GET THE APP

Journal of Thyroid Disorders & Therapy

Journal of Thyroid Disorders & Therapy
Open Access

ISSN: 2167-7948

+44 1300 500008

Research Article - (2021)Volume 10, Issue 6

Etiopathogenetic Mechanisms in Hashimoto’s Autoimmune Thyroid Disease

Isabel García1* and Sergio Abanades2
 
*Correspondence: Isabel García, Department of Endocrinology and Metabolism, Institute for Integrative and Conscious Health (ISIC), París, 3r 1a Barcelona, Spain, Email:

Author info »

Abstract

Hashimoto’s disease (HD) is an autoimmune thyroid disease often leading to hypothyroidism. The current therapy for these patients consists in traditional Hormone Replacement Therapy (HRT) to compensate inadequate thyroid function. However, clinical management of this and other autoimmune disorders usually dismisses the etiologic component, even when the gold-standard pharmacological treatment is not effective and/or the patient remains asymptomatic. Here, we propose to dig into the etiopathogenic factors as an attempt to unravel patient’s negative response to therapy and to improve therapeutic and preventive approaches as standard medical practice as well as to complement HRT. These factors may include genetic susceptibility, intestinal permeability defects, other pathologies like celiac disease, gluten intolerance and biopsychosocialstress, gender, sex hormones and micronutrients (vitamin D, iodine and selenium). Exposure to environmental toxic substances (heavy metals and others) and infectious agents (both viral and bacterial) may also trigger HD development. Finally, given the implication of multiple mechanisms of etiology and pathogenesis, a multidisciplinary strategy may be required to deliver integrated and successful patient care.

Keywords

Hashimoto’s disease; Autoimmune disease; Thyroid destruction; Genetic susceptibility; Environmental factors

Abbrevations

Hashimoto’s disease (HD); Anti-Thyroid Peroxidase Antibodies (anti-TPO); Anti-Thyroglobulin Antibodies (anti-Tg); Hormone Replacement Therapy (HRT); Thyroglobulin (Tg); Small Intestinal Bacterial Overgrowth (SIBO); Bisphenol A (BPA); Glutathione-S-Transferase (GST); Celiac Disease (CD); Non-Celiac Gluten Sensitivity (NCGS); T helper (Th); Thyrotropin- Releasing Hormone (HRT)

Introduction

Autoimmune thyroiditis or Hashimoto's Disease (HD) is an autoimmune disease characterized by an autoantibody-mediated destruction of the thyroid gland (anti-thyroid peroxidase (anti- TPO) and anti-thyroglobulin (anti-Tg) antibodies) via and immune response mediated by T-lymphocytes. Although this leads to distinct levels of thyroid hypofunction, clinical manifestations may range from diffuse or nodular goiter with euthyroidism, subclinical hypothyroidism and permanent hypothyroidism [1].

HD was named after the Japanese physician Hakaru Hashimoto who made its first description in 1912 and referred to it as lymphomatous goiter [2]. Nowadays, autoimmune hypothyroidism has achieved estimated rates of incidence as high as 2.2/100,000/year (males) and 498.4/100,000/year (females) worldwide and they will continue to increase. The same trend is observed for all types of autoimmune thyroid disease, for which women have a higher incidence compared to men [3].

Materials and Methods

The hypothesis

As a result of this exponentially increased incidence, much attention has been drawn to the etiopathogenesis of autoimmune diseases [4]. However, the current diagnostic and therapeutic approach for HD takes into consideration the hormonal deficit upon glandular destruction almost exclusively, disregarding the possible pathogenic mechanisms that may be involved in the initiation and maintenance of the disease [5]. This way, conventional Hormone Replacement Therapy (HRT), including periodical follow-up and dose adjustment, still constitutes the first-line treatment, despite its poor efficacy in some patients [6-7]. In most patients, traditional HRT is able to relieve hypothyroidism symptoms. Nevertheless, an important percentage of them remain symptomatic in spite of proper patient adherence [6-7]. Indeed, some studies have reported that in up to 15% of the patients with normal Thyroid Stimulating Hormone (TSH) values, T3 levels fall below the normal range, which may be an explanation for the persistence of symptoms [8-9]. A possible mechanism behind this observation may be a deficient T4 thyroid hormone conversion into biologically active T3 in peripheral tissues, the so-called “functional hypothyroidism”. As a result, despite minimal intracellular T3 concentration, in the absence of plasmatic T3 fluctuations, the hypothalamuspituitary- adrenal axis feedback mechanism will not be triggered, thereby TSH concentration will remain stable [10].

Results and Discussion

Hence, we hypothesise that HD is caused by a combination of etiologic and susceptibility factors and that these factors are associated to the development and persistence of the disease and also in the persistence of symptoms in apparently correctly treated patients. In light of this, we propose to explore the etiologic factors surrounding HD to improve disease prophylaxis, to better understand treatment-refractory clinical cases, and to eventually design more precise and personalized therapeutic approaches, alone or in combination with hormone replacement methods. This could be the basis of a novel integrative therapeutic approach and an effective complement to the usual repositioning treatment.

Evaluation of the hypothesis

In general, three main factors have been involved in the development of autoimmune diseases: A genetically altered immune system, a reactive mucous membraneassociated immunity (mucosal immunity) against environmental antigens, and an increased intestinal permeability that promotes their entry into the systemic circulation [11-12] (Figure 1). Similarly, HD is driven by the following suggested key factors (Figure 2).

Etiopathogenesis

Figure 1: Etiopathogenesis of autoimmune diseases. Genetic susceptibility, certain environmental antigens interacting with mucosal immunity, and intestinal permeability (mainly due to dysbiosis) are the three main factors involved in disease etiology.

Hashimoto

Figure 2: Key factors involved in Hashimoto’s disease. Several and non-mutually exclusive factors may be linked to the disease origin and development. CD, celiac disease.

Genetic factors: Multiple susceptibility genes may be involved in HD pathogenesis. Some of them are common among other autoimmune diseases, while certain genes are specific for thyroid autoimmunity. The first genetic locus identified in association with HD was the region of the major histocompatibility complex (or HLA). In Caucasian individuals, different HD subtypes have been associated with certain HLA. Alleles including DR3, DR5, DQ7, DQB1*03, DQw7 or the DRB1*04-DQB1*0301 haplotype. The associations with DRB4*0101, HLA-A2 and DRw53 and with DRw9 have been proven in the Japanese and Chinese population, respectively [13]. CTLA-4, PTPN22 and VDR genes also play an important role in the etiology of HD, other autoimmune diseases and, in the case of VDR, organspecific pathologies such as type-1-diabetes or Addison's disease. Nevertheless, Thyroglobulin (Tg) gene is the only HD-associated thyroid specific gene known so far, which also confers susceptibility to Graves-Basedow disease [13].

Intestinal permeability, dysbiosis and small intestinal bacterial overgrowth (SIBO): Intestinal permeability occurs when the intestinal barrier function is lost. In physiological conditions, it selectively allows nutrient, electrolyte and water absorption while preventing the entry of intraluminal toxins, foreign antigens and microorganisms [14]. Physical components, such as intercellular tight junctions, as well as biochemical and immunological factors are highly coordinated to maintain this intestinal barrier system. However, when integrity is compromised, harmful substances may cross the epithelium, disseminate through the bloodstream and lymphatic system and eventually affect tissue homeostasis [14]. Intestinally derived inflammatory mediators could lead to a systemic inflammatory and autoimmune response. Thus, increased intestinal permeability due to multiple putative causes has been postulated an essential trigger of an autoimmune response [14-15] (Table 1).

Risk factors associated  with  increased intestinal permeability [12,15,17]
Intestinal inflammation
Oxidative stress
Stress
Nonsteroidal anti-inflammatory drugs
Alcohol consumption
Cow’s milk intolerance
SIBO
Sepsis
Pancreatic insufficiency
Intestinal infections
Obstructive jaundice
Surgery

Table 1: Risk factors associated with increased intestinal permeability.

A healthy gut microbiota composition is also crucial to prevent intestinal permeability, although it may be altered by a diet rich in saturated fats and poor in fibre, the presence of environmental toxins and psycho-emotional stress [14]. Immune and dendritic cell dysregulation is one important consequence of dysbiosis-associated increased permeability. In fact, dendritic cells have a role in oral tolerance through the induction of specific integrin and chemokine receptors and their involvement in autoimmune thyroiditis have been shown [16]. Interestingly, the presence of intestinal dysbiosis have been associated to other conditions such as inflammatory bowel disease and/or type 1 diabetes [16]. Small Intestinal Bacterial Overgrowth (SIBO) is characterized by the presence of an abnormally high number of coliform bacteria in the small bowel, in conjunction with several gastrointestinal symptoms as diarrhoea, constipation and meteorism [17]. An increased prevalence of SIBO has been demonstrated in patients with autoimmune diseases such as multiple sclerosis, type 1 diabetes, and rheumatoid arthritis [18-20]. SIBO has been associated with GI’s dysmotility and hypochlorhydria and a can also induce intestinal permeability. Therefore, perpetuating the leakage of luminal toxins and inflammatory mediators associated with autoimmune disorders [21]. Additionally, a worsening of thyroid function parameters and an increase in anti-TPO antibodies it has also been observed in the presence of SIBO, therefore contributing with the induction of autoimmunity and hypothyroidism [22]. Furthermore, hypothyroidism and levothyroxine treatment have been both linked to the induction of SIBO and gastrointestinal dysmotility [23-24].

Infectious agents: Microorganism infections may be the leading cause of HD. Several types of microorganisms have been identified and at least 5 different mechanisms have been proposed for microorganism-induced autoimmunity. The main ones are thyroid protein molecular mimicry and the thyroid’s gland immune attack mediated by superantigens [25].

The Epstein Barr Virus (EBV) is one of the most important players in autoimmune thyroiditis onset and persistence of symptoms [26-29], both during the primary infection and upon reactivation and/or viral chronification. Increased activation of EBV in HD patients seems related to a CD8 T-cell deficiency, which underpins disease progression [30]. Additional microorganisms have been also involved such cytomegalovirus, Herpes Simplex and hepatitis C virus, B19 parvovirus, Yersinia enterocolitica, Borrelia Burgdorferi, Candida albicans and Toxoplasma gondii [31-38].

Exposure to environmental toxic substances: A growing pile of evidence supports the role of toxic substances as other important environmental triggers of autoimmune thyroiditis [39]. Exposure to Polychlorinated Biphenyls (PCB), dioxin and heavy metals has been linked to high levels of anti-Tg and anti- TPO autoantibodies [39]. Detection of Bisphenol A (BPA) in plasma has also been correlated with anti-TPO production [40]. In urine, an increased phthalate and BPA concentration has been associated with an alteration of circulating thyroid hormones [41]. Finally, exposure to phthalates and an exacerbation of autoimmune thyroid disease occurred concomitantly [42].

Environmental toxins may damage the intestinal mucosa and directly interact with follicular thyroid and immune cells. From a molecular point of view, although heavy metals exhibit distinct toxicity mechanisms, they all ultimately induce oxidative damage through the formation of Deoxy Ribonucleic Acid (DNA) and/or protein adducts, thereby impairing key respiratory enzyme activities [43]. Plasma and urine levels of mercury are not proportional to its detrimental effects in some autoimmune disorders; however in HD they are directly connected to lowered thyroid hormone concentration and the presence of anti-Tg antibodies in plasma [44-47]. Thyroid peroxidases, which catalyze the synthesis of T3, T4 are potent thyroid antioxidants which function depends on selenium concentration. Due to its extremely high affinity, mercury binds to selenium acting as an important chelator of selenium. Therefore, in the event of chronic mercury intoxication, selenium bioavailability may be reduced, consequently causing extensive cytotoxicity [48]. In addition, mercury impairs the protective antioxidant function of glutathione [49]. Autoantibodies are typically found in patients with other autoimmune diseases who have been chronically exposed to mercury. Specifically, it seems that inorganic mercury would enhance the production of autoimmune disease markers to a greater extent than organic forms [44]. However, the impact of mercury exposure on different clinical variables is a question that still remains unanswered, with studies showing conflicting results [50]. In recent years, increased susceptibility to mercury and BPA prenatal exposure have been attributed to certain genetic polymorphisms, being the Glutathione-S-Transferase (GST) GSTT1-/- and GSTM1-/- polymorphisms especially relevant as they hamper toxin excretion [51,52].

Vitamin D deficiency: A recent meta-analysis demonstrates a clear vitamin D deficiency in patients with HD [53], which may favour autoimmunity mechanisms such as the loss of tolerance to self-antigens, the production of autoantibodies and direct cell damage [54].

Celiac disease/Gluten intolerance: There is an increased prevalence of Celiac Disease (CD) in patients with autoimmune thyroid disease as revealed by a recent meta-analysis [55]. This could be partially explained by the increased immunosensitivity of celiac patients, as part of an autoimmune polyglandular syndrome, by a CD malabsorption-associated iodine and selenium deficiency or due to antibodies affecting both intestinal and thyroid tissues [4]. In addition, in non-celiac patients, gluten (gliadins and glutenins) and other wheat proteins are capable of generating intestinal permeability by directly damaging the intestinal mucosa [56-57]. The Non-Celiac Gluten Sensitivity (NCGS), also known as gluten intolerance or sensitivity to gluten, shares some clinical symptoms with irritable bowel syndrome in addition to some systemic ones such as headaches, arthromyalgia, asthenia, and mood alterations. Its prevalence ranges between 0.6%-6% of the population and diagnosis is established once CD and wheat allergy have been ruled out, i.e. patients do not present typical antibodies or pathognomonic lesions of CD in the intestinal mucosa. Still, a gluten-free diet leads to regression of the symptoms [58]. Interestingly, NCGS has been associated with the onset of HD and the consequent elevation of antithyroid antibodies. In fact, some of the symptoms of NCGS may also occur during the course of HD [59].

Excess of iodine and/or low Selenium:Iodine ratio: Iodine is absolutely required for thyroid hormone synthesis. It comes as no surprise, therefore, that insufficient iodine intake is directly correlated with a weakened hormone production. Excess iodine, however, may also lead to adverse effects depending on the underlying thyroid function, as well as the extent and duration of the excessive intake. In this regard, the rise in thyroid autoantibodies, and even thyroid autoimmune disease, may be triggered by iodine supplementation [60-63]. A similar response to iodine has been reported in studies using mice genetically predisposed to thyroid autoimmunity.

The mechanism of HD occurrence in response to iodine supplementation is not fully understood. Several mechanisms have been hypothesized such as greater immunogenicity of iodinated thyroglobulin and a direct toxic effect of iodine on thyroid cells through the increased generation of oxygen free radicals, which are byproducts of iodine peroxidation during thyroid hormone synthesis [60-63]. Glutathione peroxidase and thioredoxin oxidase are selenium-dependent enzymes that protect tissues from the harmful effect of reactive oxygen species (especially hydrogen peroxide) [63]. An excess of iodine in the context of selenium deficiency seems to reduce the activity of these and other selenoproteins [64-65] and, consequently, to enhance the accumulation of free radicals and to promote an inflammatory process within the thyroid tissue [63]. Nevertheless, their enzymatic activity may be reversed upon selenium administration [64]. A low (Selenium:Iodine) ratio also affects autoantibody production and causes a shift in the population of immune cells towards a deficit of regulatory T lymphocytes. Again, both effects may be restored by selenium supplementation [64,66-68].

Gender and hormonal factors: Gender greatly influences the development of autoimmune diseases. Numerous epidemiological studies have shown a prevalence of up to 3:1 (female:male) [69]. In a report by the National Health and Nutrition Examination Survey (NHANES), the prevalence of anti-TPO and anti-Tg levels above the normal range was significantly higher in women than in men (42.1% vs. 36.1% and 9.6% vs. 5.8%, respectively) [70]. This gender gap could be explained by a number of factors like the differences between male and female immune systems that make women more prone to react to certain antigens, which may in turn be sustained by hormonal differences. Therefore, gender bias in autoimmunity may also be explained by differences in reproductive function (puberty, pregnancy and menopause) [70]. Estrogens affect immunity by modulating the development and function of lymphocytes and promoting cytoprotection. During the onset of HD, an adequate interaction among thyroid, antigen-presenting and T cells is necessary [71]. As a result, specific cytokines produced by the innate immune system are able to influence the differentiation of bipotential T helper (Th) cells preferentially towards Th1 (cellular immunity) and not Th2 (humoral immunity) cells [69]. At this stage though, estrogens are capable of suppressing the Th1-mediated responses while Th2 activity is enhanced [70]. Hence, estrogens may have a protective role in Th1-dependant diseases like HD, yet they would tend to worsen antibody-mediated pathologies [70]. During pregnancy, there is an exponential increase in estradiol, which would reduce the Th1-mediated response. An increase in regulatory T lymphocytes also takes place, which induces immune tolerance and suppression of autoimmunity [70]. Most notably, reduced anti-TPO and anti-TG levels during pregnancy have been demonstrated, reaching the lowest values during the third trimester [72]. However, during the postpartum estrogen levels drop thus leading to higher Th1-mediated disease risk. Certainly, HD is more frequently diagnosed at this stage [73-74].

Prolactin is known for its proinflammatory effects and it also fluctuates during pregnancy. Although an exacerbation of the disease would be expected, studies have found no correlation between hyperprolactinemia and HD [75]. Other sex hormones, namely progesterone and androgens, exert anti-inflammatory and immunosuppressive effects, respectively [70]. Consequently, they would have a protective role against autoimmunity. Nevertheless, a positive effect of androgens on Th2 responses has also been described [76]. Other factors may influence autoimmune disease susceptibility such as paternal/maternal inheritance (including mitochondrial inheritance, genomic imprinting and sex chromosome inactivation), genetic predisposition and environmental stimuli through epigenetic mechanisms. The role of microchimerism, i.e. the trafficking of cells between the mother and the fetus, in HD needs to be further explored [70].

Stress: Chronic biopsychosocial stress negatively shapes health over the life span by contributing to chronic inflammation processes and infection risk [77]. However, the exact mechanisms by which stress would interact with autoimmune disease pathophysiology have not been fully elucidated. Some studies suggest that, in cases of high stress-mediated hormonal activity, T-cell balance may be skewed toward Th2 (thus increasing Graves-Basedow’s disease risk), whereas Th1 function may be deficient. In other words, immunitymay be mainly humoral [78-79]. On the other hand, other studies consider that, upon stress relieve, the decrease of the immunosuppressive effects may induce a rebound autoimmune reaction that could affect the thyroid gland. In any case, it seems that stress would not be directly related to HD [80-82]. With regard to thyroid function, cortisol rise observed in chronic stress may inhibit the production of thyrotropin-releasing hormone (TRH), which may lead to central hypothyroidism. 

Moreover, the peripheral conversion of T4 to T3 would also be reduced and shifted toward the production of biologically inactive reverse Triiodothyronine (T3), causing peripheral functional hypothyroidism [83-84].

Conclusion

We reckon that autoimmune thyroiditis disease course would be better handled by including the examination of etiopathogenic factors into its clinical assessment and patient management. We have shown that the extensive list of considerations may include genetic susceptibility, gastro-intestinal conditions (intestinal permeability defects, dysbiosis and SIBO), other pathologies like celiac disease/gluten intolerance, and specific micronutrients imbalance (vitamin D, iodine and selenium). Biopsychosocial stress, hormonal imbalances and gender differences and exposure to environmental toxic substances (heavy metals and others) and infectious agents (both viral and bacterial) also contribute to HD development and chronification. Finally, given the implication of multiple mechanisms of etiology and pathogenesis, a multidisciplinary strategy may be required to deliver integrated and successful patient care. Further studies are still needed in order to confirm the number of associations between these factors and the development and progression of HD. The study design should take into account the multiple and diverse factors as well as the multisystemic nature of the disease.

Acknowledgement

We would like to thank Neus Cantariño for her valuable help in editing this manuscript.

References

  1. Liontiris MI, Mazokopakis EE. A concise review of Hashimoto thyroiditis (HT) and the importance of iodine, selenium, vitamin D and gluten on the autoimmunity and dietary management of HT patients. Points that need more investigation. Hell J Nucl Med. 2017;20(1):51-56.
  2. Baretić M.100 years of Hashimoto thyroiditis, still an intriguing disease. Acta Med Croatica. 2011;  65(5):453-7.
  3. McGrogan A, Seaman HE, Wright JW, de Vries CS. The incidence of autoimmune thyroid disease: a systematic review of the literature. Clin Endocrinol (Oxf).2008;69(5):687-96.
  4. Lerner A, Jeremias P, Matthias T. The world incidence and prevalence of autoimmune diseases is increasing. Int J Celiac Dis.2015; 3(4):151-155.
  5. Jonklaas J, Bianco AC, Bauer AJ, Auer AJ, Burman KD, Cappola AR et al. Guidelines for the treatment of hypothyroidism: Prepared by the american thyroid association task force on thyroid hormone replacement. Thyroid. 2014; 24(12):1670-1751.
  6. Wekking EM, Appelhof BC, Fliers E, Schene AH,  Huyser J,  Tijssen JGP, et al. Cognitive functioning and well-being in euthyroid patients on thyroxine replacement therapy for primary hypothyroidism. Eur J Endocrinol. 2005; 153(6):747-753.
  7. Saravanan P, Chau WF, Roberts N, Vedhara K, Greenwood R, Dayan CM. Psychological well-being in patients on ‘adequate’ doses of l-thyroxine:Results of a large, controlled community-based questionnaire study. Clin Endocrinol. 2002; 57(5):577-585.
  8. Gullo D, Latina A, Frasca F, Le Moli R, Pellegriti G,Vigneri R. Levothyroxine monotherapy cannot guarantee euthyroidism in all athyreotic patients. PLoS One 6(8): e22552
  9. Jonklaas J, Davidson B, Bhagat S, Soldin SJ. Triiodothyronine levels in athyreotic individuals during levothyroxine therapy. JAMA. 2008; 299(7):769-777.
  10.  Holtorf K. Peripheral thyroid hormone conversion and its impact on TSH and metabolic activity. J  Restor Med. 2014; 3:30.
  11.  ​Campbell AW. Autoimmunity and the Gut. Autoimmune Dis. 2014;152428.
  12. Holtorf K. Peripheral thyroid hormone conversion and its impact on TSH and metabolic activity. J  Restor Med. 2014; 3:30.
  13.  Mu Q, Kirby J, Reilly CM, Luo XM. Leaky gut as a danger signal for autoimmune diseases. Front Immunol.2017;8:598.
  14. Arrieta MC, Bistritz L, Meddings JB. Alterations in intestinal permeability. Gut. 2006; 55(10):1512-1520.
  15. Zaletel K, Gaberšček S. Hashimoto’s thyroiditis: From genes to the disease. Curr Genomics. 2011;12(8):576-588
  16. Mu Q, Kirby J, Reilly CM, Luo XM. Leaky gut as a danger signal for autoimmune diseases. Front Immunol.2017;8:598.
  17. Bischoff SC, Barbara G, Buurman W, Ockhuizen T,  Schulzke JD, Serino M, et al. Intestinal permeability a new target for disease prevention and therapy. BMC Gastroenterol.2014; 14:189.
  18.  Malik A, Morya RK, Bhadada SK, Rana S. Type 1 diabetes mellitus: Complex interplay of oxidative stress, cytokines, gastrointestinal motility and small intestinal bacterial overgrowth. Eur J Clin Invest. 2018; 48(11):e13021.
  19. Virili C, Centanni M. Does microbiota composition affect thyroid homeostasis? Endocrine. 2015; 9:583-585
  20.  Resnick C. Nutritional protocol for the treatment of intestinal permeability defects and related conditions. J Nat Med. 2010; 2(3):14-23.
  21. Konrad P, Chojnacki J, Kaczka A, Pawłowicz M, Rudnicki C, Chojnacki C. Thyroid dysfunction in patients with small intestinal bacterial overgrowth. Pol Merkur Lekarski. 2018; 44(259):15-18.
  22. Rao SSC, Bhagatwala J. Small intestinal bacterial overgrowth: Clinical features and therapeutic management. Clin Transl Gastroenterol. 2019; 10(10):e00078
  23. Zhang Y, Liu G, Duan Y, Han X, Dong H, Geng J. Prevalence of small intestinal bacterial overgrowth in multiple sclerosis: A case-control study from China. J Neuroimmunol. 2018; 301:83-87.
  24.  Benvenga S, Guarneri S. Molecular mimicry and autoimmune thyroid disease. Rev Endocr Metab Disord. 2016; 17(4):485-498.
  25. Malik A, Morya RK, Bhadada SK, Rana S. Type 1 diabetes mellitus: Complex interplay of oxidative stress, cytokines, gastrointestinal motility and small intestinal bacterial overgrowth. Eur J Clin Invest. 2018; 48(11):e13021.
  26.  Janegova A, Janega P, Rychly B, Kuracinova K, Babal P. The role of Epstein-Barr virus infection in the development of autoimmune thyroid diseases. Endocrynol Pol. 2015; 66(2):132-136.
  27. Henriksson AE, Blomquist L, Nord CE, Midtvedt T, Uribe A. Small intestinal bacterial overgrowth in patients with rheumatoid arthritis. Ann Rheum Dis. 1993; 52(7):503-510.
  28. Lauritano EC, Bilotta AL, Gabrielli M, Scarpellini E, Lupascu A, Laginestra A, et al. Association between hypothyroidism and small intestinal bacterial overgrowth. J Clin Endocrinol Metab.2007; 92(11):4180-4184.
  29. Brechmann T, Sperlbaum A, Schmiegel W. Levothyroxine therapy and impaired clearance are the strongest contributors to small intestinal bacterial overgrowth: Results of a retrospective cohort study. World J Gastroenterol. 23(5):842–852.
  30. Kannangai R, Sachithanandham J, Kandathil AJ, Ebenezer DL, Danda D, Vasuki Z, et al. Immune responses to Epstein-Barr virus in individuals with systemic and organ specific autoimmune disorders. Indian J Med Microbiol. 2010; 28(2):120-123.
  31. Janegova A, Janega P, Rychly B, Kuracinova K, Babal P. The role of Epstein-Barr virus infection in the development of autoimmune thyroid diseases. Endocrynol Pol. 2015; 66(2):132-136.
  32.  ​Prelog M, Schönlaub J, Würzner R, Koppelstaetter C, Almanzar G, Brunner A, et al. Lower CD28+ T cell proportions were associated with CMV-seropositivity in patients with Hashimoto’s thyroiditis. BMC Endocr Disord. 2013; 13:34.
  33.  Morohoshi K, Takahashi Y, Mori K. Viral infection and innate pattern recognition receptors in induction of Hashimoto’s thyroiditis. Discov Med. 2011; 12(67):505-511.
  34.  Ferrari SM, Fallahi P, Antonelli A, Benvenga S. Environmental issues in thyroid diseases. Front Endocrinol. 2017; 8:50.
  35. ​ Fallahi P, Ferrari SM, Vita R, Benvenga S, Antonelli A.The role of human parvovirus B19 and hepatitis C virus in the development of thyroid disorders. Rev Endocr Metab Disord.2016; 17(4):529-535.
  36. Dittfeld A, Gwizdek K, Michalski M, Wojnicz R. A possible link between the Epstein-Barr virus infection and autoimmune thyroid disorders. Cent Eur J Immunol. 2016; 41(3):297-301
  37. Pender MP. D8+ T-Cell deficiency, Epstein-Barr virus infection, vitamin d deficiency, and steps to autoimmunity: A unifying hypothesis. Autoimmune Dis. 2012:189096.
  38.  Patrick L. Thyroid disruption: Mechanism and clinical implications in human health. Altern Med Rev.2009; 14(4):326-346.
  39.  Chailurkit LO, Aekplakorn W, Ongphiphadhanakul B. The association of serum bisphenol A with thyroid autoimmunity. Int J Environ Res Public Health. 2016; 13(11):1153.
  40. ​Park C, Choi W, Hwang M, Lee Y, Kim S, Yu S, et al. Associations between urinary phthalate metabolites and bisphenol A levels, and serum thyroid hormones among the Korean adult population-Korean national environmental health survey (KoNEHS) 2012-2014. Sci Total Environ. 2017; 584-585:950-957.
  41.  Duan J, Kang J, Deng T, Yang X, Chen M. Exposure to DBP and high iodine aggravates autoimmune thyroid disease through increasing the levels of IL-17 and thyroid binding globulin in Wistar rats. Toxicol Sci. 2018; 163(1):196-205.
  42. Tozzoli R, Barzilai O, Ram M, Villaltac D, Bizzarod N, Shererb Y, et al. Infections and autoimmune thyroid diseases: parallel detection of antibodies against pathogens with proteomic technology. Autoimmun Rev. 2008; 8(2):112-115.
  43. Crowe W, Allsopp PJ, Watson GE, et al. Mercury as an environmental stimulus in the development of autoimmunity-A systematic review. Autoimmun Rev. 2017; 16(1):72-80.
  44.  Hu Q, Han X, Dong G, Yan W, Wang X, Bigambo FM, et al. Association between mercury exposure and thyroid hormones levels: A meta-analysis. Environ Res. 2021; 196:110928.
  45.  Chen A, Kim SS, Chung E, Dietrich KN. Thyroid hormones in relation to lead, mercury, and cadmium exposure in the National Health and Nutrition Examination Survey, 2007-2008. Environ Health Perspect.2013; 121(2):181-186.
  46. Gallagher CM, Meliker JR. Mercury and thyroid autoantibodies in U.S. women, NHANES 2007-2008. Environ Int. 2012;40:39-43.
  47.  Raymond LJ, Ralston NVC. Mercury: Selenium interactions and health implications. Seychelles Med Dent J. 2004; 7(1):72-77.
  48.  Queiroz ML, Pena SC, Saltes TS, Saad STO,  Capitani EMD. Abnormal antioxidant system in erythrocytes of mercury-exposed workers. Human Exp Toxicol. 1998; 17:225-230.
  49.  Rachmawati D, Muris J, Scheper RJ, et al. Continuing the quest for autoimmunity due to oral metal exposure. Autoimmunity. 2015; 48(7):494-501.
  50. ​Gundacker C, Komarnicki G, Jagiello P, Gencikova A, Dahmend N, Wittmann KJ, et al. Glutathione-S-transferase polymorphism, metallothionein expression and mercury levels among students in Austria. Sci Total Environ. 2007; 385(1-3):37-47.
  51.  ​Lee YM, Hong YC, Ha M, Kim Y, Park H, Kim HS, et al. Prenatal Bisphenol-A exposure affects fetal length growth by maternal glutathione transferase polymorphisms and neonatal exposure affects child volume growth by sex: From multiregional prospective birth cohort MOCEH study. Sci Total Environ. 2018; 612:1433-1441.
  52.  ​Wang J, Lv S, Chen G, Gao C, He J, Zhong H, et al. Meta-analysis of the association between vitamin D and autoimmune thyroid disease. Nutrients. 2015; 7(4):2485-2498.
  53. ​Kamen DL, Tangpricha V. Vitamin D and molecular actions on the immune system: Modulation of innate and autoimmunity. J Mol Med. 2010; 88(5): 441-450.
  54. Thomas D, Karachaliou F, Kallergi K, Vlachopapadopoulou E, Antonaki G, Chatzimarkou F, et al. Herpes virus antibodies seroprevalence in children with autoimmune thyroid disease. Endocrine. 2008; 33(2):171-175
  55.  de Punder K, Pruimboom L. The dietary intake of wheat and other cereal grains and their role in inflammation. Nutrients. 2013; 5(3):771-787.
  56.  Sharma BR, Joshi AS, Varthakavi PK, Chadha MD, Bhagwat NM, Pawal PS. Celiac autoimmunity in autoimmune thyroid disease is highly prevalent with a questionable impact. Indian J Endocrinol Metab.2016; 20 (1):97-100.
  57.  ​Czaja-Bulsa G. Non coeliac gluten sensitivity-A new disease with gluten intolerance. Clin Nutr 2015; 34(2):189-194.
  58. ​ Carroccio A, D’Alcamo A, Cavataio F, Soresi M, Seidita A,  Sciumè C, et al. High proportions of people with nonceliac wheat sensitivity have autoimmune disease or antinuclear antibodies. Gastroenterol. 2015; 149:596-603.
  59. ​ Papanastasiou L, Vatalas I-A, Koutras DA, Mastorakos G. Thyroid autoimmunity in the current iodine environment. Thyroid. 2007; 17:729-739.
  60.  ​Burek CL. Talor MV. Environmental triggers of autoimmune thyroiditis. J Autoimmun. 2009; 33:183-189
  61.  Luo Y, Kawashima A, Ishido Y, Yoshihara A, Oda K, Hiroi N, et al. Iodine excess as an environmental risk factor for autoimmune thyroid disease. Int J Mol Sci. 2014; 15(7):12895–12912.
  62.  Laurberg P, Jørgensen T, Perrild H, Ovesen L, Knudsen N, Bülow I, et al. The Danish investigation on iodine intake and thyroid disease, DanThyr: Status and perspectives. Eur J Endocrinol. 2006; 155(2):219-228.
  63. ​ Golbidi S, Laher I. Antioxidant therapy in human endocrine disorders. Med Sci Monit. 2010; 16(1):RA9-24.
  64. Xu J, Liu XL, Yang XF, Guo HL, Zhao LN, Sun XF. Supplemental selenium alleviates the toxic effects of excessive iodine on thyroid. Biol Trace Elem Res. 2011; 141(1-3):110-118.
  65.  ​Xu J, Yang XF, Guo HL, Hou XH, Liu LG, Sun XF. Selenium supplement alleviated the toxic effects of excessive iodine in mice. Biol Trace Elem Res. 2006; 111(1-3):229-238.
  66. Barzilai O, Sherer Y, Ram M, Izhaky D, Anaya JM, Shoenfeld Y. Epstein-Barr virus and cytomegalovirus in autoimmune diseases: Are they truly notorious? A preliminary report. Ann N Y Acad Sci. 2007; 1108:567-577.
  67. Sakaguchi S, Ono M, Setoguchi R, Yagi H, Hori S, Fehervari Z, et al. Foxp3+CD25+CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol Rev. 2006; 212:8-27.
  68.  Ngo ST, Steyn FJ, McCombe PA. Gender differences in autoimmune disease. Front Neuroendocrinol. 2014; 35(3):347-369.
  69. Jain RB. Thyroid profile of the reference united states population: Data from NHANES 2007-2012. Int Arch Endocrinol Clin Res. 2015; 1:004
  70. Ajjan RA, Watson, PF, Weetman AP. Cytokines and thyroid function. Adv Neuroimmunol. 1996; 6(4):359-386.
  71.  ​Smyth PP, Wijeyaratne CN, Kaluarachi WN, Smith DF, Premawardhana LDKE, Parkes AB, et al. Sequential studies on thyroid antibodies during pregnancy. Thyroid. 2005; 15(5):474-477.
  72. Wiersinga W. Clinical relevance of environmental factors in then pathogenesis of autoimmune thyroid disease. Endocrinol Metab. 2016; 31(2):213-222.
  73. Weetman AP. Immunity, thyroid function and pregnancy: Molecular mechanisms. Nat Rev Endocrinol. 2010; 6(6):311-318.
  74.  Vanderpump MP, French JM, Appleton D, Tunbridge WM, Kendall-Taylor P. The prevalence of hyperprolactinemia and association with markers of autoimmune thyroid disease in survivors of the Whickham Survey cohort. Clin Endocrinol (Oxf). 1998; 48(1):39-44.
  75.  González DA, Díaz BB, Rodríguez Pérez MC, Hernández AG, Chico BN, de León AC. Sex hormones and autoimmunity. Immunol Lett. 2010; 133(1):6-13.
  76. Stojanovich L. Stress and autoimmunity. Autoimmun Rev. 2010; 9(5):A271-6.
  77.  ​Leclère J, Weryha G. Stress and auto-immune endocrine diseases. Horm Res. 1989; 31:90-3.
  78.  Elenkov IJ, Chrousos GP. Stress hormones, proinflammatory and antiinflammatory cytokines, and autoimmunity. Ann N Y Acad Sci. 2002; 966:290-303.
  79. Martin-du Pan RC. Triggering role of emotional stress and childbirth. Unexpected occurrence of Graves’ disease compared to 96 cases of Hashimoto thyroiditis and 97 cases of thyroid nodules. Ann Endocrinol. 1998; 59:107-112.
  80. ​ Oretti RG, Harris B, Lazarus JH, Parkes AB, Crownshaw T. Is there an association between life events, postnatal depression and thyroid dysfunction in thyroid antibody positive women? Int J Soc Psychiatry.2003; 49(1):70-76.
  81.  ​Effraimidis G, Tijssen JG, Brosschot JF, Wiersinga WM. Involvement of stress in the pathogenesis of autoimmune thyroid disease: A prospective study. Psychoneuroendocrinology. 2012; 37(8):1191-1198.
  82. Tsatsoulis A. The role of stress in the clinical expression of thyroid autoimmunity. Ann N Y Acad Sci 2006; 1088:382-395.
  83. Bános C, Takó J, Salamon F, Györgyi S, Czikkely R. Effect of ACTH-stimulated glucocorticoid hypersecretion on the serum concentrations of thyroxine-binding globulin, thyroxine, triiodothyronine, reverse triiodothyronine and on the TSH-response to TRH. Acta Med Acad Sci Hung. 1979; 36(4):381-394.

Author Info

Isabel García1* and Sergio Abanades2
 
1Department of Endocrinology and Metabolism, Institute for Integrative and Conscious Health (ISIC), París, 3r 1a Barcelona, Spain
2Department of Pharmacology, International College of Human Nutrition and Functional Medicine, París, 3r 1a Barcelona, Spain
 

Citation: García I, Abanades S (2021) Etiopathogenetic Mechanisms in Hashimoto’s Autoimmune Thyroid Disease. Thyroid Disorders Ther 10:262.

Received: 15-Nov-2021 Accepted: 29-Nov-2021 Published: 06-Dec-2021

Copyright: © 2021 García I, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top