionnaire survey of Polish physicians. *THESIS: Treatment of hypothyroidism in Europe by specialists: an international survey. Endokrynol Pol. 2021; 72(4): 357–365.
  7. Burlacu MC, Attanasio R, Hegedüs L, et al. Use of thyroid hormones in hypothyroid and euthyroid patients: a THESIS* survey of Belgian specialists *THESIS: treatment of hypothyroidism in Europe by specialists: an international survey. Thyroid Res. 2022; 15(1): 3.
  8. Riis KR, Frølich JS, Hegedüs L, et al. Use of thyroid hormones in hypothyroid and euthyroid patients: A 2020 THESIS questionnaire survey of members of the Danish Endocrine Society. J Endocrinol Invest. 2021; 44(11): 2435–2444.
  9. Planck T, Lantz M, Perros P, et al. Use of Thyroid Hormones in Hypothyroid and Euthyroid Patients: A 2020 THESIS Questionnaire Survey of Members of the Swedish Endocrine Society. Front Endocrinol (Lausanne). 2021; 12: 795111.
  10. Jiskra J, Paleček J, Attanasio R, et al. Use of thyroid hormones in hypothyroid and euthyroid patients: a 2020 THESIS questionnaire survey of members of the Czech Society of Endocrinology. BMC Endocr Disord. 2022; 22(1): 117.
  11. Buffet C, Belin L, Attanasio R, et al. Real-life practice of thyroid hormone use in hypothyroid and euthyroid patients: A detailed view from the THESIS questionnaire survey in France. Ann Endocrinol (Paris). 2022; 83(1): 27–34.
  12. Wiersinga WM. Paradigm shifts in thyroid hormone replacement therapies for hypothyroidism. Nat Rev Endocrinol. 2014; 10(3): 164–174.
  13. Brokhin M, Danzi S, Klein I. Assessment of the Adequacy of Thyroid Hormone Replacement Therapy in Hypothyroidism. Front Endocrinol (Lausanne). 2019; 10: 631.
  14. Mitchell AL, Hegedüs L, Žarković M, et al. Patient satisfaction and quality of life in hypothyroidism: An online survey by the british thyroid foundation. Clin Endocrinol (Oxf). 2021; 94(3): 513–520.
  15. Feller M, Snel M, Moutzouri E, et al. Association of Thyroid Hormone Therapy With Quality of Life and Thyroid-Related Symptoms in Patients With Subclinical Hypothyroidism: A Systematic Review and Meta-analysis. JAMA. 2018; 320(13): 1349–1359.
  16. Chaker L, Bianco A, Jonklaas J, et al. Hypothyroidism. Lancet. 2017; 390(10101): 1550–1562.
  17. Fitzgerald SP, Bean NG, Falhammar H, et al. Clinical Parameters Are More Likely to Be Associated with Thyroid Hormone Levels than with Thyrotropin Levels: A Systematic Review and Meta-Analysis. Thyroid. 2020; 30(12): 1695–1709.
  18. Cui Z, Ding X, Bian N, et al. Relatively Lower FT3 Levels Are Associated with Impaired Quality of Life in Levothyroxine-Treated Patients with Hashimoto Thyroiditis. Int J Endocrinol. 2022; 2022: 1918674.
  19. Zhang H, Che W, Shi K, et al. FT4/FT3 ratio: A novel biomarker predicts coronary microvascular dysfunction (CMD) in euthyroid INOCA patients. Front Endocrinol (Lausanne). 2022; 13: 1021326.
  20. Boesen VB, Feldt-Rasmussen U, Bjorner JB, et al. How Should Thyroid-Related Quality of Life Be Assessed? Recalled Patient-Reported Outcomes Compared to Here-and-Now Measures. Thyroid. 2018; 28(12): 1561–1570.
  21. Wenzek C, Boelen A, Westendorf AM, et al. The interplay of thyroid hormones and the immune system - where we stand and why we need to know about it. Eur J Endocrinol. 2022; 186(5): R65–R77.
  22. Li Q, Wang B, Mu K, et al. The pathogenesis of thyroid autoimmune diseases: New T lymphocytes - Cytokines circuits beyond the Th1-Th2 paradigm. J Cell Physiol. 2019; 234(3): 2204–2216.
  23. Weetman AP. An update on the pathogenesis of Hashimoto's thyroiditis. J Endocrinol Invest. 2021; 44(5): 883–890.
  24. Zheng T, Xu C, Mao C, et al. Increased Interleukin-23 in Hashimoto's Thyroiditis Disease Induces Autophagy Suppression and Reactive Oxygen Species Accumulation. Front Immunol. 2018; 9: 96.
  25. Siddiq A, Naveed AK, Ghaffar N, et al. Association of Pro-Inflammatory Cytokines with Vitamin D in Hashimoto's Thyroid Autoimmune Disease. Medicina (Kaunas). 2023; 59(5).
  26. Zhang QY, Ye XP, Zhou Z, et al. Lymphocyte infiltration and thyrocyte destruction are driven by stromal and immune cell components in Hashimoto's thyroiditis. Nat Commun. 2022; 13(1): 775.
  27. Gigena N, Alamino VA, Montesinos MD, et al. Dissecting thyroid hormone transport and metabolism in dendritic cells. J Endocrinol. 2017; 232(2): 337–350.
  28. Klein JR. Physiological relevance of thyroid stimulating hormone and thyroid stimulating hormone receptor in tissues other than the thyroid. Autoimmunity. 2003; 36(6-7): 417–421.
  29. Guan H, de Morais NS, Stuart J, et al. Discordance of serological and sonographic markers for Hashimoto's thyroiditis with gold standard histopathology. Eur J Endocrinol. 2019; 181(5): 539–544.
  30. Siegmann EM, Müller HHO, Luecke C, et al. Association of Depression and Anxiety Disorders With Autoimmune Thyroiditis: A Systematic Review and Meta-analysis. JAMA Psychiatry. 2018; 75(6): 577–584.
  31. Groenewegen KL, Mooij CF, van Trotsenburg AS. Persisting symptoms in patients with Hashimoto's disease despite normal thyroid hormone levels: Does thyroid autoimmunity play a role? A systematic review. J Transl Autoimmun. 2021; 4: 100101.
  32. Tohidi M, Baghbani-Oskouei A, Amouzegar A, et al. Serum Thyroid Peroxidase Antibody Level and Incident Hypertension in Iranian Men: A Suggestion for the Role of Thyroid Autoimmunity. Endocr Metab Immune Disord Drug Targets. 2020; 20(10): 1711–1718.
  33. Khan SR, Peeters RP, van Hagen PM, et al. Determinants and Clinical Implications of Thyroid Peroxidase Antibodies in Middle-Aged and Elderly Individuals: The Rotterdam Study. Thyroid. 2022; 32(1): 78–89.
  34. Łukawska-Tatarczuk MM, Zieliński J, Franek E, et al. Is thyroid autoimmunity associated with subclinical atherosclerosis in young women with type 1 diabetes mellitus? Endokrynol Pol. 2022; 73(2): 301–308.
  35. Łukawska-Tatarczuk MM, Pawlak A, Zieliński J, et al. Association of antithyroid peroxidase antibodies with cardiac function in euthyroid women with type 1 diabetes mellitus - assessment with two-dimensional speckle‑tracking echocardiography. Endokrynol Pol. 2022; 73(5): 812–822.
  36. Mavai M, Bhandari B, Singhal A, et al. Cardiac Autonomic Modulation and Anti-Thyroid Peroxidase (TPO) Antibodies in Subclinical Hypothyroidism: Does a Correlation Exist? Cureus. 2021; 13(10): e18844.
  37. Wang GC, Talor MV, Rose NR, et al. Thyroid autoantibodies are associated with a reduced prevalence of frailty in community-dwelling older women. J Clin Endocrinol Metab. 2010; 95(3): 1161–1168.
  38. Leyhe T, Ethofer T, Bretscher J, et al. Low performance in attention testing is associated with reduced grey matter density of the left inferior frontal gyrus in euthyroid patients with Hashimoto's thyroiditis. Brain Behav Immun. 2013; 27(1): 33–37.
  39. DeBiase JM, Avasthi D. Hashimoto's Encephalopathy: A Case Report and Literature Review of an Encephalopathy With Many Names. Cureus. 2020; 12(8): e9601.
  40. Wei C, Shen Y, Zhai W, et al. Hashimoto's encephalopathy with cerebellar ataxia as the main symptom: A case report and literature review. Front Neurol. 2022; 13: 970141.
  41. Ruggeri RM, Vicchio TM, Cristani M, et al. Oxidative Stress and Advanced Glycation End Products in Hashimoto's Thyroiditis. Thyroid. 2016; 26(4): 504–511.
  42. Kardalas E, Sakkas E, Ruchala M, et al. The role of transforming growth factor beta in thyroid autoimmunity: current knowledge and future perspectives. Rev Endocr Metab Disord. 2022; 23(3): 431–447.
  43. Chang Q, Yin D, Li H, et al. HDAC6-specific inhibitor alleviates hashimoto's thyroiditis through inhibition of Th17 cell differentiation. Mol Immunol. 2022; 149: 39–47.
  44. Li H, Min J, Mao X, et al. Edaravone ameliorates experimental autoimmune thyroiditis in rats through HO-1-dependent STAT3/PI3K/Akt pathway. Am J Transl Res. 2018; 10(7): 2037–2046.
  45. McMillan M, Rotenberg KS, Vora K, et al. Comorbidities, Concomitant Medications, and Diet as Factors Affecting Levothyroxine Therapy: Results of the CONTROL Surveillance Project. Drugs R D. 2016; 16(1): 53–68.
  46. Bolk N, Visser TJ, Nijman J, et al. Effects of evening vs morning levothyroxine intake: a randomized double-blind crossover trial. Arch Intern Med. 2010; 170(22): 1996–2003.
  47. Ruchała M, Bossowski A, Brzozka MM, et al. Liquid levothyroxine improves thyroid control in patients with different hypothyroidism aetiology and variable adherence - case series and review. Endokrynol Pol. 2022; 73(5): 893–902.
  48. Bornikowska K, Gietka-Czernel M, Raczkiewicz D, et al. Improvements in Quality of Life and Thyroid Parameters in Hypothyroid Patients on Ethanol-Free Formula of Liquid Levothyroxine Therapy in Comparison to Tablet LT4 Form: An Observational Study. J Clin Med. 2021; 10(22).
  49. Virili C, Bruno G, Santaguida MG, et al. Levothyroxine treatment and gastric juice pH in humans: the proof of concept. Endocrine. 2022; 77(1): 102–111.
  50. Gietka-Czernel M, Hubalewska-Dydejczyk A, Kos-Kudła B, et al. Expert opinion on liquid L-thyroxine usage in hypothyroid patients and new liquid thyroxine formulation - Tirosint SOL [Opinia ekspertów dotycząca stosowania płynnej postaci lewotyroksyny oraz nowego preparatu Tirosint SOL u chorych na niedoczynność tarczycy]. Endokrynol Pol. 2020; 71(5): 441–465.
  51. Skelin M, Lucijanić T, Amidžić Klarić D, et al. Factors Affecting Gastrointestinal Absorption of Levothyroxine: A Review. Clin Ther. 2017; 39(2): 378–403.
  52. Colucci P, Yue CS, Ducharme M, et al. A Review of the Pharmacokinetics of Levothyroxine for the Treatment of Hypothyroidism. Eur Endocrinol. 2013; 9(1): 40–47.
  53. Rizzo LFL, Mana DL, Serra HA. Drug-induced hypothyroidism. Medicina (B Aires). 2017; 77(5): 394–404.
  54. Benvenga S. L-T4 Therapy in the Presence of Pharmacological Interferents. Front Endocrinol (Lausanne). 2020; 11: 607446.
  55. Iwama S, Kobayashi T, Yasuda Y, et al. Immune checkpoint inhibitor-related thyroid dysfunction. Best Pract Res Clin Endocrinol Metab. 2022; 36(3): 101660.
  56. Naguib R. Potential relationships between COVID-19 and the thyroid gland: an update. J Int Med Res. 2022; 50(2): 3000605221082898.
  57. Brancatella A, Viola N, Santini F, et al. COVID-induced thyroid autoimmunity. Best Pract Res Clin Endocrinol Metab. 2023; 37(2): 101742.
  58. Duntas LH, Jonklaas J. COVID-19 and Thyroid Diseases: A Bidirectional Impact. J Endocr Soc. 2021; 5(8): bvab076.
  59. Taşkaldıran I, Altay FP, Bozkuş Y, et al. A Case Report of Conversion from Hashimoto's Thyroiditis to Graves' Disease in Type 1 Diabetic Patient Following the COVID-19 Vaccination. Endocr Metab Immune Disord Drug Targets. 2023; 23(3): 405–409.
  60. Pappa T, Refetoff S. Resistance to Thyroid Hormone Beta: A Focused Review. Front Endocrinol (Lausanne). 2021; 12: 656551.
  61. Ortiga-Carvalho TM, Sidhaye AR, Wondisford FE. Thyroid hormone receptors and resistance to thyroid hormone disorders. Nat Rev Endocrinol. 2014; 10(10): 582–591.
  62. Paisdzior S, Knierim E, Kleinau G, et al. A New Mechanism in THRA Resistance: The First Disease-Associated Variant Leading to an Increased Inhibitory Function of THRA2. Int J Mol Sci. 2021; 22(10).
  63. Tagami T. An overview of thyroid function tests in subjects with resistance to thyroid hormone and related disorders. Endocr J. 2021; 68(5): 509–517.
  64. Hattori N, Ishihara T, Shimatsu A. Variability in the detection of macro TSH in different immunoassay systems. Eur J Endocrinol. 2016; 174(1): 9–15.
  65. Hattori N, Ishihara T, Yamagami K, et al. Macro TSH in patients with subclinical hypothyroidism. Clin Endocrinol (Oxf). 2015; 83(6): 923–930.
  66. Ghazal K, Brabant S, Prie D, et al. Hormone Immunoassay Interference: A 2021 Update. Ann Lab Med. 2022; 42(1): 3–23.
  67. Paczkowska K, Otlewska A, Loska O, et al. Laboratory interference in the thyroid function test. Endokrynol Pol. 2020; 71(6): 551–560.
  68. Koulouri O, Moran C, Halsall D, et al. Pitfalls in the measurement and interpretation of thyroid function tests. Best Pract Res Clin Endocrinol Metab. 2013; 27(6): 745–762.
  69. Favresse J, Burlacu MC, Maiter D, et al. Interferences With Thyroid Function Immunoassays: Clinical Implications and Detection Algorithm. Endocr Rev. 2018; 39(5): 830–850.
  70. Moerman A, Delanghe JR. Sense and nonsense concerning biotin interference in laboratory tests. Acta Clin Belg. 2022; 77(1): 204–210.
  71. Odhaib SA, Mansour AA, Haddad NS. How Biotin Induces Misleading Results in Thyroid Bioassays: Case Series. Cureus. 2019; 11(5): e4727.
  72. Sharma H, Sahlot R, Purwar N, et al. Co-existence of type 1 diabetes and other autoimmune ailments in subjects with autoimmune thyroid disorders. Diabetes Metab Syndr. 2022; 16(2): 102405.
  73. Zeng MF, Chen LiLi, Ye HY, et al. Primary hypothyroidism and isolated ACTH deficiency induced by nivolumab therapy: Case report and review. Medicine (Baltimore). 2017; 96(44): e8426.
  74. Ohara N, Kobayashi M, Ohashi K, et al. Isolated adrenocorticotropic hormone deficiency and thyroiditis associated with nivolumab therapy in a patient with advanced lung adenocarcinoma: a case report and review of the literature. J Med Case Rep. 2019; 13(1): 88.
  75. Sánchez J, Cohen M, Zapater JL, et al. Primary Adrenal Insufficiency After COVID-19 Infection. AACE Clin Case Rep. 2022; 8(2): 51–53.
  76. Durcan E, Hacioglu A, Karaca Z, et al. Hypothalamic-Pituitary Axis Function and Adrenal Insufficiency in COVID-19 Patients. Neuroimmunomodulation. 2023; 30(1): 215–225.
  77. Kanczkowski W, Gaba WH, Krone N, et al. Adrenal Gland Function and Dysfunction During COVID-19. Horm Metab Res. 2022; 54(8): 532–539.
  78. Thavaraputta S, Dennis JA, Laoveeravat P, et al. Hypothyroidism and Its Association With Sleep Apnea Among Adults in the United States: NHANES 2007-2008. J Clin Endocrinol Metab. 2019; 104(11): 4990–4997.
  79. Bode H, Ivens B, Bschor T, et al. Hyperthyroidism and clinical depression: a systematic review and meta-analysis. Transl Psychiatry. 2022; 12(1): 362.
  80. Tang R, Wang J, Yang L, et al. Subclinical Hypothyroidism and Depression: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne). 2019; 10: 340.
  81. Wildisen L, Feller M, Del Giovane C, et al. Effect of Levothyroxine Therapy on the Development of Depressive Symptoms in Older Adults With Subclinical Hypothyroidism: An Ancillary Study of a Randomized Clinical Trial. JAMA Netw Open. 2021; 4(2): e2036645.
  82. Jørgensen P, Langhammer A, Krokstad S, et al. Diagnostic labelling influences self-rated health. A prospective cohort study: the HUNT Study, Norway. Fam Pract. 2015; 32(5): 492–499.
  83. Fan H, Ren Q, Sheng Z, et al. The role of the thyroid in polycystic ovary syndrome. Front Endocrinol (Lausanne). 2023; 14: 1242050.
  84. Pasandideh R, Hosseini SM, Veghari G, et al. The Effects of 8 Weeks of Levothyroxine Replacement Treatment on Metabolic and Anthropometric Indices of Insulin Resistance in Hypothyroid Patients. Endocr Metab Immune Disord Drug Targets. 2020; 20(5): 745–752.
  85. Krysiak R, Basiak M, Okopień B. Insulin resistance attenuates the impact of levothyroxine on thyroid autoimmunity and hypothalamic-pituitary-thyroid axis activity in women with autoimmune subclinical hypothyroidism. Clin Exp Pharmacol Physiol. 2021; 48(9): 1215–1223.
  86. Bianco AC, da Conceição RR. The Deiodinase Trio and Thyroid Hormone Signaling. Methods Mol Biol. 2018; 1801: 67–83.
  87. Halsall DJ, Oddy S. Clinical and laboratory aspects of 3,3',5'-triiodothyronine (reverse T3). Ann Clin Biochem. 2021; 58(1): 29–37.
  88. Naiyer AJ, Shah J, Hernandez L, et al. Tissue transglutaminase antibodies in individuals with celiac disease bind to thyroid follicles and extracellular matrix and may contribute to thyroid dysfunction. Thyroid. 2008; 18(11): 1171–1178.
  89. Ruggeri RM, Giovinazzo S, Barbalace MC, et al. Influence of Dietary Habits on Oxidative Stress Markers in Hashimoto's Thyroiditis. Thyroid. 2021; 31(1): 96–105.
  90. Virili C, Fallahi P, Antonelli A, et al. Gut microbiota and Hashimoto's thyroiditis. Rev Endocr Metab Disord. 2018; 19(4): 293–300.
  91. Knezevic J, Starchl C, Tmava Berisha A, et al. Thyroid-Gut-Axis: How Does the Microbiota Influence Thyroid Function? Nutrients. 2020; 12(6).
  92. Virili C, Stramazzo I, Bagaglini MF, et al. The relationship between thyroid and human-associated microbiota: A systematic review of reviews. Rev Endocr Metab Disord. 2024; 25(1): 215–237.
  93. Liu J, Qin X, Lin B, et al. Analysis of gut microbiota diversity in Hashimoto's thyroiditis patients. BMC Microbiol. 2022; 22(1): 318.
  94. Zawadzka K, Kałuzińska K, Świerz MJ, et al. Are probiotics, prebiotics, and synbiotics beneficial in primary thyroid diseases? A systematic review with meta-analysis. Ann Agric Environ Med. 2023; 30(2): 217–223.
  95. Klasson CL, Sadhir S, Pontzer H. Daily physical activity is negatively associated with thyroid hormone levels, inflammation, and immune system markers among men and women in the NHANES dataset. PLoS One. 2022; 17(7): e0270221.
  96. Roa Dueñas OH, Koolhaas C, Voortman T, et al. Thyroid Function and Physical Activity: A Population-Based Cohort Study. Thyroid. 2021; 31(6): 870–875.
  97. Ferrante M, Distefano G, Distefano C, et al. Benefits of Physical Activity during and after Thyroid Cancer Treatment on Fatigue and Quality of Life: A Systematic Review. Cancers (Basel). 2022; 14(15).
  98. Murugathasan M, Jafari A, Amandeep A, et al. Moderate exercise induces trained immunity in macrophages. Am J Physiol Cell Physiol. 2023; 325(2): C429–C442.
  99. Werneck FZ, Coelho EF, Almas SP, et al. Exercise training improves quality of life in women with subclinical hypothyroidism: a randomized clinical trial. Arch Endocrinol Metab. 2018; 62(5): 530–536.
  100. Kim MJ, Moon S, Oh BC, et al. Association Between Diethylhexyl Phthalate Exposure and Thyroid Function: A Meta-Analysis. Thyroid. 2019; 29(2): 183–192.
  101. Kwon JA, Shin B, Kim B. Urinary bisphenol A and thyroid function by BMI in the Korean National Environmental Health Survey (KoNEHS) 2012-2014. Chemosphere. 2020; 240: 124918.
  102. Milczarek-Banach J, Rachoń D, Bednarczuk T, et al. Exposure to Bisphenol A Analogs and the Thyroid Function and Volume in Women of Reproductive Age-Cross-Sectional Study. Front Endocrinol (Lausanne). 2020; 11: 587252.
  103. Beg MA, Sheikh IA. Endocrine disruption: Molecular interactions of environmental bisphenol contaminants with thyroid hormone receptor and thyroxine-binding globulin. Toxicol Ind Health. 2020; 36(5): 322–335.
  104. Rayman MP. Multiple nutritional factors and thyroid disease, with particular reference to autoimmune thyroid disease. Proc Nutr Soc. 2019; 78(1): 34–44.
  105. Wichman J, Winther KH, Bonnema SJ, et al. Selenium Supplementation Significantly Reduces Thyroid Autoantibody Levels in Patients with Chronic Autoimmune Thyroiditis: A Systematic Review and Meta-Analysis. Thyroid. 2016; 26(12): 1681–1692.
  106. Qiu Y, Xing Z, Xiang Q, et al. Insufficient evidence to support the clinical efficacy of selenium supplementation for patients with chronic autoimmune thyroiditis. Endocrine. 2021; 73(2): 384–397.
  107. Wang YS, Liang SS, Ren JJ, et al. The Effects of Selenium Supplementation in the Treatment of Autoimmune Thyroiditis: An Overview of Systematic Reviews. Nutrients. 2023; 15(14).
  108. Winther KH, Papini E, Attanasio R, et al. A 2018 European Thyroid Association Survey on the Use of Selenium Supplementation in Hashimoto's Thyroiditis. Eur Thyroid J. 2020; 9(2): 99–105.
  109. Luo J, Wang X, Yuan Li, et al. Iron Deficiency, a Risk Factor of Thyroid Disorders in Reproductive-Age and Pregnant Women: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne). 2021; 12: 629831.
  110. Garofalo V, Condorelli RA, Cannarella R, et al. Relationship between Iron Deficiency and Thyroid Function: A Systematic Review and Meta-Analysis. Nutrients. 2023; 15(22).
  111. Cappellini MD, Musallam KM, Taher AT. Iron deficiency anaemia revisited. J Intern Med. 2020; 287(2): 153–170.
  112. Moncayo R, Moncayo H. The WOMED model of benign thyroid disease: Acquired magnesium deficiency due to physical and psychological stressors relates to dysfunction of oxidative phosphorylation. BBA Clin. 2015; 3: 44–64.
  113. Wang K, Wei H, Zhang W, et al. Severely low serum magnesium is associated with increased risks of positive anti-thyroglobulin antibody and hypothyroidism: A cross-sectional study. Sci Rep. 2018; 8(1): 9904.
  114. Moncayo R, Moncayo H. Practical Guidelines for Diagnosing and Treating Thyroid Disease Based on the WOMED Metabolic Model of Disease Focusing on Glycolysis and Coenzyme Q Deficiency-A Clinical Alternative to the 2021 Retired Clinical Practice Guidelines of the Endocrine Society. Diagnostics (Basel). 2022; 12(1).
  115. Beserra JB, Morais JB, Severo JS, et al. Relation Between Zinc and Thyroid Hormones in Humans: a Systematic Review. Biol Trace Elem Res. 2021; 199(11): 4092–4100.
  116. Patel AM, Khan S, Inam AM, et al. Determination of Serum Zinc and Phosphorus Levels in Patients with Hypothyroidism. Biol Trace Elem Res. 2024; 202(7): 3018–3024.
  117. Zavros A, Giannaki CD, Aphamis G, et al. The Effects of Zinc and Selenium Supplementation on Body Composition and Thyroid Function in Individuals with Overweight or Obesity: A Systematic Review. J Diet Suppl. 2023; 20(4): 643–671.
  118. Bucci I, Napolitano G, Giuliani C, et al. Zinc sulfate supplementation improves thyroid function in hypozincemic Down children. Biol Trace Elem Res. 1999; 67(3): 257–268.
  119. Turashvili N, Javashvili L, Giorgadze E. "Vitamin D Deficiency Is More Common in Women with Autoimmune Thyroiditis: A Retrospective Study". Int J Endocrinol. 2021; 2021: 4465563.
  120. Krysiak R, Szkróbka W, Okopień B. The Effect of Vitamin D on Thyroid Autoimmunity in Levothyroxine-Treated Women with Hashimoto's Thyroiditis and Normal Vitamin D Status. Exp Clin Endocrinol Diabetes. 2017; 125(4): 229–233.
  121. Jiang H, Chen X, Qian X, et al. Effects of vitamin D treatment on thyroid function and autoimmunity markers in patients with Hashimoto's thyroiditis-A meta-analysis of randomized controlled trials. J Clin Pharm Ther. 2022; 47(6): 767–775.
  122. Murdaca G, Tonacci A, Negrini S, et al. Emerging role of vitamin D in autoimmune diseases: An update on evidence and therapeutic implications. Autoimmun Rev. 2019; 18(9): 102350.
  123. Altieri B, Muscogiuri G, Barrea L, et al. Does vitamin D play a role in autoimmune endocrine disorders? A proof of concept. Rev Endocr Metab Disord. 2017; 18(3): 335–346.
  124. Gupta R, Choudhary S, Chatterjee T. A Study on Vitamin B12 Levels in Hypothyroid Patients Presenting to a Tertiary Care Teaching Hospital. Cureus. 2023; 15(8): e44197.
  125. Issac TG, Soundarya S, Christopher R, et al. Vitamin B12 deficiency: an important reversible co-morbidity in neuropsychiatric manifestations. Indian J Psychol Med. 2015; 37(1): 26–29.
  126. Tamura J, Kubota K, Murakami H, et al. Immunomodulation by vitamin B12: augmentation of CD8+ T lymphocytes and natural killer (NK) cell activity in vitamin B12-deficient patients by methyl-B12 treatment. Clin Exp Immunol. 1999; 116(1): 28–32.
  127. Yang W, Jin C, Wang H, et al. Subclinical hypothyroidism increases insulin resistance in normoglycemic people. Front Endocrinol (Lausanne). 2023; 14: 1106968.
  128. Mazaheri T, Sharifi F, Kamali K. Insulin resistance in hypothyroid patients under Levothyroxine therapy: a comparison between those with and without thyroid autoimmunity. J Diabetes Metab Disord. 2014; 13(1): 103.
  129. Blaslov K, Gajski D, Vucelić V, et al. The Association Of Subclinical Insulin Resistance with Thyroid Autoimmunity in Euthyroid Individuals. Acta Clin Croat. 2020; 59(4): 696–702.
  130. Jia Xi, Zhai T, Zhang JA. Metformin reduces autoimmune antibody levels in patients with Hashimoto's thyroiditis: A systematic review and meta-analysis. Autoimmunity. 2020; 53(6): 353–361.
  131. Fallahi P, Ferrari SM, Elia G, et al. Myo-inositol in autoimmune thyroiditis, and hypothyroidism. Rev Endocr Metab Disord. 2018; 19(4): 349–354.
  132. Corvilain B, Laurent E, Lecomte M, et al. Role of the cyclic adenosine 3',5'-monophosphate and the phosphatidylinositol-Ca2+ cascades in mediating the effects of thyrotropin and iodide on hormone synthesis and secretion in human thyroid slices. J Clin Endocrinol Metab. 1994; 79(1): 152–159.
  133. Ferrari SM, Elia G, Ragusa F, et al. The protective effect of myo-inositol on human thyrocytes. Rev Endocr Metab Disord. 2018; 19(4): 355–362.
  134. Nordio M, Basciani S. Myo-inositol plus selenium supplementation restores euthyroid state in Hashimoto’s patients with subclinical hypothyroidism. Eur Rev Med Pharmacol Sci. 2017; 21(2_Suppl): 51–59.
  135. Payer J, Jackuliak P, Kužma M, et al. Supplementation with myo-inositol and Selenium improves the clinical conditions and biochemical features of women with or at risk for subclinical hypothyroidism. Front Endocrinol (Lausanne). 2022; 13: 1067029.
  136. Hosny EN, El-Gizawy MM, Sawie HG, et al. Neuroprotective Effect of Ashwagandha Extract against the Neurochemical Changes Induced in Rat Model of Hypothyroidism. J Diet Suppl. 2021; 18(1): 72–91.
  137. Sharma AK, Basu I, Singh S. Efficacy and Safety of Ashwagandha Root Extract in Subclinical Hypothyroid Patients: A Double-Blind, Randomized Placebo-Controlled Trial. J Altern Complement Med. 2018; 24(3): 243–248.
  138. Langade D, Thakare V, Kanchi S, et al. Clinical evaluation of the pharmacological impact of ashwagandha root extract on sleep in healthy volunteers and insomnia patients: A double-blind, randomized, parallel-group, placebo-controlled study. J Ethnopharmacol. 2021; 264: 113276.
  139. Salve J, Pate S, Debnath K, et al. Adaptogenic and Anxiolytic Effects of Ashwagandha Root Extract in Healthy Adults: A Double-blind, Randomized, Placebo-controlled Clinical Study. Cureus. 2019; 11(12): e6466.
  140. Chauhan S, Srivastava MK, Pathak AK. Effect of standardized root extract of ashwagandha () on well-being and sexual performance in adult males: A randomized controlled trial. Health Sci Rep. 2022; 5(4): e741.
  141. Ajgaonkar A, Jain M, Debnath K. Efficacy and Safety of Ashwagandha (Withania somnifera) Root Extract for Improvement of Sexual Health in Healthy Women: A Prospective, Randomized, Placebo-Controlled Study. Cureus. 2022; 14(10): e30787.
  142. Speers AB, Cabey KA, Soumyanath A, et al. Effects of (Ashwagandha) on Stress and the Stress- Related Neuropsychiatric Disorders Anxiety, Depression, and Insomnia. Curr Neuropharmacol. 2021; 19(9): 1468–1495.
  143. Midgley JEM, Larisch R, Dietrich JW, et al. Variation in the biochemical response to l-thyroxine therapy and relationship with peripheral thyroid hormone conversion efficiency. Endocr Connect. 2015; 4(4): 196–205.
  144. Carlé A, Faber J, Steffensen R, et al. Hypothyroid Patients Encoding Combined MCT10 and DIO2 Gene Polymorphisms May Prefer L-T3 + L-T4 Combination Treatment - Data Using a Blind, Randomized, Clinical Study. Eur Thyroid J. 2017; 6(3): 143–151.
  145. van der Deure WM, Appelhof BC, Peeters RP, et al. Polymorphisms in the brain-specific thyroid hormone transporter OATP1C1 are associated with fatigue and depression in hypothyroid patients. Clin Endocrinol (Oxf). 2008; 69(5): 804–811.
  146. Arici M, Oztas E, Yanar F, et al. Association between genetic polymorphism and levothyroxine bioavailability in hypothyroid patients. Endocr J. 2018; 65(3): 317–323.
  147. Al-Azzam SI, Alzoubi KH, Khabour O, et al. The associations of polymorphisms of TSH receptor and thyroid hormone receptor genes with L-thyroxine treatment in hypothyroid patients. Hormones (Athens). 2014; 13(3): 389–397.
  148. Lee KW, Shin Y, Lee S, et al. Inherited Disorders of Thyroid Hormone Metabolism Defect Caused by the Dysregulation of Selenoprotein Expression. Front Endocrinol (Lausanne). 2021; 12: 803024.
  149. Fujisawa H, Korwutthikulrangsri M, Fu J, et al. Role of the Thyroid Gland in Expression of the Thyroid Phenotype of Sbp2-Deficient Mice. Endocrinology. 2020; 161(5).
  150. Mizuma T, Watanabe M, Inoue N, et al. Association of the polymorphisms in the gene encoding thyroglobulin with the development and prognosis of autoimmune thyroid disease. Autoimmunity. 2017; 50(6): 386–392.
  151. Tomari S, Watanabe M, Inoue N, et al. The polymorphisms in the thyroid peroxidase gene were associated with the development of autoimmune thyroid disease and the serum levels of anti-thyroid peroxidase antibody. Endocr J. 2017; 64(10): 1025–1032.
  152. Bianco AC, Kim BS. Pathophysiological relevance of deiodinase polymorphism. Curr Opin Endocrinol Diabetes Obes. 2018; 25(5): 341–346.
  153. Zhang X, Sun J, Han W, et al. The Is Associated with Worse Glycemic Control in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. J Diabetes Res. 2016; 2016: 5928726.
  154. Wang X, Chen K, Zhang C, et al. The Type 2 Deiodinase Thr92Ala Polymorphism Is Associated with Higher Body Mass Index and Fasting Glucose Levels: A Systematic Review and Meta-Analysis. Biomed Res Int. 2021; 2021: 9914009.
  155. Jo S, Fonseca TL, Bocco BM, et al. Type 2 deiodinase polymorphism causes ER stress and hypothyroidism in the brain. J Clin Invest. 2019; 129(1): 230–245.
  156. Caron P, Grunenwald S, Persani L, et al. Factors influencing the levothyroxine dose in the hormone replacement therapy of primary hypothyroidism in adults. Rev Endocr Metab Disord. 2022; 23(3): 463–483.
  157. Young Cho Y, Jeong Kim H, Won Jang H, et al. The relationship of 19 functional polymorphisms in iodothyronine deiodinase and psychological well-being in hypothyroid patients. Endocrine. 2017; 57(1): 115–124.
  158. Wouters HJ, van Loon HCM, van der Klauw MM, et al. No Effect of the Thr92Ala Polymorphism of Deiodinase-2 on Thyroid Hormone Parameters, Health-Related Quality of Life, and Cognitive Functioning in a Large Population-Based Cohort Study. Thyroid. 2017; 27(2): 147–155.
  159. Millan-Alanis JM, González-González JG, Flores-Rodríguez A, et al. Benefits and Harms of Levothyroxine/L-Triiodothyronine Versus Levothyroxine Monotherapy for Adult Patients with Hypothyroidism: Systematic Review and Meta-Analysis. Thyroid. 2021; 31(11): 1613–1625.
  160. Jonklaas J, Bianco AC, Cappola AR, et al. Evidence-Based Use of Levothyroxine/Liothyronine Combinations in Treating Hypothyroidism: A Consensus Document. Thyroid. 2021; 31(2): 156–182.
  161. Weihs A, Chaker L, Martin TC, et al. Epigenome-Wide Association Study Reveals CpG Sites Associated with Thyroid Function and Regulatory Effects on . Thyroid. 2023; 33(3): 301–311.
  162. Lafontaine N, Campbell PJ, Castillo-Fernandez JE, et al. Epigenome-Wide Association Study of Thyroid Function Traits Identifies Novel Associations of fT3 With KLF9 and DOT1L. J Clin Endocrinol Metab. 2021; 106(5): e2191–e2202.
  163. Drepanos L, Gans IM, Grendler J, et al. Loss of Krüppel-like factor 9 deregulates both physiological gene expression and development. Sci Rep. 2023; 13(1): 12239.
  164. Wen L, Fu L, Shi YB. Histone methyltransferase Dot1L is a coactivator for thyroid hormone receptor during development. FASEB J. 2017; 31(11): 4821–4831.
  165. Jaimes-Hoy L, Pérez-Maldonado A, Narváez Bahena E, et al. Sex Dimorphic Changes in Trh Gene Methylation and Thyroid-Axis Response to Energy Demands in Maternally Separated Rats. Endocrinology. 2021; 162(8).
  166. Arakawa Y, Watanabe M, Inoue N, et al. Association of polymorphisms in DNMT1, DNMT3A, DNMT3B, MTHFR and MTRR genes with global DNA methylation levels and prognosis of autoimmune thyroid disease. Clin Exp Immunol. 2012; 170(2): 194–201.
  167. Kim S, Cho YH, Won S, et al. Maternal exposures to persistent organic pollutants are associated with DNA methylation of thyroid hormone-related genes in placenta differently by infant sex. Environ Int. 2019; 130: 104956.
  168. Mendoza A, Hollenberg AN. New insights into thyroid hormone action. Pharmacol Ther. 2017; 173: 135–145.
  169. Guo Q, Wu Y, Hou Y, et al. Cytokine Secretion and Pyroptosis of Thyroid Follicular Cells Mediated by Enhanced NLRP3, NLRP1, NLRC4, and AIM2 Inflammasomes Are Associated With Autoimmune Thyroiditis. Front Immunol. 2018; 9: 1197.
  170. Cui X, Liu Y, Wang S, et al. Circulating Exosomes Activate Dendritic Cells and Induce Unbalanced CD4+ T Cell Differentiation in Hashimoto Thyroiditis. J Clin Endocrinol Metab. 2019; 104(10): 4607–4618.