Skip to main content

Immunogenetics of Hashimoto's thyroiditis

Abstract

Hashimoto's thyroiditis (HT) is an organ-specific T-cell mediated disease. It is a complex disease, with a strong genetic component. To date, significant progress has been made towards the identification and functional characterization of HT susceptibility genes. In this review, we will summarize the recent advances in our understanding of the genetic input to the pathogenesis of HT.

Introduction

Hashimoto's thyroiditis (HT) is one of the most common human autoimmune diseases responsible for considerable morbidity in women [1]. It is an organ-specific T-cell mediated disease that affects the thyroid, and genetics play a contributory role in its complexity. To date, significant progress has been made in identifying and characterizing those genes involved in the disease. In this review, we will summarize recent advances in our understanding of the genetic contribution to the pathogenesis of HT.

Epidemiology and clinical features of Hashimoto's thyroiditis

Goitrous autoimmune thyroiditis, or Hashimoto's thyroiditis is a common form of chronic autoimmune thyroid disease (AITD). The disorder affects up to 2% of the general population [2] and is more common in older women and ten times more frequent in women than in men [3]. In the NHALES III study, performed in the USA, the prevalence of subclinical and clinical hypothyroidism was 4.6% and 0.3% respectively [4]. Another US epidemiological study, the Whickham survey, showed the prevalence of spontaneous hypothyroidism to be 1.5% in females and less than 0.1% in males [5]. These prevalence rates are similar to those reported in Japan [6] and Finland [7]. A significant proportion of patients have asymptomatic chronic autoimmune thyroiditis and 8% of woman (10% of woman over 55 years of age) and 3% of men have subclinical hypothyroidism [8]. According the data of the 20-year follow-up to the Whickham survey cohort, the risk of developing overt hypothyroidism is four times higher in women aged between 60 and 70 years than for women between 40 and 50 years of age [1].

Subclinical hypothyroidism is characterized by an increase in serum thyrotropin (TSH) whilst serum levels of thyroxine (T4) and triiodothyronine (T3) remain normal. The overt disease is defined by the dramatic loss of thyroid follicular cells (thyrocytes), hypothyroidism, goitre, circulating autoantibodies to two primary thyroid-specific antigens, thyroglobulin (Tg), thyroid peroxidase (TPO), and lowered concentrations of serum TSH and T4 [9]. Histological and cytological features of HT include a dense thyroidal accumulation of lymphocytes, plasma cells and occasional multinuclear giant cells. The epithelial cells are enlarged, with a distinctive eosinophilic cytoplasm, owing to increased number of mitochondria [10].

HT has been shown to often coexist with other autoimmune diseases such as type 1 diabetes (T1D), celiac disease, rheumatoid arthritis, multiple sclerosis, vitiligo, etc [11–14]. HT can also be expressed as part of an autoimmune polyendocrine syndrome type 2 (APS-2), which is usually defined by the occurrence of two or more of the following: Addison's disease (always present), AITD and/or type 1 diabetes [15], in the same patient.

In common with probably all autoimmune disorders, the harmful interaction between internal (genetic) and external (environmental and endogenous) factors is required to initiate Hashimoto's disease (Fig. 1). Environmental triggers of HT include iodine intake [16, 17], bacterial and viral infections [18, 19], cytokine therapy [20] and probably pregnancy [21, 22]. The role of dietary iodine is well defined in epidemiological studies [23, 24] and in animal models [25–27] and seems to be the most significant environmental factor to induce thyroiditis.

Figure 1
figure 1

Possible pathogenic mechanism of Hashimoto's thyroiditis. Genetically predisposed individuals could be influenced by an environmental trigger (i.e., dietary iodine, infection, pregnancy, cytokine therapy) that induces an autoimmune response against thyroid-specific antigens by infiltrating immune cells. The autoimmune process results in preferential T helper type 1 (TH1)-mediated immune response and induction of apoptosis of thyroid cells that leads to hypothyroidism.

Pathogenesis of Hashimoto's thyroiditis

Autoimmunity in Hashimoto's thyroiditis

The development of the autoimmune failure of the thyroid is a multistep process, requiring several genetic and environmental abnormalities to converge before full-blown disease develops (Fig. 2). At the onset of disease, major histocompatibility complex (MHC) class II-positive antigen-presenting cells (APC), particularly dendritic cells, and different subclasses of macrophages, accumulate in the thyroid [28, 29]. APC present thyroid-specific autoantigens to the naïve T cells, leading to activation and clonal expansion of the latter. Thus, the initial stage of the disease is followed by a clonal expansion phase and maturation of autoreactive T and B lymphocytes in the draining lymph nodes.

Figure 2
figure 2

A scheme of autoimmune events in Hashimoto's thyroiditis. In an initial stage, antigen-presenting cells (APC), mostly dendritic cell and macrophage (Mφ) derived, infiltrate the thyroid gland. The infiltration can be induced by an envinromental triggering factor (dietary iodine, toxins, virus infection, etc.) which causes insult of thyrocytes and releasing of thyroid-specific proteins. These proteins serve as a source of self-antigenic peptides that are presented on the cell surface of APC after processing. Taking up relevant autoantigens, APC travel from the thyroid to the draining lymph node. A central phase occurs in the draining lymph node in which interactions between APC, autoreactive (AR) T cells (that survive as result of dysregulation or breakage of immune tolerance) and B cells result in inducing production of thyroid autoantibodies. In the next step, antigen-producing B lymphocytes, cytotoxic T cells and macrophages infiltrate and accumulate in the thyroid through expansion of lymphocyte clones and propagation of lymphoid tissue within the thyroid gland. This process is preferentially mediated by T helper type 1 (TH1) cells which secrete regulatory cytokines (interleukin-12, interferon-γ and tumor necrosis factor-α). In a final stage, the generated autoreactive T cells, B cells and antibodies cause massive depletion of thyrocytes via antibody-dependent, cytokine-mediated and apoptotic mechanisms of cytotoxity that leads to hypothyroidism and Hashimoto's disease.

In autoimmune thyroditis animal models, genetically determined immune defects have been suggestively linked to the breakdown of immunological self-tolerance that results in the presentation of host autoantigens and expansion of autoreactive lymphocyte clones. These immune defects are associated with the presence of particular MHC class II haplotypes, but other immune and immune regulatory genes (i.e., CTLA-4 and others) are also involved [30–32].

Breakdown of the immune tolerance might occur in several ways including interrupting central tolerance (e.g. deletion of autoreactive T cells in the thymus), defects in maintaining peripheral tolerance (e.g. activation-induced T-cell death and suppressing activity of regulatory T lymphocytes) and anergy (e.g. the expression of MHC class II molecules on non-professional APC). Animal models genetically predisposed to develop an autoimmune disease, and patients with AITD, showed a lack of, or a deficiency in, a subpopulation of regulatory T cells with suppressive function [33–35].

The mechanisms, whereby autoreactive T cells escape deletion and anergy, and become activated, remain uncertain. There is evidence that the thyroid cell itself, by "aberrantly" expressing MHC molecules, can play the role of "non-professional " APS and present disease-initiating antigen directly to the T cells [36, 37]. The concept of aberrant MHC class II expression was supported by studies in mice. They developed a type of Graves' Disease (GD) after being injected with fibroblasts coexpressing MHC class II and the TSH receptor (TSHR). TPO antibody production was induced after injection with fibroblasts coexpressing class II molecules and TPO [38, 39].

Iodine is a necessary component of normal thyroid hormonogenesis. Incorporation of iodine into thyrosine residues of Tg leads to the formation of mono-iodotyrosine and di-idothyrosine derivates that subsequently undergo an oxidative coupling event resulting in the producing of T3 and T4. Iodine can promote antithyroid immunity in a number of ways. Several studies suggest that iodination of Tg is crucial for recognition by Tg-reactive T cells [40, 41]. Iodine excess can affect the Tg molecule directly, creating new epitopes or exposing "cryptic" epitopes. It has been demonstrated that a highly iodinated thyroglobulin molecule is a better immunogen than Tg of low iodine content [41, 42]. Therefore, highly iodinated Tg may facilitate antigen uptake and processing by APC. Additionally, high doses of iodine were shown to directly affect macrophages, dendritic cells, B and T lymphocytes, resulting in stimulation of macrophage myeloperoxidase activity, acceleration of the maturation of dendritic cells, increasing the number of circulating T cells and stimulating B cell immunoglobulin production [25]. Excessive amounts of iodide ion are rapidly oxidized by TPO, thereby generating excessive amounts of reactive intermediates such as hypoiodous acid and oxygen radicals. These oxidative species damage thyrocyte cell membrane by oxidation of membrane lipids and proteins causing thyrocyte necrosis [43]. The state of severe iodine deficiency itself namely leads to a lowering of thyroid autoimmunity and an immunodeficient state in autoimmune-prone BB-DP rats. This hampers the autoreactcive T-cell generation and autoantibody production [25]. A lower degree of Tg iodination also makes this molecule less antigenic [42].

An influx of dendritic cells and macrophages to the thyroid may occur as a consequence of inflammatory events in the gland. Early non-specific necrosis of thyrocytes due to toxins (i.e. iodine, etc.), and perhaps viral or bacterial infection, can attract these cells to the thyroid. Moreover, these immune cells are normal constituents of the thyroid that are able to regulate the growth and function of thyrocytes via interleukin-1 (IL-1) and IL-6-mediated pathways [44].

A central phase of HT is characterized by the recognition of presented autoantigens by the lymphocytes, followed by an apparent uncontrolled production of autoreactive CD4+ T cells, CD8+ cytotoxic T cells and immunoglobulin G (IgG) autoantibodies. Initially, the production of self-reactive cells and autoantibodies occurs in the draining lymph nodes (Fig. 2). Later, the lymphoid tissue often develops directly in the thyroid gland itself. This tissue is generally very well organized, with cords of anti-Tg-antibody-producing plasma cells in the periphery. It is usually non-destructive and shows a peaceful co-existence with adjacent thyrocytes.

Thyroglobulin, the main protein synthesized in the thyroid, serves both in the synthesis and in the storage of thyroid hormones. Human Tg molecules contain at least four thyroid hormone synthesis sites from the iodinated tyrosine residues at positions 5, 2553, 2567 and 2746 [45]. The hormone synthesis sites and the iodine content of Tg play an important role in its autoantigenicity [40]. Tg is one of the major autoantigens in thyroid autoimmunity and serologic studies have shown that there are at least 40 antigenic epitopes on human Tg [16, 46]. Tg-antibodies are detected in almost all patients with AITD [47]. Anti-thyroglobulin antibodies were also reported in up to 27% of normal individuals [48]. However, numerous studies have clearly shown that the epitope recognition pattern of the natural anti-Tg antibodies is differented from that of AITD-associated anti-Tg antibodies. Most studies have demonstrarted a restricted epitope recognition pattern of AITD subjects by anti-Tg antibodies, in contrast to polyclonal reactivity observed with anti-Tg antibodies from healthy individuals [49, 50]. Human or mouse Tg immunization induces experimental autoimmune thyroiditis (EAT) in mice [51]. The EAT induction is HLA-dependent implying an interaction between the Tg molecule and the MHC glycoproteins [52]. In addition, alterations to Tg could explain interactions between genetic and environmental factors in the aetiology of HT.

Thyroid peroxidase is another significant autoantigen in the thyroid of patients affected with HT and AITD. This enzyme catalyses the oxidation of iodine to an iodinating species that forms iodotyrosines in a Tg molecule and subsequently iodotyronines [53]. TPO antibodies are heterogeneous. To date, around 180 human TPO anribodies have been cloned and sequenced. This allows for the possible identification of major features of the TPO-directed antibodies repertoire during AITD. In Graves' disease patients, heavy chain VH domains of anti-TPO antibodies preferentially use D proximal IGHV1 genes. IGHV3 genes, mainly located in the middle of the immunoglobulin heavy chain gene (IGH) cluster on chromosome 15q11, characterize HT patients more frequently. A large proportion of the anti-TPO heavy chain VH domain comes about following a VDJ recombination process that uses inverted D genes [54, 55].

Autoantibodies against other thyroid-specific antigens such as thyrotropin receptor and sodium iodide symporter were also found in serum of HT patients. However, these antibodies occur at low frequency and do not appear to contribute any diagnostic power for HT [56, 57].

In a final, destructive step of Hashimoto's thyroiditis, the autoreactive T cells diffusely accumulate in large numbers and infiltrate thyroid parenchyma (Fig. 2). In the BB-DP rat model, T-helper type 1 (TH1)-mediated mechanisms involving production of IL-12, tumor necrosis factor-α (TNF-α) and interferon-γ play a major role in the destruction of thyrocytes, rather than TH2 type mechanisms directed by IL-4 and IL-10 [58]. The infiltration of activated scavenger macrophages into the thyroid follicles, thus destroying the thyroid cells, is compatible with TH1-mediated mechanisms [59]. Fas and Fas ligand (FasL) expression was higher in rats with lympholytic thyroiditis indicating a role of these apoptotic molecules in thyrocyte death [60].

Apoptosis in Hashimoto's thyroiditis

Autoimmune responses against specific antigens are primary determinants in thyroid autoimmunity. Other molecular mechanisms including cell apoptosis may play a role in determining the opposite phenotypic outcomes of AITD such as thyroid destruction in HT and thyroid hyperplasia in GD. T-helper lymphocytes produce cytokines that influence both immune and target cells at several levels. The predominance of TH1 or TH2 cytokines might regulate thyrocyte survival through the induction of pro-apoptotic and anti-apoptotic proteins. TH1-mediated mechanisms lead to thyrocyte depletion in Hashimoto's thyroiditis through the involvement of death receptors and cytokine-regulated apoptotic pathways [61, 62].

The normal thyroid gland has been shown to act as an immune privileged site having carefully regulated mechanisms of cell death and self-protection against attack by infiltrating activated T-cells induced by apoptosis [63, 64]. Cell apoptosis occurs in the normal thyroid at a low level. As new thyrocytes are produced, old cells are destroyed in order to maintain normal thyroid volume and function. Deregulation of apoptosis, which is weakly determined by genetic susceptibility, can lead to destructive processes. Initiation of an out-of-control apoptotic mechanism in thyroid cells may be caused by various non-genetic injuries that affect expression of apoptosis inhibitor molecule Bcl-2 or membrane ligand FasL [65]. Thyrocytes from HT thyroid glands are able to hyperproduce Fas and FasL on their surfaces thus inducing fratricide apoptosis [66]. IL-1β, abundantly produced in HT glands, induces Fas expression in normal thyrocytes, the cross-linking of Fas resulting in massive thyrocyte apoptosis. This can play a role in the progression of Hashimoto's thyroiditis [67].

Immune-mediated apoptosis of thyrocytes is directed by CD8+ cells. Receptors on the target cell are triggered by lymphocyte ligands and/or released soluble factors are delivered to the target cell [68]. Receptors involved in immune-mediated apoptosis include the TNF R1 receptor, the Fas receptor and death receptors DR3 and DR4, whereas soluble mediators include substances such as perforines and TNF [68–70].

The common apoptotic pathway consists of subsequent activation of specific intracellular proteases known as caspases. These caspases are themselves activated by specific proteolytic cleavage or may be activated by cleavage performed by other caspases. The caspase cascade ultimately induces enzymes that progressively destroy the cell and its genetic material, finally lead to cell death. The apoptosis, or programmed cell death, can be initiated by binding death ligands, such as TNF, TNF-related apoptosis-induced ligand (TRAIL) and FasL, to the cell surface. This in turn starts intracellular signal cascading of caspases [71].

Several apoptosis signalling pathways, initiated by molecules such as FasL and TRAIL, have been shown to be active in thyrocytes and may be involved in destructive thyroiditis [72]. Fas-mediated apoptosis seems to be a general mechanism of cell destruction in AITD. In GD patients, reduced levels of Fas/FasL and increased levels of antiapoptotic molecule Bcl-2 favour thyroid cell survival and apoptosis of infiltrating lymphocytes. In contrast, the regulation of Fas/FasL/Bcl-2 expression in HT can promote thyrocyte apoptosis through homophylic Fas-FasL interactions and a gradual reduction in thyrocyte numbers leading to hypothyroidism [61].

Thus, the rate of thyrocyte apoptosis dictates the clinical outcome of thyroid autoimmunity. Though rare in normal thyroid, it markedly increases during HT, but not in GD. Therefore, regulation of thyrocyte survival is a crucial pathogenic determinant.

Genetics of Hashimoto's thyroiditis

Evidence for genetic susceptibility to Hashimoto's thyroiditis

Abundant epidemiologic data (population-based and family-based studies, twin studies) suggest a strong genetic contribution to the development of HT. The disease clusters in families [22, 73]. Thyroid abnormalities with clinical outcomes were observed in 33% of offspring of patients with HT or GD [73]. The sibling risk ratio (λS), that is the ratio of the prevalence of disease in siblings to the prevalence in the general population, can be used as a quantitative measure of the genetic contribution to the disease. Usually, a λS of more than five indicate a significant genetic contribution to the disease development. Based on historical data, the λS for AITD is estimated to be greater than 10, supporting a strong case of genetic influence on disease development [74]. Using HT prevalence data from the NHAHES III study, an estimated λS value is about 28 for HT [74].

In Danish twin study, the concordance rates for Hashimoto's disease were 38% for monozygotic (MZ) twins and 0 for dizygotic (DZ) twins [75]. For HT, a recent twin study in California confirmed these results, showing concordance rates of 55% and 0% in MZ and DZ twins, respectively [76]. For thyroid antibodies, the concordance rate in the Danish twin study was twice high in MZ twins (80%) than that in DZ twins [75]. In a recent twin study in the UK, the concordance rates for Tg-antibodies were 59% and 23% in in MZ and DZ twins, respectively [77]. In this study, the concordance rates for TPO-autoantibodies were 47% and 29% in MZ and DZ twins, respectively [77]. These data suggest that HT and other AITD outcomes such as antibody production against thyroid-specific antigens have a substantial inherited susceptibility. HT seems to be a polygenic disease with a complex mode of inheritance. Immunomodulatory genes are expected to play an important role in predisposing and modulating the pathogenesis of Hashimoto's thyroiditis.

Animal models of autoimmune thyroiditis

Animal models of AITD still hold immense promise for the discovery of pathways, genes and environmental factors that determine the development of thyroid autoimmunity. Animals affected by experimental autoimmune thyroiditis (EAT) provide a unique opportunity to uncover disease-associated pathways, which are complicated to define in man.

One of the oldest inbred models is the obese strain chicken (OS), which develops goitrous lympholytic thyroiditis with the subsequent atrophic lympholytic thyroiditis followed by a rapid onset of hypothyroidism [78]. The biobreeding diabetes-prone (BB-DP) rat expresses a form of focal lympholytic thyroiditis that under normal conditions does not lead to hypothyroidism [79]. The nonobese diabetes (NOD) mouse strain NOD-H2h4 spontaneously develops iodine-induced autoimmune thyroiditis but not diabetes [26]. In particular, this murine strain has been extensively used to evaluate the role of iodine in the development of autoimmune thyroiditis [16].

EAT can be induced in mice by injecting with murine or human Tg, [80] and in normal syngenic recipients it is induced by the adoptive transfer of in vitro activated T cells from Tg-immunized mice [81]. The induced disease is characterized by the production of murine Tg-specific antibodies and infiltration of the thyroid by lymphocytes and other monocytes, with murine or human Tg-specific CD4+ T cells as the primary effector cells [80, 82].

Clinical features of EAT induced in the animal models mentioned above are similar to those of human HT. For example, autoimmune thyroiditis in the NOD-H2h4 mouse is induced by dietary iodine that supports epidemiologic data on human populations. In addition, the iodinified mouse represents high levels of IgG2b that is similar to HT patients expressing the predominance of IgG2 subclass, the human analog of murine IgG2b [83]. IgM class generally restricts Tg-antibodies of normal individuals and mice, while HT individuals and affected mice commonly produce Tg-antibodies of the IgG isotype [17]. However, anti-TPO antibodies generally detectable in HT patients could not be found in NOD-H2h4 mice. Despite some differences between EAT and HT, these animal models have greatly contributed to the knowledge concerning the etiology and the pathogenesis of thyroid autoimmunity, most notably on the events occurring in the very early prodromal phases.

Major Histocompatibility Complex (MHC) molecules are thought to play an important role in the initial stages of the development of HT and AITD. MHC molecules, or Human Leukocyte Antigen (HLA) homologs, play a pivotal role in T-cell repertoire selection in the thymus and in antigen presentation in the periphery. Crystal structures of MHC molecules show a peptide-binding cleft containing the variable region of these molecules. Genetic polymorphism of the MHC molecule determines the specificity and affinity of peptide binding and T-cell recognition. Therefore, polymorphisms within MHC class I and class II loci can play a significant role in predisposition to autoimmune disease [84].

A role of selected HLA class II genes susceptible to HT has been significantly clarified using transgenic NOD (H2Ag7) class II-knockout mice with EAT as a model for HT [85, 86]. In mouse genome, the H2 class II locus is homologous to the human HLA class II region [51]. A role for HLA-DRB1 polymorphism as a determining factor in HT-susceptibility, with DR3-directed predisposition and DR2-mediated resistance to the disease, was demonstrated using H2 class II-negative mice injected with HLA-DRA/DRB1*0301 (DR3) and HLA-DRB1*1502 (DR2) transgenes [85]. A role for HLA-DQ polymorphism was shown with human thyroglobulin-induced EAT in HLA-DQ*0301/DQB1*0302 (DQ8), but not HLA-DQ*0103/DQB1*0601 (DQ6), transgenic mice [52]. In summary, DR3 and DQ8 alleles are found to be susceptible, whereas DR2, DR4 and DQ6 alleles are resistant [30, 87]. Studies on EAT-developing mice showed the differential effects of class II molecules on EAT induction. Susceptibility can be determined when class II molecules from a single locus, H2A or HLA-DQ, are examined in transgenic mice, but the overall effect may depend upon the presence of both class II molecules H2A and H2E in mice and HLA-DQ and HLA-DR in humans [88]. Polymorphism within DQ alleles can determine predisposition to HT while DRB1 molecules associated with susceptibility to HT may appear to play a permissive role. The combination of susceptibility-inducing HLA-DQ and permissive DR alleles is responsible for the association of the HLA class II region with the disease.

T cells recognize an antigenic peptide via interaction of their membrane T cell receptors (TcR) with antigen-MHC complexes presented on the surface of APC. Biased or restricted TcR gene use has been reported in a variety of human or murine autoimmune diseases [89]. Biased TcR V gene in intrathyroidal T cells was also observed in mice with spontatenous (NOD strain) or human Tg-induced (CBA/J strain) thyroiditis. This confirms the primary role played by T cells in initiating EAT and the phenomenon of oligoclonal expansion of intrathyroidal T lymphocytes in early thyroiditis [90]. Sequencing of amplified TCR V beta cDNA showed that within each NOD thyroid sample at least one of the overexpressed V beta gene families was clonally expanded. For example, in the CBA/J mouse immunized with human Tg, clonally expressed T cells were shown to primarily express the murine TcR Vβ1 and Vβ13 sequences [91].

A new murine model that developed destructive thyroiditis with histological and clinical features comparable with human HT has been recently reported [92]. The transgenic mice express the TcR of the self-reactive T-cell clone derived from a patient with autoimmune thyroiditis. The T-cell clone is specific for the autoantigen thyroid peroxidase (TPO) peptide comprising amino acid residues at positions 535–551 (TPO535–551) of the TPO amino acid sequence. This includes a cryptic epitope (TPO536–547) preferentially displayed after endogenous processing during inflammation [93]. These results underline the pathogenic role of autoreactive human T cells and the potential significance of recognition of cryptic epitopes in target molecules such as TPO for inducing thyroid-specific autoimmune response.

The two-signal theory for T cell activation requires TcR engagement of its cognate antigen-MHC complex and CD28 binding to B7 ligands (B7-1 and B7-2) on APC. Activation of T cells results in increased expression of the cytotoxic T cell antigen-4 (CTLA-4) molecule that shares homology with CD28. Although B7-1 (CD80) and B7-2 (CD86) expressed on APC can bind to both CD28 and CTLA-4 (CD152), because of higher affinity, they preferentially bind to CTLA-4 on activated T cells and attenuate the T cell response [94].

The importance of CTLA-4 in the down-regulation of T cell responses and in the induction of anergy and tolerance to alloantigens, tumors and pathogens, has been clearly demonstrated in experiments with CTLA-4 deficient mice. The mice developed a severe inflammatory disorder due to up-regulated proliferation of T cells [95, 96]. CTLA-4 can down-regulate T cell responses involving binding and sequestering B7 molecules from CD28, therefore preventing CD28-mediated co-stimulation. Another possibility is that CTLA-4 through its intracellular domain could actively transmit a negative signal resulting in down-regulation of activated T cells [97]. The crucial role of CTLA-4 in maintaining self-tolerance breakdown of which leads to the initiatition of a primary autoimmune response has been demonstrated in several murine models of autoimmune diabetes [98] and autoimmune thyroiditis [32].

Human Leukocyte Antigen class I and II genes

Genes of the human MHC region are clustered on chromosome 6p21 and encode HLA glycoproteins and a number of additional proteins, which are predominantly related to immune response. The MHC locus itself contains three groups of genes: class I genes encoding HLA antigens A, B and C, class II genes encoding HLA-DR, DP and DQ molecules and class III genes [99].

Previous studies in the early 1980s investigated the HLA locus in relation to the genetics of HT. Associations between HLA and HT have both been analysed by serologic typing of HLA and DNA typing using sequence-specific oligonucleotide probe analysis or restriction fragment length polymorphism. In Asians, HLA class I (A2, B16, B35, B46, B51, B54, C3) and HLA class II (DR2, DR9, DR53, DQ4) genes showed an association with the disease [31, 100–105]. In Caucasians, HT is associated with HLA class II genes such as DR3, DR4, DR5, DQA1*0301, DQB1*0201 and DQB1*0301 [106–120] but not with the HLA-DP and HLA class I (HLA-A, HLA-B and HLA-C) genes [113, 114, 121]. However, some studies could not reveal an association between HLA-DQ and DR genes and Hashimoto thyroiditis [114, 122, 123]. Reports of disease-associated alleles are not consistent, but associations appear to be strongest with alleles in the HLA-DR and -DQ loci. This has also been suggested by studies in transgenic mouse [30, 52, 85–87].

Early linkage, non-genome-wide studies of the HLA region have failed to detect linkage between the HLA locus and HT [124–129]. Using dataset of 56 US Caucasian multigenerational families, genome-wide scans has revealed a susceptibility locus AITD-1 located on chromosome 6p [130]. The AITD-1 locus is common for both general forms of thyroid autoimmunity, HT and GD [130]. This locus was replicated in the expanded dataset of 102 US Caucasian families but is distinct from the HLA gene cluster [131]. Whole-genome scans of a large family with members affected with vitiligo and HT mapped a HT susceptibility locus that shared both the MHC region and the non-MHC AITD-1 [132]. However, evidence for linkage between the HLA locus and HT (or autoimmune thyroid disease) has not been confirmed by further whole-genome scans of other affected families [133, 134], sibling pairs [135], or within HLA-DR3 positive families [120]. The lack of linkage means, for instance, the DR3 gene did not cause the familial segregation of Hashimoto's disease while a relatively strong and consistent association showed that HLA-DR3 conferred a generalized increased risk of HT in the general population. These data did not support a major role for the HLA region in the susceptibility to HT and may imply that the DR3 gene modulates the effect of other non-HLA susceptibility gene.

However, a linkage between the HLA region and HT was recently shown in the data set of 40 US multiplex families affected with AITD and type 1 diabetes [136]. The linkage to HT was found to be weaker than to diabetes, suggesting that additional, non-HLA loci were contributing to the joint susceptibility to AITD and T1D. Among HLA-DR alleles, HLA-DR3 was detected as the only associated gene for Hashimoto's thyroiditis and diabetes [136]. Indeed, DR3 seems to represent the major HLA allele, which contributes to the shared susceptibility to T1D and AITD. These findings, however, need to be replicated in larger data sets because early family [137, 138] and case-control [139, 140] studies have not shown the unique role for HLA-DR3 allele in conferring shared susceptibility to T1D and thyroid autoimmunity.

The HLA region has been established to be involved in multiple autoimmune disorders [141]. The mechanisms by which HLA molecules influence the susceptibility to autoimmune disorders become more and more clear. Different HLA alleles could have different affinities to autoantigenic peptides. Therefore, certain alleles can bind the autoantigenic peptide, with the subsequent recognition by T cells that have escaped self-tolerance, whereas others may not [142]. The possibility of certain class II alleles to bind and present thyroid-specific antigens such as TSHR or Tg peptides has been shown in vitro [143] and in mice with EAT [144].

Thyroid autoantigens need to occur in the thyroid or its draining lymph nodes in order for them to be presented by HLA molecules. It has been suggested that an aberrant intrathyroidal expression of MHC class II molecules by thyrocytes is necessary to initiate thyroid autoimmunity [145, 146]. This hypothesis is supported by detection of the expression of HLA class II molecules by thyroid epithelial cells in HT and GD patients [147, 148] and in studies on animal models with experimentally induced thyroid autoimmunity [85, 145, 149, 150]. The aberrant expression of HLA class II antigen by thyrocytes can initiate autoimmune responses through direct thyroid self-antigen presentation or a secondary event following on from cytokine secretion by infiltrated T lymphocytes [148, 151].

Genetic contribution of HLA varies depending on the disease. HLA involvement in T1D, rheumatoid arthritis or multiple sclerosis is large and can constitute more than 50% of the genetic risk [84, 152]. Contributions of HLA alleles as genetic risk factors to HT are much weaker [118, 153]. HLA class I and II genes appear to contribute to the autoimmunity in general but not to organ specificity. Their role in the predisposition to HT is rather non-specific [62, 117]. The HLA class I and II genes appear not to be the primary HT genes, and are likely to be modulating genes that increase the risk for AITD contribution by other genes. HLA class III and other non-HLA genes, located in the HLA region, are also critical to the immune response. It is possible that HLA associations as seen in thyroid autoimmunity are due partially to genetic variation in these closely linked immune regulatory genes and their linkage disequilibrium with class I and II genes [154].

HLA class III genes and non-HLA genes of the HLA region

The HLA class III region lies between class I and II genes and encodes important immunoregulatory proteins such as cytokines [tumour necrosis factor (TNF), lymphotoxin alpha (LT-α) and beta (LT-β)], complement components (C2, C4, properdin factor B) and heat shock proteins (HSP) [155]. Both TNF and LT-α mediate B-cell proliferation and humoral immune responses [154]. TNF has been found to enhance cellular expression of HLA class I and II antigens, and enhances adhesion and complement regulatory molecules in the thyroid gland of HT patients. Alterations to the above could promote the autoimmune process [156]. However, case-control studies showed no association between polymorphisms within the TNF and LT-α genes and HT in Germans [112], UK Caucasians [118] and Koreans [157].

HSP70 gene cluster consists of three genes encoding HSP70-1, HSP70-2 and HSP-Hom proteins. They are expressed in response to heat shock and a variety of other stress stimuli (e.g. oxidative free radicals, toxic metal ions and metabolic stress). HSPs are also important for antigen processing and presentation [158]. Genetic variations within all three HSP70 genes were tested in British patients with HT and no associations were found [118]. Polymorphisms of complement component-encoding genes have not yet been evaluated in relation to HT. Meanwhile, finding a link between frequency disturbances in BI and C4A allotypes and one of the forms of thyroid autoimmunity, postpartum thyroiditis [159], may be an intriguing future study in HT patients.

Other genes crucial to the immune response, including TAP (transporters associated with antigen processing), LMP (large multifunctional protease), DMA and DMB genes are located within the HLA class II region [155]. Protein products of TAP (TAP1 and TAP2) and LMP2 (LMP2 and LMP7) genes participate in the proteolysis of endogenous cytoplasmic proteins into small fragments and subsequent transportation of these self-peptides from the cytoplasm into the endoplasmic reticulum, the site of HLA class I assembly [160]. To date, one investigation has been concerned with the association between TAP1 and TAP2 genes and Hashimoto's thyroiditis. No significant association was observed in the British population [118]. The genetic role of LMP in HT has not yet been examined. An association between the R60 allele of the LMP2 gene and GD was observed [161]. Additionally, quantitative defects in the amount of transcription products of TAP1, TAP2, LMP2 and LMP7 genes were found in lymphocytes of patients with AITD [160]. These findings suggest that defective transcription of HLA class I-processing genes could contribute to the quantitative defect in cell-surface expression in autoimmune lymphocytes in HT. Further evaluation of the role of such class I-processing genes as TAP and LMP is necessary.

DMA and DMB genes are involved in the assembly of HLA class II peptides. These genes encode subunits of a functional heterodimer that is critical for class II antigen presentation [160, 162]. Based on nucleotide variation within exon 3, three rare DMB alleles (DMB*01kv1, DMB*01kv2 and DMB*01kv3) have been detected in Korean HT patients while these DMB variants have not been found in healthy subjects [163]. However, these DMB alleles have not yet been functionally characterized. In summary, there is a significant dearth of information on how HLA class III genes and non-HLA genes, located in the HLA region, contribute to the pathogenesis of HT. Further studies are required to clarify the involvement of these genes in HT susceptibility.

CTLA-4 gene

The CTLA-4 gene is the most frequently studied of the immune modulatory genes located outside the HLA region, in relation to the genetics of HT. This gene encodes a costimulatory molecule, which suppresses T-mediated immune response and is crucial in the maintenance of peripheral immunological self-tolerance [164]. An inventory of case-control studies based on the association between three polymorphic markers within the CTLA-4 gene [A49G dimorphism in the leader peptide, C (-318) T substitution in the promoter region and a dinucleotide repeat polymorphism at the 3'-untranslated region (3'-UTR)] and HT is reviewed in [165]. Results of these studies, except for those for the C (-318) T single nucleotide polymorphism, suggest that polymorphisms within the CTLA-4 gene are associated with the development of HT.

Family studies showed linkage between CTLA-4 and GD [166], thyroid antibody production [167] and autoimmune thyroid disease [12, 62] but not specifically to HT, probably due to lack of their power [129, 130, 135]. Classical linkage analysis is suitable for detecting susceptibility loci with major genetic effects. CTLA-4 demonstrates a modest but significant effect in the genetics of HT. To detect a locus with a modest genetic effect, a large number (at least 400) of affected families should be tested [168].

This investigation has recently been performed involving about 600 AITD families and more than 1300 affected patients [169]. The CTLA-4 gene has been found to play a critical role in the pathogenesis of autoimmune diseases such as GD, HT and T1D [62, 153, 169]. Disease susceptibility was mapped in the 6.1-kb 3' untranslating region of CTLA-4. Allelic variation was correlated to altered mRNA levels of soluble form of CTLA-4 [169]. This alternative splice form of CTLA-4 lacks exon 3 encoding the transmembrane domain but maintains exon 2 encoding the ligand-binding domain [170]. The short form of CTLA-4 can bind CD80/86 and inhibit T-cell proliferation [171]. The soluble CTLA-4 (sCTLA-4) is expressed constitutively by T regulatory cells suppressing the effector T-cell response [172]. Its role in autoimmune disease is not exactly clear, but sCTLA-4 was observed significantly more often in patients with AITD [173] and myasthenia gravis [174] in comparison with non-affected subjects. Patients with AITD and myasthenia gravis had an aberrant expression of the CTLA-4 products, with high levels of sCTLA-4 and low levels of the intracellular form [175]. Soluble CTLA-4 might play an important role in immune regulation by binding with the B7 molecules, thus interfering with the binding of CD28 and/or full-length CTLA-4. Interference of sCTLA-4 with B7/CTLA-4 interactions could block suppressive signals transferred via surface-bound CTLA-4. Therefore, high concentrations of sCTLA-4 in serum might contribute to disease manifestations through interference of sCTLA-4 with B7/CTLA-4 interaction.

It may be that the amino acid change at codon 17 of the signal peptide could alter the function of the signal peptide to direct intracellulat trafficking of CTLA-4. In in vitro expreriments, the Ala17 (G49) allele was found to represent a translation product, which was not glycosylated in one of two N-linked glycosylation sites [176]. This aberrantly glycosylated product was shown to be further translocated from the endoplasmic reticulum back to cytoplasm and, probably, to become a target for proteolytic degradation. In addition, the distribution of Ala17 CTLA-4 variant on the surface of COS1 cells is significantly less density than the Thr17 variant of CTLA-4 [176]. These fundings suggest that the Ala17 allele is linked to the inefficient glycosylation of CTLA-4, which subsequently could affect suppressing effects of the CTLA-4 molecule. This could also explain observations showing that the G49 allele of the CTLA-4 signal peptide is associated with accelerated proliferation of T lymphocytes in human subjects homozygous for this allele, and with suppression of the downregulation of T-cell activation in response to IL-2 [177, 178].

The codon 17 single nucleotide polymorphism (SNP) is shown to be in tight linkage disequilibrium with another SNP situated at position (-318) of the CTLA-4 promoter region and with the (AT)n repeat polymorphism at the 3'-UTR of the CTLA-4 gene [176, 179–181]. For the C (-318) T SNP, the protective T (-318) allele demonstrated higher promoter activity than the alternative C allele in a luciferase expression assay [182]. Since the (-318) dimorphism occurs in a potential regulatory region, this suggets that this nucleotide substitution may influence the expression of CTLA-4. However, this possibility remains to be explored.

The (AT)n repeat polymorphism at the 3'-UTR of the CTLA-4 gene has been shown to affect the expression of this costimulatory molecule [174]. Adenylate- and uridylate-rich elements (AUREs) presented in the 3'-UTRs can regulate stability of eukaryotic mRNAs, and their presence correlates with rapid RNA turnover and translational and posttranslational control [183, 184]. The AT repeats in the 3'-UTR of CTLA-4 might represent a special type of of AUREs. CTLA-4 mRNA with longer (AT)n alleles have shorter half-lives and, hence, are more unstable [174, 185]. Indeed, the (AT)n microsatellite in the 3'-UTR influences the mRNA stability. Additionally, the CTLA-4 AT-repeat polymorphism was recently shown to alter the inhibitory function of CTLA-4. The long AT-repeat allele is associated with reduced control of T-cell proliferation and thus contributes to the pathogenesis of GD [186].

The AT-repeat may also affect splicing of one or more of the alternative CTLA-4 transcripts but this should be clarified. Ueda et al. [169] showed that another polymorphism (A6230G, or CT60 SNP) located in the first position of the 3'-UTR correlates with higher expression of a soluble CTLA-4. In this study, the highest power of linkage with GD was found for this SNP and three other SNPs (JO27, JO30 and JO31) within a 6.1-kb segment of the 3'-UTR, but not for the (AT)n repeat polymorphism [169]. However, no T-cell function data were presented. Thus, further investigations are necessary to evaluate functional significance of these SNPs. Due to the linkage disequilibrium, it is currently not possible to determine whether one, or both, are of physiological importance. It can not be excluded that allele combination of several closely linked CTLA-4 polymorphisms might form a functionally significant haplotype that is directly involved in the susceptibility to autoimmune disease [187, 188].

It should be noted that the genomic region 2q33 linked to autoimmune disease contains cluster of three genes encoding costimulatory molecules CTLA-4, CD28 and inducible costimulator (ICOS) [189]. However, genetic studies showed that the AITD gene in the 2q33 locus is the CTLA-4 gene and not the CD28 or ICOS genes [167, 169, 181].

The CTLA4 gene should be recognised as the first major known non-HLA locus of human autoimmunity and that its role in the pathogenesis of HT is rather general and non-specific [74, 153]. Association of CTLA-4 with the production of thyroid antibodies [167, 190], an event that often represents the subclinical stage of AITD [1], can explain non-specific mechanism of CTLA-4-mediated susceptibility to the development of thyroid autoimmunity. The association of the CTLA-4 gene with several autoimmune diseases such as T1D [153, 169], Addison's disease [191, 192], multiple sclerosis [193, 194], myasthenia gravis [175] and all clinical outcomes of AITD [74], can also explain the general contribution of CTLA-4 to autoimmunity. Interestingly, AITD, Addison's disease and autoimmune diabetes frequently coexist in patients with the autoimmune polyendocrine syndrome type II as mentioned above. The above disorders seem to share a genetic background, and CTLA-4 could represent a common susceptibility focus for them [195]

Other immune regulatory genes

In initial phases of AITD, oligoclonal expansion of T lymphocytes occurs in the thyroid gland. These T cells are restricted by their T cell receptor V gene use [89, 90]. Therefore, the TcR may be considered a likely candidate gene for AITD and HT. Early case-control investigations showed a lack of association between HT and the T-cell receptor-α gene in the US white population [111] but not the T-cell receptor-β gene in the Japanese [102]. Linkage analysis using a US Caucasian AITD family dataset [129] and Tunisian affected pedigree [196] has eliminated the T-cell receptor V alpha and V beta gene complexes, located on 14q11 and 7q35, respectively, as candidate genes for susceptibility to thyroid autoimmunity. Therefore, the TcR genes are not major susceptibility genes for HT and AITD.

Another likely candidate among immune-related genes was the IGH gene because HT individuals commonly produce Tg-autoantibodies restricted by IgG class [50]. Early investigations found an association between IgH Gm allotypes and AITD in the Japanese [197, 198]. However, these findings have not been confirmed in Caucasians [129, 196].

Cytokines are crucial in the regulation of immune and inflammatory responses. Multiple investigations showed the important role of these regulatory molecules in directing autoimmune and apoptotic pathogenic processes, of particular, in central and late stages of the development of HT [72, 80, 199]. Therefore, cytokine genes might be good candidates for HT. Intrathyroidal inflammatory cells and thyroid follicular cells produce a variety of cytokines, including interleukin-1α (IL-1α), IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, IL-13, IL-14, tumor necrosis factor-α, and interferon-γ [200]. Hunt et al. [201] evaluated 15 polymorphisms within nine cytokine genes for IL-1α, IL-1β, IL-1 receptor antagonist (IL1RN), IL-1 receptor 1, IL-4, IL-4 receptor, IL-6, IL-10, and transforming growth factor-β in British patients with AITD. They only found a significant association for one of those. The T-allele of the IL-4 promoter [T (-590) C] polymorphism was associated with lower risk of GD and AITD but not HT [201]. Blakemore et al. [202] failed to find an association between a polymorphic minisatellite in the IL1RN gene and HT in another group of affected patients from UK. Thus, it may be concluded that these genes are not major susceptibility genes for thyroid autoimmunity but need to be further studied.

The autoimmune regulator (AIRE1) gene is known to contribute to the pathogenesis of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), a rare monogenic autoimmune disease with endocrine components including T1D, adrenal failure, and thyroid dysfunction, with major autoantibodies directed against adrenal, pancreas, and thyroid tissue [203]. However, studies in UK patients showed no relation between a 13-bp deletion at nucleotide 964 in exon 8 (964del13) of the AIRE1 gene, a common disease-associated marker for APECED in British population, and HT [204].

The vitamin D-mediated endocrine system plays a role in the regulation of calcium homeostasis, cell proliferation and (auto) immunity. 1,25-Dihydroxi-vitamin D3 (1,25(OH)2D3) is the most active natural vitamin D metabolite that effectively prevents the development of autoimmune thyroiditis in an animal model [205] and inhibits HLA class II expression on endocrine cells [206]. C/T polymorphism located at intron 6 of the vitamin D 1α-hydroxylase (CYP1α) gene failed to show association with HT in Germans [207]. Two polymorphic markers within the vitamin D-binding protein gene encoding another member of the vitamin D metabolic pathway also showed no association with HT in the German population [208]. However, among two polymorphic sites tested at the vitamin D receptor (VDR) gene, the Fok I(+) allele of the FokI/restriction fragment length polymorphism was found to be associated with higher risk HT in Japanese females [209]. Meanwhile, the VDR gene remains to be a likely candidate for the common autoimmune susceptibility gene because it has been found to be associated with autoimmune disorders such as GD [210], Addison's disease [211], multiple sclerosis [212] and T1D [213].

Thus, a wide variety of non-HLA immune regulatory genes located outside the HLA region showed no significant linkage or association with HT and AITD except for the CTLA4 gene. However, we still cannot estimate whether or not these genes significantly contribute to HT susceptibility due to a serious shortfall in information about their role in this disorder. It cannot be excluded that other genes in linkage disequilibrium with these genes are the susceptibility genes at these loci.

Thyroid-specific genes

Antibodies against thyroid peroxidase are one of the most specific features of HT [214]. Therefore, the TPO gene is expected to be a putative candidate responsible not only for susceptibility to HT but also for specific determination between two common outcomes of AITD, such as HT and GD. Genetic transmission of the recognition by antibody of the TPO immunodominant region and the TPO B domain has been described in families affected with HT [215]. This transmission could be explained by genetic variations within the TPO gene. However, case-control studies showed lack of association between the TPO gene polymorphisms and AITD [113, 216]. These data suggest that the thyroid peroxidase gene does not play an important role in predisposition to HT. Subsequent studies are necessary to clarify exactly whether this gene is a true susceptibility gene for AITD.

Within the other thyroid-specific gene, the TSHR gene, the T52P amino acid substitute was examined in US white and Thai populations but no association with HT was found [217, 218]. Various genome-wide scans have failed to detect linkage between the thyrotropin receptor gene and HT or AITD [130, 133–135, 219, 220]. However, two microsatellites, an (AT)n marker at intron 2 of the TSHR gene and a (CA)n marker that was mapped to approximately 600 kb of the TSHR gene, have been shown to be strongly associated with HT in Japanese patients [221, 222]. The TSHR gene, therefore, does not seem to be a major susceptibility gene for HT, although a minor role cannot be excluded.

Tg-specific autoantibodies are common in AITD. The thyroglobulin gene makes a significant contribution to HT and AITD. Whole-genome scans in Japanese-affected sibling pairs have detected a HT susceptibility locus on chromosome 8q24, with a maximum linkage to marker D8S272 [135]. This marker is separated by 4.6 megabases (Mb) from the Tg gene. Subsequent studies of the mixed US and European Caucasian family dataset has confirmed the susceptibility locus to be on chromosome 8q24, with the maximum linkage to markers D8S514 and D8S284 [74, 131, 223]. These markers border a large region of chromosome 8 spanning about 15 Mb. The thyroglobulin gene is located within this region. Moreover, a new microsatellite marker Tgms2 inside intron 27 of the Tg gene showed strong evidence of linkage and association with AITD [223, 224]. Two new microsatellites have recently been described in introns 29 and 30 of the thyroglobulin gene that can be useful for further linkage studies in families with autoimmune thyroid diseases [225]. Using a high-density panel of SNPs within the human and murine Tg genes, Ban et al. [226] identified a unique SNP haplotype, consisting of an exon 10–12 SNP cluster in both genes and, additionally, exon 33 SNP in the human gene, associated with AITD in humans and with EAT in mice. Taken together, these data strongly suggest that the thyroglobulin gene could represent the susceptibility gene for HT and AITD on 8q24 [74, 227] and, therefore, be characterized as the first thyroid-specific susceptibility gene for thyroid autoimmunity [228].

The Tg gene spanning over 300 kilobases long is expected to harbour more than one haplotype block associated with AITD since the length of a linkage disequilibrium block of SNPs is shown to be less than 100 kilobases [229]. It seems that this gene is AITD-specific but is not a HT-specific susceptibility gene. The manner in which the Tg gene can be a predisposition to AITD remains unclear. It could be that amino acid variations within the Tg gene can affect the immunogenicity of Tg. The evidence that iodination of thyroglobulin affects its immunogenicity favours this suggestion [230, 231]. However, additional studies are required to evaluate that.

Recent investigation in Tunisians showed significant association of two polymorphic microsatellites (D7S496 and D7S2459) close to the PDS gene (7q31) with GD and HT, and one of them, D7S496, was linked to GD only [232]. The PDS gene encodes a transmembrane protein known as pendrin. Pendrin is a chloride/iodide transporting protein identified in the apical membrane of the thyroid gland [233]. Data of Kacem et al. [232] suggest that the PDS gene might be considered a new susceptibility gene to autoimmune thyroid diseases, having a different involvement with different diseases. However, studies in other populations are necessary to support a role for the PDS gene in thyroid autoimmunity and HT.

Finally, a role for other genes specifically expressed in the thyroid gland, has yet to be defined. These genes include those encoding thyrotropin-β, thyroid-specific factor-1, sodium iodide (Na+/I) symporter and paired box transcription factor-8, among others. They also need to be evaluated for any putative impact on HT.

Apoptotosis-related genes

Two polymorphic sites within the FasL gene were recently tested in HT Caucasian patients from Italy and Germany. No association between these polymorphisms and the disorder was shown [234]. Assuming a lack of association of the naturally occurring FasL gene polymorphisms with multiple other autoimmune diseases tested, we conclude that genetic variation within this gene does not contribute to autoimmunity. Inactivating mutations within the Fas and FasL genes are associated with carcinogenesis [235, 236]. This situation is common among apoptotic-related genes encoding caspases, death receptors, decoy receptors and death ligands as well as for genes that encode other types of signalling molecules [237]. However, since apoptotic mechanisms play a critical role in pathogenesis and progression of HT, genes associated with programming cell death should be evaluated whether or not they confer susceptibility to HT.

Other genes

Due to the prevalence of thyroid autoimmunity in females, gender-related genes could also be considered as putative candidates for HT susceptibility. Some of these genes, such as the CYP19 gene encoding aromatase that participates in estrogen synthesis, and genes for estrogen receptor-α (ESR1) and -β (ESR2), were examined but showed no linkage with HT [238]. The ESR1 and ESR2 genes demonstrated no association with AITD in the Japanese [239, 240]. It seems that the CYP19 and both estrogen receptor genes do not predispose to HT and AITD. Other gender-specific genes could contribute to AITD. A possible involvement of such genes has been shown for GD with the discovery of a susceptible locus on chromosome X [238].

The SEL1L gene, encoding a novel transcription factor, was recently described [241]. The gene is located on chromosome 14q24.3-q31 close to the GD-1 susceptibility locus [128, 130, 219] and considered a likely candidate for thyroid autoimmunity. However, a case-control study in the Japanese population detected no association of a dinucleotide (CA)n repeat polymorphism in the intron 20 of the SEL1L gene with AITD [242]. This gene may be a potentially predisposing gene to T1D because it is specifically expressed in adult pancreas and islets of Langerhans [241]. It lies in the vicinity to IDDM11, a susceptibility locus to this autoimmune disease, on chromosome 14q24.3-q31 [243].

A new zink-finger gene designated ZFAT (a novel zink-finger gene in AITD susceptibility region) has been recently found on chromosome 8q24 [244]. The T allele of the Ex9b-SNP10 dimorphism representing an adenine-to-thymidine substitution within intron 9 of this gene was shown to be associated with high risk for AITD in Japanese patients. Functional studies showed that the Ex9b-SNP10 significantly affects the expression of the small antisense transcipt of ZFAT (SAS-ZFAT) in vitro and this expression results in the decreasing expression of the truncated form of ZFAT (TR-ZFAT) [244]. This SNP is located in the 3'-UTR of TR-ZFAT and in the promoter region of SAS-ZFAT. Full-length ZFAT and TR-ZFAT encode a protein with unknown function, which has eighteen and eleven repeats of zink-finger domains, respectively. Both molecular variants of ZFAT are expressed in different tissues including peripheral blood lymphocytes, while SAS-ZFAT is exclusively expressed in peripheral blood B cells and represents a non-coding RNA with putative regulatory function [245]. The disease-associated polymorphism can play a significant role in B cell function by enfluencing the expression level of TR-ZFAT through regulation of transcription of SAS-ZFAT. Interestingly, Shirasawa et al. found no association of the thyroglobulin gene with AITD when studying different ethnic groups [244]. These results suggest that the ZFAT gene could implicate the susceptibilty to AITD on chromosome 8q24 but that it needs to be strongly replicated in other populations. Additionally, the ZFAT gene should be functionally studied to clarify whether the ZFAT or thyroglobulin gene are true contributors of genetic susceptibility to AITD and HT on 8q24.

Non-defined susceptibility loci for Hashimoto's thyroiditis and autoimmune thyroid disease

At present, the CTLA-4 (chromosome 2q33), thyroglobulin (or ZFAT) (8q24) and likely HLA genes (6p21.3) are the only susceptibility loci for HT and thyroid autoimmunity to be mapped. Two HT-specific susceptibility loci that have been detected in mixed Caucasian families from USA and Europe, HT-1 (13q) near marker D13S173 and HT-2 on chromosome 12q in the vicinity of marker D12S351, are still not defined [130]. HT-2 locus has been subsequently replicated in the extended dataset, with a peak linkage close to marker D12S346, which HT-1 does not have [223]. Possible candidate genes for susceptibility to HT positioned within the HT-2 locus may include the BTG1 and CRADD genes. The BTG1 gene encodes B-cell translocation protein-1, which play an immune regulatory role as a negative regulator of the proliferation of B cells [246]. The GRADD gene encodes CASP-2 and RIPK-domain-containing adaptor with death domain, that represents apoptotic function, inducing cell apoptosis via recruiting caspase 2/ICH1, TNF receptor 1, RIPK-RIP kinase and other proteins [247].

AITD-1 locus located on chromosome 6p is very close yet distinct from the HLA region [120, 130]. It has been shown that the AITD-1 is positioned in the same location as susceptibility loci for T1D (locus IDDM15) [248] and systemic lupus erythematosus [249]. This may imply that a general autoimmunity susceptibility gene is located in this region. The AITD-1 locus contains an interesting positional candidate gene such the SOX-4 gene, encoding a transcription factor that modulates differentiation of lymphocytes [250].

A whole-genome scan in Japanese showed evidence for linkage with AITD on chromosome 5q31-q33 [135, 251]. The 5q31 locus was replicated by recent genome-wide scan in Caucasian population, the Old Order Amish of Lancaster County, from Pennsylvania [220]. This locus harbours a cluster of cytokine genes and, therefore, several positional candidate genes occur in this region and need to be evaluated.

In the Chinese, a whole-genome screening for AITD susceptibility found two chromosome regions (9q13 and 11q12) linked to AITD [134]. Susceptibility genes have yet to be defined within these regions. However, the 9q13 locus harbours a putative candidate gene such as the ANXA1 gene, whose product annexin A1 prevents the production of inflammatory mediators [252]. The 11q12 locus contains several interesting candidate genes encoding immune modulators (CD5 and CD6) and possible components of antigenic peptide processing (PSMC3) and transport (PTH2).

In a large Tunisian family affected with AITD, a susceptibility locus was mapped on 2p24 [133] This locus harbors two possible candidate genes such as the FKBP1B gene, product of which demonstrates immune modulating activity [253], and the TP53I3 gene encoding p53-inducible protein 3 that is involved in p53-mediated apoptotic pathway [254].

These data suggest that both HT and AITD show genetic heterogeneity in different populations. Susceptibility loci differ in their chromosome location depending on the population being tested. The contributory value of these genes to the disease pathology varies significantly depending on the ethnic background. A gene, that has a major effect on the susceptibility to HT in one population, may contribute weakly in other population. To date, several regions of linkage to HT and AITD have been defined. Further studies are required to find a true susceptibility gene in these genomic regions to reveal the functional significance of disease-associated polymorphisms within these genes.

Conclusion

AITD can be initiated in individuals genetically predisposed to AITD by non-genetic (environmental) triggers such as dietary iodine, infection, pregnancy, cytokine therapy (Fig. 1). This interaction leads to different clinical phenotypes of thyroid autoimmunity such as Graves' disease, Hashimoto's thyroiditis or production of thyroid antibodies. HT and GD are two distinct but related clinical outcomes of AITD. It seems that both thyroid diseases have common pathogenic mechanisms as their initial steps including breakdown of the immune tolerance and accumulation of T lymphocytes in the thyroid gland.

Sequence variants of CTLA-4, associated with increased levels of the soluble form of this immune costimulator and with stability of CTLA-4 mRNA, could play a crucial role in the earliest stages of AITD (i. e. breakdown of self-tolerance and surviving autoreactive T lymphocytes). This role might be sufficient to regulate subsequent steps in the development of autoimmune responses to the production of thyroid autoantobodies [167].

Environmental factors (particularly, iodine intake and infection) could cause insult of the thyrocyte followed by abnormal expression of MHC class I and class II molecules, as well as changes to genes or gene products (such as MHC class III and costimulatory molecules) needed for the thyrocyte to become an APC [255]. In this stage, a modulating role of sequence variants of HLA class II molecules could become pivotal in binding and presenting thyroid antigenic peptides derived from Tg, TPO and TSHR. Genetic variations in Tg, and probably in TSHR and other thyroid-specific genes, might be responsible for generating an autoimmune response.

In later stages, thyroid autoimmunity could be switched towards GD or HT. GD is characterized by TH2-mediated switching of thyroid-infiltrating T cells. These induce the production of stimulating anti-TSHR antibodies by B cells and anti-apoptotic mechanisms that lead to clinical hyperthyroidism. In HT, preferential TH1 response initiates apoptosis of thyroid cells and results in clinical hypothyroidism [22].

It is clear that a number of loci and genes determine genetic predisposition to HT, with varying effects. These loci could be unique to HT or general for both HT and GD. Several whole-genome scans showed results suggesting that there is significant shared susceptibility to HT and GD [130, 131, 134, 135]. This is also supported by the frequent coexistance of both diseases in affected families [74, 133]. Preliminary data suggest that shared genetic susceptibility involves both immune regulatory (i. e. CTLA-4 and HLA) and thyroid-specific genes (i.e. Tg). These genes are not responsible for the determination of pathogenic mechanisms of thyroid autoimmunity distinct for HT and GD. It remains unclear which susceptibility genes are specifically involved in the HT pathogenesis.

The CD40 gene, an important immune modulator, appears to act as a GD-specific susceptibility gene. The gene is located within the 20q11 locus and shows significant linkage to GD, but not to HT, in UK Caucasians [74, 130, 131, 256, 257]. Subsequent analysis found the CD40 gene to be associated with GD [258]. However, this finding needs to be independently confirmed in other population samples.

A probable susceptibility gene that could direct switching towards GD or HT is thought to be located within the 5q31 locus, which is linked to AITD and contains a cytokine gene cluster. Different sets of cytokines are known to regulate switching to TH1 or TH2 type mechanisms [58]. There may be two susceptibility genes, each of which uniquely contributes to the development of HT- or GD-specific pathogenesis. The IL-4 promoter [T(-590) C] polymorphism also appears to be associated with GD, but not with HT [199]. IL-4 mediates TH2 type mechanism, which can lead to hyperthyroidism [259].

Another HT-specific susceptibility gene(s) may be an apoptotic gene. Apoptosis of thyroid follicular cells is the hallmark of HT and might be the primary cause of death of thyrocytes compared to T cell-mediated cytotoxity [69].

Thus, it is necessary to identify additional susceptibility genes and disease-associated polymorphisms in apoptotic genes in AITD- and HT-linked loci by using a fine mapping approach and high-density panels of SNPs. Further functional analysis and search for correlations between genotype and phenotype will help to evaluate the role of these genes in the development of autoimmune thyroid disease. Susceptibility genes interact with thyroid autoimmunity [62, 130], and the level of these interactions could affect disease severity and clinical expressions. The molecular mechanisms of these interactions is unknown. However, significant progress has been made in identifying susceptibility genes to HT and AITD along with intriguing findings regarding the functional characterization of disease-associated polymorphisms. These should stimulate further studies towards the in-depth understanding of the mechanisms by which these genes contribute to thyroid autoimmunity.

References

  1. Vanderpump MP, Tunbridge WM: Epidemiology and prevention of clinical and subclinical hypothyroidism. Thyroid. 2002, 12: 839-847. 10.1089/105072502761016458.

    PubMed  Google Scholar 

  2. Wang C, Crapo LM: The epidemiology of thyroid disease and implications for screening. Endocrinol Metab Clin North Am. 1997, 26: 189-218.

    CAS  PubMed  Google Scholar 

  3. Tunbridge WM, Vanderpump MP: Population screening for autoimmune thyroid disease. Endocrinol Metab Clin North Am. 2000, 29: 239-253.

    CAS  PubMed  Google Scholar 

  4. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, Braverman LE: Serum TSH, T4, and thyroid antibodies in the United states population (1988 to 1994): National health and Nutrition Examination Durvey (NHALES III). J Clin Endocrind Metab. 2002, 87: 489-499. 10.1210/jc.87.2.489.

    CAS  Google Scholar 

  5. Tunbridge WM, Evered DC, Hall R, Appleton D, Brewis M, Clark F, Evans JG, Young E, Bird T, Smith PA: The spectrum of thyroid disease in a community: the Whickham survey. Clin Endocrinol (Oxf). 1977, 7: 481-493.

    CAS  Google Scholar 

  6. Okamura K, Ueda K, Sone H, Ikenoue H, Hasuo Y, Sato K, Yoshinary M, Fujishima M: A sensitive thyroid hormone assay for screening a thyroid functional disorder in elderly Japanese. J Am Geriatr Soc. 1989, 37: 317-322.

    CAS  PubMed  Google Scholar 

  7. Gordin A, Heinonen OP, Saarinen P, Lamberg BA: Serum thyrotropin in symptomless autoimmune thyroiditis. Lancet. 1972, 1: 551-554. 10.1016/S0140-6736(72)90353-4.

    CAS  PubMed  Google Scholar 

  8. Vanderpump MP, Tunbridge WM, French JM, Appleton D, Bates D, Clark F, Grimley EJ, Hasan D, Rodgers H, Tunbridge F, Young ET: The incidence of thyroid disorders in the community: a twenty-year follow-up of the Whickham Survey. Clin Endocrinol (Oxf). 1995, 43: 55-69.

    CAS  Google Scholar 

  9. Hueston WJ: Treatment of hypothyroidism. Am Fam Physician. 2001, 64: 1717-1724.

    CAS  PubMed  Google Scholar 

  10. Devendra D, Eisenbarth GS: 17. Immunological endocrine disorders. J Allergy Clin Immunol. 2003, 111: 624-636. 10.1067/mai.2003.81.

    Google Scholar 

  11. Jenkins RC, Weetman AP: Disease associations with autoimmune thyroid disease. Thyroid. 2002, 12: 977-988. 10.1089/105072502320908312.

    PubMed  Google Scholar 

  12. Einarsdottir E, Soderstrom I, Lofgren-Burstrom A, Haraldsson S, Nilsson-Ardnor S, Penha-Goncalves C, Lind L, Holmgren G, Holmberg M, Asplund K, Holmberg D: The CTLA4 region as a general autoimmunity factor: an extended pedigree provides evidence for synergy with the HLA locus in the etiology of type 1 diabetes mellitus, Hashimoto's thyroiditis and Graves' disease. Eur J Hum Genet. 2003, 11: 81-84. 10.1038/sj.ejhg.5200903.

    CAS  PubMed  Google Scholar 

  13. Niederwieser G, Buchinger W, Bonelli RM, Berghold A, Reisecker F, Koltringer P, Archelos JJ: Prevalence of autoimmune thyroiditis and non-immune thyroid disease in multiple sclerosis. J Neurol. 2003, 250: 672-675. 10.1007/s00415-003-1053-9.

    PubMed  Google Scholar 

  14. Zettinig G, Tanew A, Fischer G, Mayr W, Dudczak R, Weissel M: Autoimmune diseases in vitiligo: do anti-nuclear antibodies decrease thyroid volume?. Clin Exp Immunol. 2003, 131: 347-354. 10.1046/j.1365-2249.2003.02089.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  15. Betterle C, Zanchetta R: Update on autoimmune polyendocrine syndromes (APS). Acta Biomed Ateneo Parmense. 2003, 74: 9-33.

    Google Scholar 

  16. Rose NR, Saboori AM, Rasooly L, Burek CL: The role of iodine in autoimmune thyroiditis. Crit Rev Immunol. 1997, 17: 511-517.

    CAS  PubMed  Google Scholar 

  17. Rose NR, Bonita R, Burek CL: Iodine: an environmental trigger of thyroiditis. Autoimmun Rev. 2002, 1: 97-103. 10.1016/S1568-9972(01)00016-7.

    CAS  PubMed  Google Scholar 

  18. Tomer Y, Davies TF: Infection, thyroid disease and autoimmunity. Endocrine Rev. 1993, 14: 107-120. 10.1210/er.14.1.107.

    CAS  Google Scholar 

  19. Mine H, Kawai H, Yokoi K, Akaike M, Saito S: High frequencies of human T-lymphotropic virus type I (HTLV-I) infection and presence of HTLV-II proviral DNA in blood donors with anti-thyroid antibodies. J Mol Med. 1996, 74: 471-477. 10.1007/s001090050049.

    CAS  PubMed  Google Scholar 

  20. Kakizaki S, Takagi H, Murakami M, Takayama H, Mori M: HLA antigens in patients with interferon-alpha-induced autoimmune thyroid disorders in chronic hepatitis C. J Hepatol. 1999, 30: 794-800. 10.1016/S0168-8278(99)80131-7.

    CAS  PubMed  Google Scholar 

  21. Imaizumi M, Pritsker A, Kita M, Ahmad L, Unger P, Davies T: Pregnancy and murine thyroiditis: thyroglobulin immunization leads to fetal loss in specific allogeneic pregnancies. Endocrinology. 2001, 142: 823-829. 10.1210/en.142.2.823.

    CAS  PubMed  Google Scholar 

  22. Tomer Y: Genetic dissection of familial autoimmune thyroid diseases using whole genome screening. Autoimmun Rev. 2002, 1: 198-204. 10.1016/S1568-9972(02)00053-8.

    CAS  PubMed  Google Scholar 

  23. Boukis MA, Koutras DA, Souvatzoglou A, Evangepolou K, Vrontakis M, Moulapoulaos SD: Thyroid hormone and immunologic studies in endemic goiter. J Clin Endocrinol Metab. 1983, 57: 859-862.

    CAS  PubMed  Google Scholar 

  24. Harach HR, Eslacante DA, Onativia A, Lederer Outes J, Saravia Day E, Williams ED: Thyroid carcinoma and thyroiditis in an endemic goitre region before and after iodine phospholaxis. Acta Endocrinol. 1985, 108: 55-60.

    CAS  PubMed  Google Scholar 

  25. Allen EM, Appel MC, Braverman LE: The effect of iodide ingestion on the development of spontaneous lympholytic thyroiditis in the diabetesprone BB/W rat. Endocrinology. 1986, 118: 1977-81.

    CAS  PubMed  Google Scholar 

  26. Rasooly L, Burek CL, Rose NR: Iodine-induced autoimmune thyroiditis in NOD-H-2h4 mice. Clin Immunol Immunopathol. 1996, 81: 287-292. 10.1006/clin.1996.0191.

    CAS  PubMed  Google Scholar 

  27. Ruwhof C, Draxhage HA: Iodine and thyroid autoimmune disease in animal models. Thyroid. 2001, 11: 427-436. 10.1089/105072501300176381.

    CAS  PubMed  Google Scholar 

  28. Kabel PJ, Voorbij HA, de Haan M, van der Gaag RD, Drexhage HA: Intarthyroidal dendritic cells. J Clin Endocrinol Metab. 1988, 66: 199-207.

    CAS  PubMed  Google Scholar 

  29. Many M-C, Maniratunga S, Varis I, Dardenne M, Draxhage HA, Denef J-F: Two-step development of a Hashimoto-like thyroiditis in genetically autoimmune prone non-obese diabetic (NOD) mice. Effects of iodine-induced cell necrosis. J Endocrinol. 1995, 147: 311-320.

    CAS  PubMed  Google Scholar 

  30. Wan Q, Shah R, Panos JC, Giraldo AA, David CS, Kong YM: HLA-DR and HLA-DQ polymorphism in human thyroglobulin-induced autoimmune thyroiditis: DR3 and DQ8 transgenic mice are susceptible. Hum Immunol. 2002, 63: 301-310. 10.1016/S0198-8859(02)00360-9.

    CAS  PubMed  Google Scholar 

  31. Wan XL, Kimura A, Dong RP, Honda K, Tamai H, Sasazuki T: HLA-A and -DRB4 genes in controlling the susceptibility to Hashimoto's thyroiditis. Hum Immunol. 1995, 42: 131-136. 10.1016/0198-8859(94)00089-9.

    CAS  PubMed  Google Scholar 

  32. Vasu C, Gorla SR, Prabhakar BS: Targeted engagement of CTLA-4 prevents autoimmune thyroiditis. Int Immunol. 2003, 15: 641-654. 10.1093/intimm/dxg061.

    CAS  PubMed  Google Scholar 

  33. Greiner DL, Handler ES, Nakano K, Mordes JP, Rossini AA: Absence of the RT-6 T cells in diabetes-prone BB/W rats. J Immunol. 1986, 136: 148-151.

    CAS  PubMed  Google Scholar 

  34. Wick G, Brezinschek HP, Hala K, Dietrich H, Wolf H, Kroemer G: The obese strain of chickens: an animal model for sponataneous autoimmune thyroiditis. Adv Immunol. 1989, 47: 433-500.

    CAS  PubMed  Google Scholar 

  35. Martin A, Davies TF: T cells and human autoimmune thyroid disease: emerging data show lack of need to invoke suppressor T cell problems. Thyroid. 1992, 2: 247-261.

    CAS  PubMed  Google Scholar 

  36. Bottazzo GF, Pujol-Borell R, Hanafusa T, Feldmann M: Role of aberrant HLA-DR expression and antigen presentation in induction of endocrine autoimmunity. Lancet. 1983, 2: 1115-1118. 10.1016/S0140-6736(83)90629-3.

    CAS  PubMed  Google Scholar 

  37. Rapoport B, McLachlan SM: Thyroid autoimmunity. J Clin Invest. 2001, 108: 1253-1259. 10.1172/JCI200114321.

    PubMed Central  CAS  PubMed  Google Scholar 

  38. Shimojo N, Kohno Y, Yamaguchi K, Kikuoka S, Hoshioka A, Niimi H, Hirai A, Tamura Y, Saito Y, Kohn LD, Tahara K: Induction of Graves-like disease in mice by immunization with fibroblasts transfected with the thyrotropin receptor and a class II molecule. Proc Natl Acad Sci USA. 1996, 93: 11074-11079. 10.1073/pnas.93.20.11074.

    PubMed Central  CAS  PubMed  Google Scholar 

  39. Jaume JC, Guo J, Wang J, Rapoport B, McLachlan SM: Cellular thyroid peroxidase (TPO), unlike purified TPO and adjuvant, induces antibodies in mice that resemble autoantibodies in human autoimmune thyroid disease. J Clin Endocrinol Metab. 1999, 84: 1651-1657. 10.1210/jc.84.5.1651.

    CAS  PubMed  Google Scholar 

  40. Champion BR, Page KR, Parish N, Rayner DC, Dawe K, Biswas-Hughes G, Cooke A, Geysen M, Roitt IM: Identification of a thyroxine-containing self-epitope of thyroglobulin which triggers thyroid autoreactive T cells. J Exp Med. 1991, 174: 363-370. 10.1084/jem.174.2.363.

    CAS  PubMed  Google Scholar 

  41. Rasooly L, Rose NR, Saboori AM, Ladenson PW, Burek CL: Iodine is essential for human T cell recognition of human thyroglobulin. Autoimmunity. 1998, 27: 213-219.

    CAS  PubMed  Google Scholar 

  42. Ebner SA, Lueprasitsakul W, Alex S, Fang SL, Appel MC, Braverman LE: Iodine content of rat thyroglobuline affects its autogenicity in inducing lympholytic thyroiditis in the BB/Wor rat. Autoimmunity. 1992, 13: 209-214.

    CAS  PubMed  Google Scholar 

  43. Mahmoud I, Colin I, Many MC, Denef JF: Direct toxic effect effect of iodide in excess on iodine-deficient thyroid gland: epithelial necrosis and inflammation associated with lipofuscin accumulation. Exp Mol Pathol. 1986, 44: 259-271. 10.1016/0014-4800(86)90040-7.

    CAS  PubMed  Google Scholar 

  44. Simons PJ, Delamarre FG, Drexhage HA: Antigen-presenting dendritic cells as regulators of the growth of thyrocytes: a role for interleukin-1 beta and interleukin-6. Endocrinology. 1998, 139: 3148-3156. 10.1210/en.139.7.3148.

    CAS  PubMed  Google Scholar 

  45. Lamas L, Anderson PC, Fox JW, Dunn JT: Consensus sequences for early iodination and hormonogenesis in human thyroglobulin. J Biol Chem. 1989, 264: 13541-13545.

    CAS  PubMed  Google Scholar 

  46. Male DK, Champion BR, Pryce G, Matthews H, Shepherd P: Antigenic determinans of human thyreoglobulin differentiated using antigen fragments. Immunology. 1985, 54: 419-427.

    PubMed Central  CAS  PubMed  Google Scholar 

  47. Tomer Y: Anti-thyroglobulin autoantibodies in autoimmune thyroid diseases: cross-reactive or pathogenic?. Clin Immunol Immunopathol. 1997, 82: 3-11. 10.1006/clin.1996.4243.

    CAS  PubMed  Google Scholar 

  48. Kohno T, Tsinetoshi Y, Ishikawa E: Existence of antithyroglobulin IgG in healthy subjects. Biochem Biophys Res Commun. 1988, 155: 224-229.

    CAS  PubMed  Google Scholar 

  49. Piechaszuk M, Bouananni M, Salhi SL, Baldet L, Bastide M, Pau B, Bastide JM: Antigenic domains of the human thyroglobulin molecule recognized by autoantibodies in patients' sera and by natural autoantibodies isolated from sera of healthy subjects. Clin Inmunol Immunopathol. 1987, 45: 114-121. 10.1016/0090-1229(87)90117-6.

    Google Scholar 

  50. Dietrich G, Piechaszuk M, Pau B, Kasatchkine MD: Evidence for a restricted idiotypic and epitopic specificity of anti-thyroglobulin autoantibodies in patients with autoimmune thyroiditis. Eur J Immunol. 1991, 21: 811-814.

    CAS  PubMed  Google Scholar 

  51. Taneja V, David CS: HLA transgenic mice as humanized mouse models of disease and immunity. J Clin Invest. 1998, 101: 921-926.

    PubMed Central  CAS  PubMed  Google Scholar 

  52. Kong YM, David CS, Lomo LC, Fuller BE, Motte RW, Giraldo AA: Role of mouse and human class II transgenes in susceptibility to and protection against mouse autoimmune thyroiditis. Immunogenetics. 1997, 46: 312-317. 10.1007/s002510050277.

    CAS  PubMed  Google Scholar 

  53. Dunn JT, Dunn AD: Update on the intrathyroidal iodine metabolism. Thyroid. 2001, 11: 407-414. 10.1089/105072501300176363.

    CAS  PubMed  Google Scholar 

  54. McLachlan SM, Rapoport B: Genetic and epitopic analysis of thyroid peroxidase (TPO) autoantibodies: markers of the human thyroid autoimmune response. Clin Exp Immunol. 1995, 101: 200-206.

    PubMed Central  CAS  PubMed  Google Scholar 

  55. Chardes T, Chapal N, Bresson D, Bes C, Giudicelli V, Lefranc MP, Peraldi-Roux S: The human anti-thyroid peroxidase autoantibody repertoire in Graves' and Hashimoto's autoimmune thyroid diseases. Immunogenetics. 2002, 54: 141-157. 10.1007/s00251-002-0453-9.

    CAS  PubMed  Google Scholar 

  56. Akamizu T: Antithyrotropin receptor antibody: an update. Thyroid. 2001, 11: 1123-1134. 10.1089/10507250152740966.

    CAS  PubMed  Google Scholar 

  57. Heufelder AE, Joba W, Morgenthaler NG: Autoimmunity involving the human sodium/iodide symporter: fact or fiction?. Exp Clin Endocrinol Diabetes. 2001, 109: 35-40. 10.1055/s-2001-11011.

    CAS  PubMed  Google Scholar 

  58. Blucher H, Krohn K, Wallaschofski H, Braverman LE, Paschke R: Cytokine gene expression in autoimmune thyroiditis in BioBreeding/Worcester rats. Thyroid. 1999, 9: 1049-1055.

    Google Scholar 

  59. Mooij P, de Wit HJ, Drexhage HA: An excess of dietary iodine accelerates the development of thyroid-associated lymphoid tissue in autoimmune prone BB rats. Clin Immunol Immunopathol. 1993, 69: 189-198. 10.1006/clin.1993.1169.

    CAS  PubMed  Google Scholar 

  60. Blucher H, Krohn K, Wallaschofski H, Braverman LE, Paschke R: Fas and Fas ligand gene expression in autoimmune thyroiditis in BB/W rats. Eur J Endocrinol. 1999, 141: 506-511. 10.1530/eje.0.1410506.

    Google Scholar 

  61. Salmaso C, Bagnasco M, Pesce G, Montagna P, Brizzolara R, Altrinetti V, Richiusa P, Galluzzo A, Giordano C: Regulation of apoptosis in endocrine autoimmunity: insights from Hashimoto's thyroiditis and Graves' disease. Ann N Y Acad Sci. 2002, 966: 496-501.

    CAS  PubMed  Google Scholar 

  62. Weetman AP: Autoimmune thyroid disease: propagation and progression. Eur J Endocrinol. 2003, 148: 1-9. 10.1530/eje.0.1480001.

    CAS  PubMed  Google Scholar 

  63. Eguchi K: Apoptosis in autoimmune diseases. Intern Med. 2001, 40: 275-284.

    CAS  PubMed  Google Scholar 

  64. Stassi G, De Maria R: Autoimmune thyroid disease: new models of cell death in autoimmunity. Nat Rev Immunol. 2002, 21: 195-204. 10.1038/nri750.

    Google Scholar 

  65. Giordano C, Richiusa P, Bagnasco M, Pizzolanti G, Di Blasi F, Sbriglia MS, Mattina A, Pesce G, Montagna P, Capone F, Misiano G, Scorsone A, Pugliese A, Galluzzo A: Differential regulation of Fas-mediated apoptosis in both thyrocyte and lymphocyte cellular compartments correlates with opposite phenotypic manifestations of autoimmune thyroid disease. Thyroid. 2001, 11: 233-244. 10.1089/105072501750159615.

    CAS  PubMed  Google Scholar 

  66. Limachi F, Basso S: Apoptosis: life trough planned cellular death regulating mechanisms, control systems, and relations with thyroid disease. Thyroid. 2002, 12: 27-34. 10.1089/105072502753451931.

    Google Scholar 

  67. Giordano C, Stassi G, De Maria R, Todaro M., Richiusa P, Papoff G, Ruberti G, Bagnasco M, Testi R, Galluzzo A: Potential involvement of Fas and its ligand in the pathogenesis of Hashimoto's thyroiditis. Science. 1997, 275: 960-963.

    CAS  PubMed  Google Scholar 

  68. Lowin B, Hahne M, Mattmann C, Tschopp J: Cytolytic T-cell cytotoxity is mediated through perforin and Fas lytic pathways. Nature. 1994, 370: 650-652. 10.1038/370650a0.

    CAS  PubMed  Google Scholar 

  69. Arscott PL, Baker JR: Apoptosis and thyroiditis. Clin Immunol Immunopathol. 1998, 87: 207-217. 10.1006/clin.1998.4526.

    CAS  PubMed  Google Scholar 

  70. Zaccone P, Fehervari Z, Cooke A: Tumour necrosis factor-α is a fundamental cytokine in autoimmune thyroid disease induced by thyroglobulin and lypopolysacharide in interleukin-12 p40 deficient C57BL/6 mice. Immunology. 2003, 108: 50-54. 10.1046/j.1365-2567.2003.01547.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  71. Strasser A., O'Connor L, Dixit VM: Apoptosis signalling. Annu Rev Biochem. 2000, 69: 217-245. 10.1146/annurev.biochem.69.1.217.

    CAS  PubMed  Google Scholar 

  72. Bretz JD, Baker JR: Apoptosis and autoimmune thyroid disease: following a TRAIL to thyroid destruction?. Clin Endocrinol. 2001, 55: 1-11. 10.1046/j.1365-2265.2001.01345.x.

    CAS  Google Scholar 

  73. Hall R, Stanbury JB: Familial studies of autoimmune thyroiditis. Clin Exp Immunol. 1967, 2 (Suppl): 719-725.

    PubMed Central  PubMed  Google Scholar 

  74. Tomer Y, Davies TF: Searching for the autoimmune thyroid disease susceptibility genes: from gene mapping to gene function. Endocr Rev. 2003, 24: 694-717. 10.1210/er.2002-0030.

    CAS  PubMed  Google Scholar 

  75. Brix TH, Kyvik KO, Hegedus L: A population-based study of chronic autoimmune hypothyroidism in Danish twins. J Clin Endocrinol Metab. 2000, 85: 536-539. 10.1210/jc.85.2.536.

    CAS  PubMed  Google Scholar 

  76. Ringold DA, Nicoloff JT, Kesler M, Davis H, Hamilton A, Mack T: Further evidence for a strong genetic influence on the development of autoimmune thyroid disease: the California twin study. Thyroid. 2002, 12: 647-653. 10.1089/105072502760258613.

    PubMed  Google Scholar 

  77. Phillips DI, Osmond C, Baird J, Huckle A, Rees-Smith B: Is birthweight associated with thyroid autoimmunity? A study in twins. Thyroid. 2002, 12: 377-380. 10.1089/105072502760043440.

    PubMed  Google Scholar 

  78. Rose NR: Avian models of autoimmune disease: lessons from the birds. Poult Sci. 1994, 73: 984-990.

    CAS  PubMed  Google Scholar 

  79. Lam-Tse WK, Lernmark A, Drexhage HA: Animal models of endocrine/organ-specific autoimmune diseases: do they really help us to understand human autoimmunity?. Springer Semin Immunopathol. 2002, 24: 297-321. 10.1007/s00281-002-0110-2.

    CAS  PubMed  Google Scholar 

  80. Stafford EA, Rose NR: Newer insights into the pathogenesis of experimental autoimmune thyroiditis. Int Rev Immunol. 2000, 19: 501-533.

    CAS  PubMed  Google Scholar 

  81. Braley-Mullen H, Johnson M, Sharp GC, Kuriakos M: Induction of experimental autoimmune thyroiditis in mice with in vitro activated splenic T cells. Cell Immunol. 1985, 93: 132-143. 10.1016/0008-8749(85)90394-6.

    CAS  PubMed  Google Scholar 

  82. Fuller BE, Giraldo AA, Waldmann H, Cobbold SP, Kong YC: Depletion of CD4+ and CD8+ cells eliminates immunologic memory of thyroiditogenicity in murine experimental autoimmune thyroiditis. Autoimmunity. 1994, 19: 161-168.

    CAS  PubMed  Google Scholar 

  83. Caturegli P, Kuppers RC, Mariotti S, Burek CL, Pinchera A, Ladenson PW, Rose NR: IgG subclass distribution of thyroglobulin antibodies in patients with thyroid disease. Clin Exp Immunol. 1994, 98: 464-469.

    PubMed Central  CAS  PubMed  Google Scholar 

  84. Gebe JA, Swanson E, Kwok WW: HLA class II peptide-binding and autoimmunity. Tissue Antigens. 2002, 59: 78-87. 10.1034/j.1399-0039.2002.590202.x.

    CAS  PubMed  Google Scholar 

  85. Kong YM, Lomo LC, Motte RW, Giraldo AA, Baisch J, Strauss G, Hammerling GJ, David CS: HLA-DRB1 polymorphism determines susceptibility to autoimmune thyroiditis in transgenic mice: definitive association with HLA-DRB1*0301 (DR3) gene. J Exp Med. 1996, 184: 1167-1172. 10.1084/jem.184.3.1167.

    CAS  PubMed  Google Scholar 

  86. Flynn JC, Fuller BE, Giraldo AA, Panos JS, David CS, Kong JM: Flexibility of TCR repertoire and permissiveness of HLA-DR3 molecules in experimental autoimmune thyroiditis in nonobese diabetic mice. J Autoimmun. 2001, 17: 7-15. 10.1006/jaut.2001.0528.

    CAS  PubMed  Google Scholar 

  87. Flynn JC, Wan Q, Panos JC, McCormick DJ, Giraldo AA, Davied CS, Kong YM: Coexpression of susceptible and resistant HLA class II transgenes in murine experimental autoimmune thyroiditis: DQ8 molecules downredulate DR3-mediated thyroiditis. J Autoimmun. 2002, 18: 213-220. 10.1006/jaut.2002.0587.

    PubMed  Google Scholar 

  88. Kong YM, David CS: New revelations in susceptibility to autoimmune thyroiditis by the use of H2 and HLA class II transgenic models. Int Rev Immunol. 2000, 19: 573-585.

    CAS  PubMed  Google Scholar 

  89. Marguerie C, Lunardi C, So A: PCR-based analysis of the TCR repertoire in human autoimmune diseases. Immunol Today. 1992, 13: 336-338. 10.1016/0167-5699(92)90166-5.

    CAS  PubMed  Google Scholar 

  90. Matsuoka N, Bernard N, Concepcion ES, Graves PN, Ben-Nun A, Davies TF: T-cell receptor V region beta-chain gene expression in the autoimmune thyroiditis of non-obese diabetic mice. J Immunol. 1993, 151: 1691-1701.

    CAS  PubMed  Google Scholar 

  91. Matsuoka N, Unger P, Ben-Nun A, Graves P, Davies TF: Thyroglobulin-induced murine thyroiditis assessed by intrathyroidal T cell receptor sequencing. J Immunol. 1994, 152: 2562-2568.

    CAS  PubMed  Google Scholar 

  92. Quaratino S, Badami E, Pang YY, Bartok I, Dyson J, Kioussis D, Londei M, Maiuri L: Degenerate self-reactive human T-cell receptor causes spontaneous autoimmune disease in mice. Nat Med. 2004, 10: 920-926. 10.1038/nm1092.

    CAS  PubMed  Google Scholar 

  93. Quaratino S, Feldmann M, Dayan CM, Acuto O, Londei M: Human self-reactive T cell clones expressing identical T cell receptor β chains differ in their ability to recognize a cryptic self-epitope. J Exp Med. 1996, 183: 349-358. 10.1084/jem.183.2.349.

    CAS  PubMed  Google Scholar 

  94. Salomon B, Bluestone JA: Complexes of CD28/B7-CTLA-4 costimulatory pathways in autoimmunity and transplantation. Annu Rev Immunol. 2001, 19: 225-252. 10.1146/annurev.immunol.19.1.225.

    CAS  PubMed  Google Scholar 

  95. Greenwald RJ, Boussotis VA, Lorsbach RB, Abbas AK, Sharpe AH: CTLA-4 regulates induction of anergy in vivo. Immunity. 2001, 14: 145-155.

    CAS  PubMed  Google Scholar 

  96. Eagar TN, Karandikar NJ, Bluestone JA, Miller SD: The role of CTLA-4 in induction and maintaince of peripheral T cell tolerance. Eur J Immunol. 2002, 32: 972-981. 10.1002/1521-4141(200204)32:4<972::AID-IMMU972>3.3.CO;2-D.

    CAS  PubMed  Google Scholar 

  97. Carreno BM, Bennett F, Chau TA, Ling V, Luxenberg D, Jussif J, Baroja ML, Madrenas J: CTLA-4 (CD152) can inhibit T cell activation by two different mechanisms depending on its level of cell surface expression. J Immunol. 2000, 165: 1352-1356.

    CAS  PubMed  Google Scholar 

  98. Prud'homme GJ, Chang Y, Li X: Immunoinhibitory DNA vaccine protects against autoimmune diabetes through cDNA encoding a selective CTLA-4 (CD152) ligand. Hum Gene Ther. 2002, 13: 395-406. 10.1089/10430340252792521.

    PubMed  Google Scholar 

  99. Campbell RD, Trowsdale J: Map of human MHC. Immunol Today. 1993, 14: 349-352. 10.1016/0167-5699(93)90234-C.

    CAS  PubMed  Google Scholar 

  100. Sakurami T, Ueno Y, Iwaki Y, Park MS, Terasaki PI, Saji H: HLA-DR specificities among Japanese with several autoimmune diseases. Tissue Antigens. 1982, 19: 129-133.

    CAS  PubMed  Google Scholar 

  101. Hawkins BR, Lam KS, Ma JT, Wang CC, Yeung RT: Strong association between HLA DRw9 and Hashimoto's thyroiditis in Southern Chinese. Acta Endocrinol. 1987, 114: 543-546.

    CAS  PubMed  Google Scholar 

  102. Ito M, Tanimoto M, Kamura H, Yoneda M, Morishima Y, Yamauchi K, Itatsu T, Takatsuki K, Saito H: Association of HLA antigen and restriction fragment length polymorphism of T cell receptor beta-chain gene with Graves' disease and Hashimoto's thyroiditis. J Clin Endocrinol Metab. 1989, 69: 100-104.

    CAS  PubMed  Google Scholar 

  103. Inoue D, Sato K, Maeda M, Inoko H, Tsuji K, Mori T, Imura H: Genetic differences shown by HLA typing among Japanese patients with euthyroid Graves' ophthalmopathy, Graves' disease and Hashimoto's thyroiditis: genetic characteristics of euthyroid Graves' ophthalmopathy. Clin Endocrinol (Oxf). 1991, 34: 57-62.

    CAS  Google Scholar 

  104. Inoue D, Sato K, Sugawa H, Akamizu T, Maeda M, Inoko H, Tsuji K, Mori T: Apparent genetic difference between hypothyroid patients with blocking-type thyrotropin receptor antibody and those without, as shown by restriction fragment length polymorphism analyses of HLA-DP loci. J Clin Endocrinol Metab. 1993, 77: 606-610. 10.1210/jc.77.3.606.

    CAS  PubMed  Google Scholar 

  105. Onuma H, Ota M, Sugenoya A, Fukushima H, Inoko H, Iida F: Association of HLA-DR53 and lack of association of DPB1 alleles with Hashimoto's thyroiditis in Japanese. Tissue Antigens. 1993, 42: 150-152.

    CAS  PubMed  Google Scholar 

  106. Moens H, Farid NR, Sampson L, Noel EP, Barnard JM: Hashimoto's thyroiditis is associated with HLA-DRw3. N Engl J Med. 1978, 299: 133-134.

    CAS  PubMed  Google Scholar 

  107. Weissel M, Hofer R, Zasmeta H, Mayr WR: HLA-DR and Hashimoto's thyroiditis. Tissue Antigens. 1980, 16: 256-257.

    CAS  PubMed  Google Scholar 

  108. Farid NR, Sampson L, Moens H, Barnard JM: The association of goitrous autoimmune thyroiditis with HLA-DR5. Tissue Antigens. 1981, 17: 265-268.

    CAS  PubMed  Google Scholar 

  109. Thomsen M, Ryder LP, Bech K, Bliddal H, Feldt-Rasmussen U, Molholm J, Kappelgaard E, Nielsen H, Svejgaard A: HLA-D in Hashimoto's thyroiditis. Tissue Antigens. 1983, 21: 173-175.

    CAS  PubMed  Google Scholar 

  110. Tandon N, Zhang L, Weetman AP: HLA associations with Hashimoto's thyroiditis. Clin Endocrinol (Oxf). 1991, 34: 383-386.

    CAS  Google Scholar 

  111. Stenszky V, Balazs C, Kraszits E, Juhasz F, Kozma L, Balazs G, Farid NR: Association of goitrous autoimmune thyroiditis with HLA-DR3 in eastern Hungary. J Immunogenet. 1987, 14: 143-148.

    CAS  PubMed  Google Scholar 

  112. Badenhoop K, Schwarz G, Walfish PG, Drummond V, Usadel KH, Bottazzo GF: Susceptibility to thyroid autoimmune disease: molecular analysis of HLA-D region genes identifies new markers for goitrous Hashimoto's thyroiditis. J Clin Endocrinol Metab. 1990, 71: 1131-1137.

    CAS  PubMed  Google Scholar 

  113. Mangklabruks A, Cox N, DeGroot LJ: Genetic factors in autoimmune thyroid disease analyzed by restriction fragment length polymorphisms of candidate genes. J Clin Endocrinol Metab. 1991, 73: 236-244.

    CAS  PubMed  Google Scholar 

  114. Bogner U, Badenhoop K, Peters H, Schmieg D, Mayr WR, Usadel KH, Schleusener H: HLA-DR/DQ gene variation in nongoitrous autoimmune thyroiditis at the serological and molecular level. Autoimmunity. 1992, 14: 155-158.

    CAS  PubMed  Google Scholar 

  115. Shi Y, Zou M, Robb D, Farid NR: Typing for major histocompatibility complex class II antigens in thyroid tissue blocks: association of Hashimoto's thyroiditis with HLA-DQA0301 and DQB0201 alleles. J Clin Endocrinol Metab. 1992, 75: 943-946. 10.1210/jc.75.3.943.

    CAS  PubMed  Google Scholar 

  116. Nicholson LB, Wong FS, Ewins DL, Butler J, Holland A, Demaine AG, McGregor AM: Susceptibility to autoimmune thyroiditis in Down's syndrome is associated with the major histocompatibility class II DQA 0301 allele. Clin Endocrinol (Oxf). 1994, 41: 381-383.

    CAS  Google Scholar 

  117. Barbesino G, Chiovato L: The genetics of Hashimoto's disease. Endocrinol Metab Clin North Am. 2000, 29: 357-374.

    CAS  PubMed  Google Scholar 

  118. Hunt PJ, Marshall SE, Weetman AP, Bunce M, Bell JI, Wass JA, Welsh KI: Histocompatibility leucocyte antigens and closely linked immunomodulatory genes in autoimmune thyroid disease. Clin Endocrinol. 2001, 55: 491-499. 10.1046/j.1365-2265.2001.01356.x.

    CAS  Google Scholar 

  119. Petrone A, Giorgi G, Mesturino CA, Capizzi M, Cascino I, Nistico L, Osborn J, Di Mario U, Buzzetti R: Association of DRB1*04-DQB1*0301 haplotype and lack of association of two polymorphic sites at CTLA-4 gene with Hashimoto's thyroiditis in an Italian population. Thyroid. 2001, 11: 171-175. 10.1089/105072501300042901.

    CAS  PubMed  Google Scholar 

  120. Ban Y, Davies TF, Greenberg DA, Concepcion ES, Tomer Y: The influence of human leucocyte antigen (HLA) genes on autoimmune thyroid disease (AITD): results of studies in HLA-DR3 positive AITD families. Clin Endocrinol. 2002, 57: 81-88. 10.1046/j.1365-2265.2002.01566.x.

    CAS  Google Scholar 

  121. Wu Z, Stephens HA, Sachs JA, Biro PA, Cutbush S, Magzoub MM, Becker C, Schwartz G, Bottazzo GF: Molecular analysis of HLA-DQ and -DP genes in Caucasoid patients with Hashimoto's thyroiditis. Tissue Antigens. 1994, 43: 116-119.

    CAS  PubMed  Google Scholar 

  122. Jenkins D, Penny MA, Fletcher JA, Jacobs KH, Mijovic CH, Franklyn JA, Sheppard MC: HLA class II gene polymorphism contributes little to Hashimoto's thyroiditis. Clin Endocrinol (Oxf). 1992, 37: 141-145.

    CAS  Google Scholar 

  123. Badenhoop K, Walfish PG, Rau H, Fischer S, Nicolay A, Bogner U, Schleusener H, Usadel KH: Susceptibility and resistance alleles of human leukocyte antigen (HLA) DQA1 and HLA DQB1 are shared in endocrine autoimmune disease. J Clin Endocrinol Metab. 1995, 80: 2112-2117. 10.1210/jc.80.7.2112.

    CAS  PubMed  Google Scholar 

  124. Bode HH, Dorf ME, Forbes AP: Familial lympholytic thyroiditis: analysis of linkage with hitocompatibility and blood groups. J Clin Endocrinol Metab. 1973, 37: 692-697.

    CAS  PubMed  Google Scholar 

  125. Hawkins BR, Ma JT, Lam KS, Wang CC, Yeung RT: Analysis of linkage between HLA haplotype and susceptibility to Graves' disease in multiple-case Chinese families in Hong Kong. Acta Endocrinol. 1985, 110: 66-69.

    CAS  PubMed  Google Scholar 

  126. Roman SH, Greenberg D, Rubinstein P, Wallenstein S, Davies TF: Genetics of autoimmune thyroid disease: lack of evidence for linkage to HLA within families. J Clin Endocrinol Metab. 1992, 74: 496-503. 10.1210/jc.74.3.496.

    CAS  PubMed  Google Scholar 

  127. O'Connor G, Neufeld DS, Greenberg DA, Concepcion ES, Roman SH, Davies TF: Autoimmunity. 1993, 14: 237-241.

    PubMed  Google Scholar 

  128. Tomer Y, Barbesino G, Keddache M, Greenberg DA, Davies TF: Mapping of a major susceptibility locus for Graves' disease (GD-1) to chromosome 14q31. J Clin Endocrinol Metab. 1997, 82: 1645-1648. 10.1210/jc.82.5.1645.

    CAS  PubMed  Google Scholar 

  129. Barbesino G, Tomer Y, Concepcion ES, Davies TF, Greenberg DA: 1. Selected immunoregulatory genes. International Consortium for the Genetics of Autoimmune Thyroid Disease. J Clin Endocrinol Metab. 1998, 83: 1580-1584. 10.1210/jc.83.5.1580.

    CAS  PubMed  Google Scholar 

  130. Tomer Y, Barbesino G, Greenberg DA, Concepcion E, Davies TF: Mapping the major susceptibility loci for familial Graves' and Hashimoto's diseases: evidence for genetic heterogeneity and gene interactions. J Clin Endocrinol Metab. 1999, 84: 4656-4664. 10.1210/jc.84.12.4656.

    CAS  PubMed  Google Scholar 

  131. Tomer Y, Ban Y, Concepcion E, Barbesino G, Villanueva R, Greenberg DA, Davies TF: Common and unique susceptibility loci in Graves and Hashimoto diseases: results of whole-genome screening in a data set of 102 multiplex families. Am J Hum Genet. 2003, 73: 736-747. 10.1086/378588.

    PubMed Central  CAS  PubMed  Google Scholar 

  132. Alkhateeb A, Stetler GL, Old W, Talbert J, Uhlhorn C, Taylor M, Fox A, Miller C, Dills DG, Ridgway EC, Bennett DC, Fain PR, Spritz RA: Mapping of an autoimmunity susceptibility locus (AIS1) to chromosome 1p31.3-p32.2. Hum Mol Genet. 2002, 11: 661-667. 10.1093/hmg/11.6.661.

    CAS  PubMed  Google Scholar 

  133. Maalej A, Makni H, Ayadi F, Bellassoued M, Jouida J, Bouguacha N, Abid M, Ayadi H: A full genome screening in a large Tunisian family affected with thyroid autoimmune disorders. Genes Immun. 2001, 2: 71-75. 10.1038/sj.gene.6363736.

    CAS  PubMed  Google Scholar 

  134. Villanueva R, Tomer Y, Greenberg DA, Mao C, Concepcion ES, Tucci S, Estilo G, Davies TF: Autoimmune thyroid disease susceptibility loci in a large Chinese family. Clin Endocrinol. 2002, 56: 45-51. 10.1046/j0300-0664.2001.01429.x.

    CAS  Google Scholar 

  135. Sakai K, Shirasawa S, Ishikawa N, Ito K, Tamai H, Kuma K, Akamizu T, Tanimura M, Furugaki K, Yamamoto K, Sasazuki T: Identification of susceptibility loci for autoimmune thyroid disease to 5q31-q33 and Hashimoto's thyroiditis to 8q23-q24 by multipoint affected sib-pair linkage analysis in Japanese. Hum Mol Genet. 2001, 10: 1379-1386. 10.1093/hmg/10.13.1379.

    CAS  PubMed  Google Scholar 

  136. Levin L, Ban Y, Concepcion E, Davies TF, Greenberg DA, Tomer Y: Analysis of HLA genes in families with autoimmune diabetes and thyroiditis. Hum Immunol. 2004, 65: 640-647. 10.1016/j.humimm.2004.02.026.

    CAS  PubMed  Google Scholar 

  137. Torfs CP, King MC, Huey B, Malmgren J, Grumet FC: Genetic interrelationship between insulin-dependent diabetes mellitus, the autoimmune thyroid diseases, and rheumatoid arthritis. Am J Hum Genet. 1986, 38: 170-187.

    PubMed Central  CAS  PubMed  Google Scholar 

  138. Payami H, Joe S, Thomson G: Autoimmune thyroid disease in type 1 diabetes. Genet Epidemiol. 1989, 6: 137-141.

    CAS  PubMed  Google Scholar 

  139. Allen DB, MacDonald MJ, Gottschall JL, Hunter JB: Autoimmune thyroid phenomena are not evidence for human lymphocyte antigen-genetic heterogeneity in insulin-dependent diabetes. Am J Med Genet. 1989, 33: 405-408.

    CAS  PubMed  Google Scholar 

  140. Djilali-Saiah I, Bertin E, Larger E, Timsit J, Assan R, Boitard C, Bach JF, Caillat-Zucman S: Major histocompatibility class II genes polymorphism in insulin dependent diabetes mellitus with or without associated thyroid autoimmunity. Hum Immunol. 1998, 59: 176-182. 10.1016/S0198-8859(98)00002-0.

    CAS  PubMed  Google Scholar 

  141. Fischer GF: Molecular genetics of HLA. Vox Sanguinis. 2000, 78: 261-264. 10.1159/000031194.

    CAS  PubMed  Google Scholar 

  142. Faas S, Trucco M: The genes influencing the susceptibility to IDDM in humans. J Endocrinol Invest. 1994, 17: 477-495.

    CAS  PubMed  Google Scholar 

  143. Sawai Y, DeGroot LJ: Binding of human thyrotropin receptor peptides to a Graves' disease-predisposing human leukocyte antigen class II molecule. J Clin Endocrinol Metab. 2000, 85: 1176-1179. 10.1210/jc.85.3.1176.

    CAS  PubMed  Google Scholar 

  144. Dawe KI, Hutchings PR, Geysen M, Champion BR, Cooke A, Roitt IM: Unique role of thyroxine in T cell recognition of a pathogenic peptide in experimental autoimmune thyroiditis. Int Rev Immunol. 2000, 19: 573-585.

    Google Scholar 

  145. Kuhr T, Hala K, Dietrich H, Herold M, Wick G: Genetically determined target organ susceptibility in the pathogenesis of spontaneous autoimmune thyroiditis: aberrant expression of MHC-class II antigens and the possible role of virus. J Autoimmun. 1994, 7: 13-25. 10.1006/jaut.1994.1002.

    CAS  PubMed  Google Scholar 

  146. Kong JM: Graves' hyperthyroidism and thyroiditis in HLA-DRB1*0301 (DR3) transgenic mice after immunization with thyrotropin receptor DNA. Clin Exp Immunol. 2004, 135: 35-40. 10.1111/j.1365-2249.2004.02333.x.

    PubMed Central  PubMed  Google Scholar 

  147. Londei M, Lamb JR, Bottazzo GF, Feldmann M: Epithelial cells expressing aberrant MHC class II determinants can present antigen to cloned human T cells. Nature. 1984, 312: 639-641. 10.1038/312639a0.

    CAS  PubMed  Google Scholar 

  148. Hamilton F, Black M, Farquharson MA, Stewart C, Foulis AK: Spatial correlation between thyroid epithelial cells expressing class II MHC molecules and interferon-gamma-containing lymphocytes in human thyroid autoimmune disease. Clin Exp Immunol. 1991, 83: 64-68.

    PubMed Central  CAS  PubMed  Google Scholar 

  149. Shimojo N, Arima T, Yamaguchi K, Kikuoka S, Kohn LD, Kohno Y: A novel mouse model of Graves' disease: implications for a role of aberrant MHC class II expression in its pathogenesis. Int Rev Immunol. 2000, 19: 619-631.

    CAS  PubMed  Google Scholar 

  150. Platzer M, Neufeld DS, Piccinini LA, Davies TF: Induction of rat thyroid cell MHC class II antigen by thyrotropin and gamma-interferon. Endocrinology. 1987, 121: 2087-2092.

    CAS  PubMed  Google Scholar 

  151. Nagataki S, Eguchi K: Cytokines and immune regulation in thyroid autoimmunity. Autoimmunity. 1992, 13: 27-34.

    CAS  PubMed  Google Scholar 

  152. Todd JA: Genetic analysis of type 1 diabetes using whole genome approaches. Proc Natl Acad Sci USA. 1995, 92: 8560-8565.

    PubMed Central  CAS  PubMed  Google Scholar 

  153. Lewin L, Tomer Y: The etiology of autoimmune diabetes and thyroiditis: evidence for common genetic susceptibility. Autoimmun Rev. 2003, 2: 377-386. 10.1016/S1568-9972(03)00080-6.

    Google Scholar 

  154. Makhatadze NJ: Tumor necrosis factor locus: genetic organisation and biological implications. Hum Immunol. 1998, 59: 571-579. 10.1016/S0198-8859(98)00056-1.

    CAS  PubMed  Google Scholar 

  155. Milner CM, Campbell RD: Genetic organization of the human MHC class III region. Front Biosci. 2001, 6: D914-926.

    CAS  PubMed  Google Scholar 

  156. Weetman AP, Ajjan RA, Watson PF: Cytokines and Graves disease. Baillieres Clin Endocrinol Metab. 1997, 11: 481-497. 10.1016/S0950-351X(97)80708-2.

    CAS  PubMed  Google Scholar 

  157. Chung JH, Cho BY, Lee HK, Kim TG, Han H, Koh CS: The tumor necrosis factor beta * 1 allele is linked significantly to HLA-DR8 in Koreans with atrophic autoimmune thyroiditis who are positive for thyrotropin receptor blocking antibody. J Korean Med Sci. 1994, 9: 155-161.

    PubMed Central  CAS  PubMed  Google Scholar 

  158. De Nagel DC, Pierce SK: Heat shock proteins implicated in antigen processing and presentation. Semin Immunol. 1991, 3: 65-71.

    CAS  PubMed  Google Scholar 

  159. Parkes AB, Darke C, Othman S, Thomas M, Young N, Richards CJ, Hall R, Lazarus JH: Major histocompatibility complex class II and complement polymorphisms in postpartum thyroiditis. Eur J Endocrinol. 1996, 134: 449-453.

    CAS  PubMed  Google Scholar 

  160. Fu Y, Yan G, Shi L, Faustman D: Antigen processing and autoimmunity. Evaluation of mRNA abundance and function of HLA-linked genes. Ann N Y Acad Sci. 1998, 842: 138-155.

    CAS  PubMed  Google Scholar 

  161. Heward JM, Allahabadia A, Sheppard MC, Barnett AH, Franklyn JA, Gough SC: Association of the large multifunctional proteasome (LMP2) gene with Graves' disease is a result of linkage disequilibrium with the HLA haplotype DRB1*0304-DQB1*02-DQA1*0501. Clin Endocrinol. 1999, 51: 115-118. 10.1046/j.1365-2265.1999.00755.x.

    CAS  Google Scholar 

  162. Fling SP, Arp B, Pious D: HLA-DMA and -DMB genes are both required for MHC class II/peptide complex formation in antigen-presenting cells. Nature. 1994, 368: 554-558. 10.1038/368554a0.

    CAS  PubMed  Google Scholar 

  163. Kim TG, Carrington M, Choi HB, Kim HY, Han H: Three HLA-DMB variants in Korean patients with autoimmune diseases. Hum Immunol. 1996, 46: 58-60. 10.1016/0198-8859(95)00171-9.

    CAS  PubMed  Google Scholar 

  164. Ostrov DA, Shi W, Schwartz JC, Almo SC, Nathenson SG: Structure of murine CTLA-4 and its role in modulating T cell responsiveness. Science. 2000, 290: 816-819. 10.1126/science.290.5492.816.

    CAS  PubMed  Google Scholar 

  165. Chistiakov DA, Turakulov RI: CTLA4 and its role in autoimmune thyroid disease. J Mol Endocrinol. 2003, 31: 21-36. 10.1677/jme.0.0310021.

    CAS  PubMed  Google Scholar 

  166. Vaidya B, Imrie H, Perros P, Young ET, Kelly WF, Carr D, Large DM, Toft AD, McCarthy MI, Kendall-Taylor P, Pearce SH: The cytotoxic T lymphocyte antigen-4 is a major Graves' disease locus. Hum Mol Genet. 1999, 8: 1195-1199. 10.1093/hmg/8.7.1195.

    CAS  PubMed  Google Scholar 

  167. Tomer Y, Greenberg DA, Barbesino G, Concepcion E, Davies TF: CTLA-4 and not CD28 is a susceptibility gene for thyroid autoantibody production. J Clin Endocrinol Metab. 2001, 86: 1687-1693. 10.1210/jc.86.4.1687.

    CAS  PubMed  Google Scholar 

  168. Allahabadia A, Gough SC: The different approaches to the genetic analysis of autoimmune thyroid disease. J Endocrinol. 1999, 163: 7-13. 10.1677/joe.0.1630007.

    CAS  PubMed  Google Scholar 

  169. Ueda H, Howson JM, Esposito L, Heward J, Snook H, Chamberlain G, Rainbow DB, Hunter KM, Smith AN, Di Genova G, Herr MH, Dahlman I, Payne F, Smyth D, Lowe C, Twells RC, Howlett S, Healy B, Nutland S, Rance HE, Everett V, Smink LJ, Lam AC, Cordell HJ, Walker NM, Bordin C, Hulme J, Motzo C, Cucca F, Hess JF, Metzker ML, Rogers J, Gregory S, Allahabadia A, Nithiyananthan R, Tuomilehto-Wolf E, Tuomilehto J, Bingley P, Gillespie KM, Undlien DE, Ronningen KS, Guja C, Ionescu-Tirgoviste C, Savage DA, Maxwell AP, Carson DJ, Patterson CC, Franklyn JA, Clayton DG, Peterson LB, Wicker LS, Todd JA, Gough SC: Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature. 2003, 423: 506-511. 10.1038/nature01621.

    CAS  PubMed  Google Scholar 

  170. Magistrelli G, Jeannin P, Herbault N, Benoit De Coignac A, Gauchat JF, Bonnefoy JY, Delneste Y: A soluble form of CTLA-4 generated by alternative splicing is expressed by nonstimulated human T cells. Eur J Immunol. 1999, 29: 3596-3602. 10.1002/(SICI)1521-4141(199911)29:11<3596::AID-IMMU3596>3.3.CO;2-P.

    CAS  PubMed  Google Scholar 

  171. Oaks MK, Hallett KM, Penwell RT, Stauber EC, Warren SJ, Tector AJ: A native soluble form of CTLA-4. Cell Immunol. 2000, 201: 144-153. 10.1006/cimm.2000.1649.

    CAS  PubMed  Google Scholar 

  172. Manzotti CN, Tipping H, Perry LC, Mead KI, Blair PJ, Zheng Y, Sansom DM: Inhibition of human T cell proliferation by CTLA-4 utilizes CD80 and requires CD25+ regulatory T cells. Eur J Immunol. 2002, 32: 2888-2896. 10.1002/1521-4141(2002010)32:10<2888::AID-IMMU2888>3.0.CO;2-F.

    CAS  PubMed  Google Scholar 

  173. Oaks MK, Hallett KM: Cutting edge: a soluble form of CTLA-4 in patients with autoimmune thyroid disease. J Immunol. 2000, 164: 5015-5018.

    CAS  PubMed  Google Scholar 

  174. Wang XB, Kakoulidou M, Giscombe R, Qiu Q, Huang D, Pirskanen R, Lefvert AK: Abnormal expression of CTLA-4 by T cells from patients with myasthenia gravis: effect of an AT-rich gene sequence. J Neuroimmunol. 2002, 130: 224-232. 10.1016/S0165-5728(02)00228-X.

    CAS  PubMed  Google Scholar 

  175. Huang D, Liu L, Noren K, Xia SQ, Trifunovic J, Pirskanen R, Lefvert AK: Genetic association of Ctla-4 to myasthenia gravis with thymoma. J Neuroimmunol. 1998, 88: 192-198. 10.1016/S0165-5728(98)00119-2.

    CAS  PubMed  Google Scholar 

  176. Anjos S, Nguyen H, Ounissi-Benkalha H, Tessier MC, Polychronakos C: A common autoimmunity predisposing signal peptide variant of the cytotoxic T-lymphocyte antigen 4 results in inefficient glycosylation of the susceptibility allele. J Biol Chem. 2002, 277: 46478-46486. 10.1074/jbc.M206894200.

    CAS  PubMed  Google Scholar 

  177. Kouki T, Sawai Y, Gardine AC, Fisfalen ME, Alegre ML, DeGroot LJ: CTLA-4 gene polymorphism at position 49 in exon 1 reduces the inhibitory function of CTLA-4 and contributes to the pathogenesis of Graves disease. J Immunol. 2000, 165: 6606-6611.

    CAS  PubMed  Google Scholar 

  178. Maurer M, Loserth S, Kolb-Maurer A, Ponath A, Wiese S, Kruse N, Rieckmann P: A polymorphism in the human cytotoxic T-lymphocyte antigen 4 (CTLA4) gene (exon 1 +49) alters T-cell activation. Immunogenetics. 2002, 54: 1-8. 10.1007/s00251-002-0429-9.

    PubMed  Google Scholar 

  179. Holopainen PM, Partanen JA: Technical note: linkage disequilibrium and disease-associated CTLA4 gene polymorphisms. J Immunol. 2001, 167: 2457-2458.

    CAS  PubMed  Google Scholar 

  180. Kouki T, Gardine CA, Yanagawa T, Degroot LJ: Relation of three polymorphisms of the CTLA-4 gene in patients with Graves' disease. J Endocrinol Invest. 2002, 25: 208-213.

    CAS  PubMed  Google Scholar 

  181. Ban Y, Davies TF, Greenberg DA, Kissin A, Marder B, Murphy B, Concepcion ES, Villanueva RB, Barbesino G, Ling V, Tomer Y: Analysis of the CTLA-4, CD28, and inducible costimulator (ICOS) genes in autoimmune thyroid disease. Genes Immun. 2003, 4: 586-593. 10.1038/sj.gene.6364018.

    CAS  PubMed  Google Scholar 

  182. Wang XB, Zhao X, Giscombe R, Lefvert AK: A CTLA-4 gene polymorphism at position -318 in the promoter region affects the expression of protein. Genes Immun. 2002, 3: 233-234. 10.1038/sj.gene.6363869.

    PubMed  Google Scholar 

  183. Chen CY, Shyu AB: AU-rich elements: characterization and importance in mRNA degradation. Trends Biochem Sci. 1995, 20: 465-470. 10.1016/S0968-0004(00)89102-1.

    CAS  PubMed  Google Scholar 

  184. Thomas M, Karlen S, D'Ercole , Sanderson C: Analysis of the 5' and 3' UTRs in the posttranslational regulation of the interleukin-5 gene. Biochim Biophys Acta. 1999, 1444: 61-68.

    CAS  PubMed  Google Scholar 

  185. Huang D, Giscombe R, Zhou Y, Pirskanen R, Lefvert AK: Dinucleotide repeat expansion in the CTLA-4 gene leads to T-cell hyperreactivity via the CD28 pathway in myasthenia gravis. J Neuroimmunol. 2000, 105: 69-77. 10.1016/S0165-5728(00)00191-0.

    CAS  PubMed  Google Scholar 

  186. Takara M, Kouki T, DeGroot LJ: CTLA-4 AT-repeat polymorphism reduces the inhibitory function of CTLA-4 in Graves' disease. Thyroid. 2003, 13: 1083-1089. 10.1089/10507250360731479.

    CAS  PubMed  Google Scholar 

  187. Anjos S, Polychronakos C: Mechanisms of genetic susceptibility to type 1 diabetes: beyond HLA. Mol Genet Metab. 2004, 81: 187-195. 10.1016/j.ymgme.2003.11.010.

    CAS  PubMed  Google Scholar 

  188. Johnson GC, Esposito L, Barratt BJ, Smith AN, Heward J, Di Genova G, Ueda H, Cordell HJ, Eaves IA, Dudbridge F, Twells RC, Payne F, Hughes W, Nutland S, Stevens H, Carr P, Tuomilehto-Wolf E, Tuomilehto J, Gough SC, Clayton DG, Todd JA: Haplotype tagging for the identification of common disease genes. Nat Genet. 2001, 29: 233-237. 10.1038/ng1001-233.

    CAS  PubMed  Google Scholar 

  189. Ling V, Wu PW, Finnerty HF, Agostino MJ, Graham JR, Chen S, Jussiff JM, Fisk GJ, Miller CP, Collins M: Assembly and annotation of human chromosome 2q33 sequence containing the CD28, CTLA4, and ICOS gene cluster: analysis by computational, comparative, and microarray approaches. Genomics. 2001, 78: 155-168. 10.1006/geno.2001.6655.

    CAS  PubMed  Google Scholar 

  190. Zaletel K, Krhin B, Gaberscek S, Pirnat E, Hojker S: The influence of the exon 1 polymorphism of the cytotoxic T lymphocyte antigen 4 gene on thyroid antibody production in patients with newly diagnosed Graves' disease. Thyroid. 2002, 12: 373-376. 10.1089/105072502760043431.

    CAS  PubMed  Google Scholar 

  191. Vaidya B, Imrie H, Geatch DR, Perros P, Ball SG, Baylis PH, Carr D, Hurel SJ, James RA, Kelly WF, Kemp EH, Young ET, Weetman AP, Kendall-Taylor P, Pearce SH: Association analysis of the cytotoxic T lymphocyte antigen-4 (CTLA-4) and autoimmune regulator-1 (AIRE-1) genes in sporadic autoimmune Addison's disease. J Clin Endocrinol Metab. 2000, 85: 688-691. 10.1210/jc.85.2.688.

    CAS  PubMed  Google Scholar 

  192. Kristiansen OP, Larsen ZM, Pociot F: CTLA-4 in autoimmune diseases – a general susceptibility gene to autoimmunity?. Genes Immun. 2000, 1: 170-184. 10.1038/sj.gene.6363655.

    CAS  PubMed  Google Scholar 

  193. Harbo HF, Celius EG, Vartdal F, Spurkland A: CTLA4 promoter and exon 1 dimorphisms in multiple sclerosis. Tissue Antigens. 1999, 53: 106-110. 10.1034/j.1399-0039.1999.530112.x.

    CAS  PubMed  Google Scholar 

  194. Ligers A, Xu C, Saarinen S, Hillert J, Olerup O: The CTLA-4 gene is associated with multiple sclerosis. J Neuroimmunol. 1999, 97: 182-190. 10.1016/S0165-5728(99)00072-7.

    CAS  PubMed  Google Scholar 

  195. Robles DT, Fain PR, Gottlieb PA, Eisenbarth GS: The genetics of autoimmune polyendocrine syndrome type II. Endocrinol Metab Clin North Am. 2002, 31: 353-368.

    PubMed  Google Scholar 

  196. Fakhfakh F, Maalej A, Makni H, Abid M, Jouida J, Zouali M, Ayadi H: Analysis of immunoglobulin VH and TCR beta polymorphisms in a large family with thyroid autoimmune disorder. Exp Clin Immunogenet. 1999, 16: 185-191. 10.1159/000019110.

    CAS  PubMed  Google Scholar 

  197. Nakao Y, Matsumoto H, Miyazaki T, Nishitani H, Takatsuki K, Kasukawa R, Nakayama S, Izumi S, Fujita T, Tsuji K: IgG heavy chain allotypes (Gm) in autoimmune diseases. Clin Exp Immunol. 1980, 42: 20-26.

    PubMed Central  CAS  PubMed  Google Scholar 

  198. Uno H, Sasazuki T, Tamai H, Matsumoto H: Two major genes, linked to HLA and Gm, control susceptibility to Graves' disease. Nature. 1981, 292: 768-770. 10.1038/292768a0.

    CAS  PubMed  Google Scholar 

  199. Rasmussen AK: Cytokine actions on the thyroid gland. Dan Med Bull. 2000, 47: 94-114.

    CAS  PubMed  Google Scholar 

  200. Ajjan RA, Watson PF, Weetman AP: Cytokines and thyroid function. Adv Neuroimmunol. 1996, 6: 359-386.

    CAS  PubMed  Google Scholar 

  201. Hunt PJ, Marshall SE, Weetman AP, Bell JI, Wass JA, Welsh KI: Cytokine gene polymorphisms in autoimmune thyroid disease. J Clin Endocrinol Metab. 2000, 85: 1984-1988. 10.1210/jc.85.5.1984.

    CAS  PubMed  Google Scholar 

  202. Blakemore AI, Watson PF, Weetman AP, Duff GW: Association of Graves' disease with an allele of the interleukin-1 receptor antagonist gene. J Clin Endocrinol Metab. 1995, 80: 111-115. 10.1210/jc.80.1.111.

    CAS  PubMed  Google Scholar 

  203. Pearce SH, Cheetham T, Imrie H, Vaidya B, Barnes ND, Bilous RW, Carr D, Meeran K, Shaw NJ, Smith CS, Toft AD, Williams G, Kendall-Taylor P: A common and recurrent 13-bp deletion in the autoimmune regulator gene in British kindreds with autoimmune polyendocrinopathy type 1. Am J Hum Genet. 1998, 63: 1675-1684. 10.1086/302145.

    PubMed Central  CAS  PubMed  Google Scholar 

  204. Nithiyananthan R, Heward JM, Allahabadia A, Barnett AH, Franklyn JA, Gough SC: A heterozygous deletion of the autoimmune regulator (AIRE1) gene, autoimmune thyroid disease, and type 1 diabetes: no evidence for association. J Clin Endocrinol Metab. 2000, 85: 1320-1322. 10.1210/jc.85.3.1320.

    CAS  PubMed  Google Scholar 

  205. Fournier C, Gepner P, Sadouk M, Charreire J: In vivo beneficial effects of cyclosporin A and 1,25-dihydroxyvitamin D3 on the induction of experimental autoimmune thyroiditis. Clin Immunol Immunopathol. 1990, 54: 53-63. 10.1016/0090-1229(90)90005-B.

    CAS  PubMed  Google Scholar 

  206. Hahn HJ, Kuttler B, Mathieu C, Bouillon R: 1,25-Dihydroxyvitamin D3 reduces MHC antigen expression on pancreatic beta-cells in vitro. Transplant Proc. 1997, 29: 2156-2157. 10.1016/S0041-1345(97)00273-X.

    CAS  PubMed  Google Scholar 

  207. Pani MA, Regulla K, Segni M, Krause M, Hofmann S, Hufner M, Herwig J, Pasquino AM, Usadel KH, Badenhoop K: Vitamin D 1alpha-hydroxylase (CYP1alpha) polymorphism in Graves' disease, Hashimoto's thyroiditis and type 1 diabetes mellitus. Eur J Endocrinol. 2002, 146: 777-781. 10.1530/eje.0.1460777.

    CAS  PubMed  Google Scholar 

  208. Pani MA, Regulla K, Segni M, Hofmann S, Hufner M, Pasquino AM, Usadel KH, Badenhoop K: A polymorphism within the vitamin D-binding protein gene is associated with Graves' disease but not with Hashimoto's thyroiditis. J Clin Endocrinol Metab. 2002, 87: 2564-2567. 10.1210/jc.87.6.2564.

    CAS  PubMed  Google Scholar 

  209. Ban Y, Taniyama M, Ban Y: Vitamin D receptor gene polymorphism is associated with Graves' disease in the Japanese population. J Clin Endocrinol Metab. 2000, 85: 4639-4643. 10.1210/jc.85.12.4639.

    CAS  PubMed  Google Scholar 

  210. Ban Y, Taniyama M, Ban Y: Vitamin D receptor gene polymorphisms in Hashimoto's thyroiditis. Thyroid. 2001, 11: 607-608. 10.1089/105072501750302967.

    CAS  PubMed  Google Scholar 

  211. Pani MA, Seissler J, Usadel KH, Badenhoop K: Vitamin D receptor genotype is associated with Addison's disease. Eur J Endocrinol. 2002, 147: 635-640. 10.1530/eje.0.1470635.

    CAS  PubMed  Google Scholar 

  212. Fukazawa T, Yabe I, Kikuchi S, Sasaki H, Hamada T, Miyasaka K, Tashiro K: Association of vitamin D receptor gene polymorphism with multiple sclerosis in Japanese. J Neurol Sci. 1999, 166: 47-52. 10.1016/S0022-510X(99)00112-4.

    CAS  PubMed  Google Scholar 

  213. Gyorffy B, Vasarhelyi B, Krikovszky D, Madacsy L, Tordai A, Tulassay T, Szabo A: Gender-specific association of vitamin D receptor polymorphism combinations with type 1 diabetes mellitus. Eur J Endocrinol. 2002, 147: 803-808. 10.1530/eje.0.1470803.

    CAS  PubMed  Google Scholar 

  214. Pedersen IB, Knudsen N, Jorgensen T, Perrild H, Ovesen L, Laurberg P: Thyroid peroxidase and thyroglobulin autoantibodies in a large survey of populations with mild and moderate iodine deficiency. Clin Endocrinol. 2003, 58: 36-42. 10.1046/j.1365-2265.2003.01633.x.

    Google Scholar 

  215. Jaume JC, Guo J, Pauls DL, Zakarija M, McKenzie JM, Egeland JA, Burek CL, Rose NR, Hoffman WH, Rapoport B, McLachlan SM: Evidence for genetic transmission of thyroid peroxidase autoantibody epitopic "fingerprints". J Clin Endocrinol Metab. 1999, 84: 1424-1431. 10.1210/jc.84.4.1424.

    CAS  PubMed  Google Scholar 

  216. Pirro MT, De Filippis V, Di Cerbo A, Scillitani A, Liuzzi A, Tassi V: Thyroperoxidase microsatellite polymorphism in thyroid diseases. Thyroid. 1995, 5: 461-464.

    CAS  PubMed  Google Scholar 

  217. Cuddihy RM, Dutton CM, Bahn RS: A polymorphism in the extracellular domain of the thyrotropin receptor is highly associated with autoimmune thyroid disease in females. Thyroid. 1995, 5: 89-95.

    CAS  PubMed  Google Scholar 

  218. Sunthornthepvarakul T, Kitvitayasak S, Ngowngarmaratana S, Konthong P, Deerochanawong C, Sarinnapakorn V, Phongviratchai S: Lack of association between a polymorphism of human thyrotropin receptor gene and autoimmune thyroid disease. J Med Assoc Thai. 1999, 82: 1214-1219.

    CAS  PubMed  Google Scholar 

  219. Tomer Y, Barbesino G, Greenberg DA, Concepcion E, Davies TF: Linkage analysis of candidate genes in autoimmune thyroid disease. III. Detailed analysis of chromosome 14 localizes Graves' disease-1 (GD-1) close to multinodular goiter-1 (MNG-1). International Consortium for the Genetics of Autoimmune Thyroid Disease. J Clin Endocrinol Metab. 1998, 83: 4321-4327. 10.1210/jc.83.12.4321.

    CAS  PubMed  Google Scholar 

  220. Allen EM, Hsueh WC, Sabra MM, Pollin TI, Ladenson PW, Silver KD, Mitchell BD, Shuldiner AR: A genome-wide scan for autoimmune thyroiditis in the Old Order Amish: replication of genetic linkage on chromosome 5q11.2-q14.3. J Clin Endocrinol Metab. 2003, 88: 1292-1296. 10.1210/jc.2002-020634.

    CAS  PubMed  Google Scholar 

  221. Sale MM, Akamizu T, Howard TD, Yokota T, Nakao K, Mori T, Iwasaki H, Rich SS, Jennings-Gee JE, Yamada M, Bowden DW: Association of autoimmune thyroid disease with a microsatellite marker for the thyrotropin receptor gene and CTLA-4 in a Japanese population. Proc Assoc Am Physicians. 1997, 109: 453-461.

    CAS  PubMed  Google Scholar 

  222. Akamizu T, Sale MM, Rich SS, Hiratani H, Noh JY, Kanamoto N, Saijo M, Miyamoto Y, Saito Y, Nakao K, Bowden DW: Association of autoimmune thyroid disease with microsatellite markers for the thyrotropin receptor gene and CTLA-4 in Japanese patients. Thyroid. 2000, 10: 851-858.

    CAS  PubMed  Google Scholar 

  223. Tomer Y, Greenberg DA, Concepcion E, Ban Y, Davies TF: Thyroglobulin is a thyroid specific gene for the familial autoimmune diseases. J Clin Endocrinol Metab. 2002, 87: 404-407. 10.1210/jc.87.1.404.

    CAS  PubMed  Google Scholar 

  224. Collins JE, Heward JM, Carr-Smith J, Daykin J, Franklyn JA, Gough SC: Association of a rare thyroglobulin gene microsatellite variant with autoimmune thyroid disease. J Clin Endocrinol Metab. 2003, 88: 5039-5042. 10.1210/jc.2003-030093.

    CAS  PubMed  Google Scholar 

  225. Rivolta CM, Moya CM, Mendive FM, Targovnik HM: Genotyping and characterization of two polymorphic microsatellite markers located within introns 29 and 30 of the human thyroglobulin gene. Thyroid. 2002, 12: 773-779. 10.1089/105072502760339334.

    CAS  PubMed  Google Scholar 

  226. Ban Y, Greenberg DA, Concepcion E, Skrabanek L, Villanueva R, Tomer Y: Amino acid substitutions in the thyroglobulin gene are associated with susceptibility to human and murine autoimmune thyroid disease. Proc Natl Acad Sci U S A. 2003, 100: 15119-15124. 10.1073/pnas.2434175100.

    PubMed Central  CAS  PubMed  Google Scholar 

  227. Ban Y, Tomer Y: The contribution of immune regulatory and thyroid specific genes to the etiology of Graves' and Hashimoto's diseases. Autoimmunity. 2003, 36: 367-379. 10.1080/08916930310001603037.

    CAS  PubMed  Google Scholar 

  228. Tomer Y, Greenberg D: The thyroglobulin gene as the first thyroid-specific susceptibility gene for autoimmune thyroid disease. Trends Mol Med. 2004, 10: 306-308. 10.1016/j.molmed.2004.05.002.

    CAS  PubMed  Google Scholar 

  229. Wall JD, Pritchard JK: Haplotype blocks and linkage disequilibrium in the human genome. Nat Rev Genet. 4: 587-597. 10.1038/nrg1123.

  230. Hutchings PR, Cooke A, Dawe K, Champion BR, Geysen M, Valerio R, Roitt IM: A thyroxine-containing peptide can induce murine experimental autoimmune thyroiditis. J Exp Med. 1992, 175: 869-872. 10.1084/jem.175.3.869.

    CAS  PubMed  Google Scholar 

  231. Champion BR, Rayner DC, Byfield PGH, Page KR, Chan CTJ, Roitt JM: Critical role of iodination for T cell recognition of thyroglobulin in experimental murine thyroid autoimmunity. J Immunol. 1987, 139: 3665-3670.

    CAS  PubMed  Google Scholar 

  232. Kacem HH, Rebai A, Kaffel N, Masmoudi S, Abid M, Ayadi H: PDS is a new susceptibility gene to autoimmune thyroid diseases: association and linkage study. J Clin Endocrinol Metab. 2003, 88: 2274-2280. 10.1210/jc.2002-021460.

    CAS  Google Scholar 

  233. Nilsson M: Iodide handling by the thyroid epithelial cell. Exp Clin Endocrinol Diabetes. 2001, 109: 13-17. 10.1055/s-2001-11014.

    CAS  PubMed  Google Scholar 

  234. Stuck BJ, Pani MA, Besrour F, Segni M, Krause M, Usadel KH, Badenhoop K: Fas ligand gene polymorphisms are not associated with Hashimoto's thyroiditis and Graves' disease. Hum Immunol. 2003, 64: 285-289. 10.1016/S0198-8859(02)00775-9.

    CAS  PubMed  Google Scholar 

  235. Park WS, Oh RR, Kim YS, Park JY, Lee SH, Shin MS, Kim SY, Kim PJ, Lee HK, Yoo NY, Lee JY: Somatic mutations in the death domain of the Fas (Apo-1/CD95) gene in gastric cancer. J Pathol. 2001, 193: 162-168. 10.1002/1096-9896(2000)9999:9999<::AID-PATH759>3.0.CO;2-A.

    CAS  PubMed  Google Scholar 

  236. Boldrini L, Loggini B, Gisfredi S, Zucconi Y, Baldinotti F, Fogli A, Simi P, Cervadoro G, Barachini P, Basolo F, Pingitore R, Fontanini G: Mutations of Fas (APO-1/CD95) and p53 genes in nonmelanoma skin cancer. J Cutan Med Surg. 2003, 7: 112-118.

    PubMed  Google Scholar 

  237. Lynch RA, Wagoner L, Li S, Sparks L, Molkentin J, Dorn GW: Novel and nondetected human signaling protein polymorphisms. Physiol Genomics. 2002, 10: 159-168.

    CAS  PubMed  Google Scholar 

  238. Barbesino G, Tomer Y, Concepcion ES, Davies TF, Greenberg DA: Linkage analysis of candidate genes in autoimmune thyroid disease. II. Selected gender-related genes and the X-chromosome. International Consortium for the Genetics of Autoimmune Thyroid Disease. J Clin Endocrinol Metab. 1998, 83: 3290-3295. 10.1210/jc.83.9.3290.

    CAS  PubMed  Google Scholar 

  239. Ban Y, Taniyama M, Tozaki T, Tomita M, Ban Y: Estrogen receptor alpha dinucleotide repeat polymorphism in Japanese patients with autoimmune thyroid diseases. BMC Med Genet. 2000, 1: 1-10.1186/1471-2350-1-1.

    PubMed Central  CAS  PubMed  Google Scholar 

  240. Ban Y, Tozaki T, Taniyama M, Tomita M, Ban Y: Lack of association between estrogen receptor beta dinucleotide repeat polymorphism and autoimmune thyroid diseases in Japanese patients. BMC Med Genet. 2001, 2: 1-10.1186/1471-2350-2-1.

    PubMed Central  CAS  PubMed  Google Scholar 

  241. Biunno I, Bernard L, Dear P, Cattaneo M, Volorio S, Zannini L, Bankier A, Zollo M: SEL1L, the human homolog of C. elegans sel-1: refined physical mapping, gene structure and identification of polymorphic markers. Hum Genet. 2000, 106: 227-235. 10.1007/s004390051032.

    CAS  PubMed  Google Scholar 

  242. Ban Y, Taniyama M, Tozaki T, Yanagawa T, Tomita M, Ban Y: SEL1L microsatellite polymorphism in Japanese patients with autoimmune thyroid diseases. Thyroid. 2001, 11: 335-338. 10.1089/10507250152039064.

    CAS  PubMed  Google Scholar 

  243. Field LL, Tobias R, Thomson G, Plon S: Susceptibility to insulin-dependent diabetes mellitus maps to a locus (IDDM11) on human chromosome 14q24.3-q31. Genomics. 1996, 33: 1-8. 10.1006/geno.1996.0153.

    CAS  PubMed  Google Scholar 

  244. Shirasawa S, Harada H, Furugaki K, Akamizu T, Ishikawa N, Ito K, Ito K, Tamai H, Kuma K, Kubota S, Hiratani H, Tsuchiya T, Baba I, Ishikawa M, Tanaka M, Sakai K, Aoki M, Yamamoto K, Sasazuki T: SNPs in the promoter of a B cell-specific antisense transcript, SAS-ZFAT, determine susceptibility to autoimmune thyroid disease. Hum Mol Genet. 2004, 13: 2221-2231. 10.1093/hmg/ddh245.

    CAS  PubMed  Google Scholar 

  245. Tufarelly C, Stanley JAS, Carrick D, Sharpe JA, Ayyub H, Wood WG, Higgs DR: Transcription of antisense RNA leading to gene silensing and methylation as a novel cause of human genetic disease. Nat Genet. 2003, 34: 157-165. 10.1038/ng1157.

    Google Scholar 

  246. Rouault JP, Rimokh R, Tessa C, Paranhos G, Ffrench M, Duret L, Garoccio M, Germain D, Samarut J, Magaud JP: BTG1, a member of a new family of antiproliferative genes. EMBO J. 1992, 11: 1663-1670.

    PubMed Central  CAS  PubMed  Google Scholar 

  247. Ahmad M, Srinivasula SM, Wang L, Talanian RV, Litwack G, Fernandes-Alnemri T, Alnemri ES: CRADD, a novel human apoptotic adaptor molecule for caspase-2, and FasL/tumor necrosis factor receptor-interacting protein RIP. Cancer Res. 1997, 57: 615-619.

    CAS  PubMed  Google Scholar 

  248. Delepine M, Pociot F, Habita C, Hashimoto L, Froguel P, Rotter J, Cambon-Thomsen A, Deschamps I, Djoulah S, Weissenbach J, Nerup J, Lathrop M, Julier C: Evidence of a non-MHC susceptibility locus in type I diabetes linked to HLA on chromosome 6. Am J Hum Genet. 1997, 60: 174-187.

    PubMed Central  CAS  PubMed  Google Scholar 

  249. Gaffney PM, Kearns GM, Shark KB, Ortmann WA, Selby SA, Malmgren ML, Rohlf KE, Ockenden TC, Messner RP, King RA, Rich SS, Behrens TW: A genome-wide search for susceptibility genes in human systemic lupus erythematosus sib-pair families. Proc Natl Acad Sci U S A. 1998, 95: 14875-14879. 10.1073/pnas.95.25.14875.

    PubMed Central  CAS  PubMed  Google Scholar 

  250. Van de Wetering M, Oosterwegel M, van Norren K, Clevers H: Sox-4, an Sry-like HMG box protein, is a transcriptional activator in lymphocytes. EMBO J. 1993, 12: 3847-3854.

    PubMed Central  CAS  PubMed  Google Scholar 

  251. Akamizu T, Hiratani H, Ikegami S, Rich SS, Bowen DW: Association study of autoimmune disease at 5q23-q33 in Japanese patients. J Hum Genet. 2003, 48: 236-242. 10.1007/s10038-003-0017-3.

    CAS  PubMed  Google Scholar 

  252. Walther A, Riehemann K, Gerke V: A novel ligand of the formyl peptide receptor: annxin I regulates neutrophil extravasation by interacting with FRP. Mol Cell. 2000, 5: 831-840. 10.1016/S1097-2765(00)80323-8.

    CAS  PubMed  Google Scholar 

  253. Lam E, Martin MM, Timerman AP, Sabers C, Fleischer S, Lukas T, Abraham RT, O'Keefe SJ, O'Neill EA, Wiederrecht GJ: A novel FK506 binding protein can mediate the immunosuppressive effects of FK506 and is associated with the cardiac ryanodine receptor. J Biol Chem. 1995, 270: 26511-26522. 10.1074/jbc.270.44.26511.

    CAS  PubMed  Google Scholar 

  254. Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B: A model for p53-induced apoptosis. Nature. 1997, 389: 300-305. 10.1038/38525.

    CAS  PubMed  Google Scholar 

  255. Flynn JC, Rao PV, Gora M, Alsharabi G, Wei W, Giraldo AA, David CS, Banga JP, Kohn LD, Napolitano G, Singer DS, Molteni M, Scorza R, Shimojo N, Kohno Y, Mozes E, Nakazato M, Ulianich L, Chung HK, Matoba H, Saunier B, Suzuki K, Schuppert F, Saji M: Graves' disease: a host defense mechanism gone awry. Int Rev Immunol. 2000, 19: 633-664.

    Google Scholar 

  256. Tomer Y, Barbesino G, Greenberg DA, Concepcion E, Davies TF: A new Graves disease-susceptibility locus maps to chromosome 20q11.2. Am J Hum Genet. 1998, 63: 1749-1756. 10.1086/302146.

    PubMed Central  CAS  PubMed  Google Scholar 

  257. Pearce SH, Vaidya B, Imrie H, Perros P, Kelly WF, Toft AD, McCarthy MI, Young ET, Kendall-Taylor P: Further evidence for a susceptibility locus on chromosome 20q13.11 in families with dominant transmission of Graves disease. Am J Hum Genet. 1999, 65: 1462-1465. 10.1086/302610.

    PubMed Central  CAS  PubMed  Google Scholar 

  258. Tomer Y, Concepcion E, Greenberg DA: A C/T single-nucleotide polymorphism in the region of the CD40 gene is associated with Graves' disease. Thyroid. 2002, 12: 1129-1135. 10.1089/105072502321085234.

    CAS  PubMed  Google Scholar 

  259. Watson PF, Pickerill AP, Davies R, Weetman AP: Analysis of cytokine gene expression in Graves' disease and multinodular goiter. J Clin Endocrinol Metab. 1994, 79: 355-360. 10.1210/jc.79.2.355.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dimitry A Chistiakov.

Additional information

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

ABFG

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chistiakov, D.A. Immunogenetics of Hashimoto's thyroiditis. J Autoimmune Dis 2, 1 (2005). https://doi.org/10.1186/1740-2557-2-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1740-2557-2-1

Keywords