You are on page 1of 9

Autoimmunity Reviews 19 (2020) 102649

Contents lists available at ScienceDirect

Autoimmunity Reviews
journal homepage: www.elsevier.com/locate/autrev

Hashimoto's thyroiditis: An update on pathogenic mechanisms, diagnostic T


protocols, therapeutic strategies, and potential malignant transformation

Massimo Rallia, , Diletta Angelettia, Marco Fioreb, Vittorio D'Aguannoa, Alessandro Lambiasea,
Marco Articoa, Marco de Vincentiisc, Antonio Grecoa
a
Department of Sense Organs, Sapienza University of Rome, Italy
b
CNR, Institute of Cell Biology and Neurobiology, Italy
c
Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Italy

A R T I C LE I N FO A B S T R A C T

Keywords: Hashimoto's thyroiditis, characterized by thyroid-specific autoantibodies, is one of the commonest autoimmune
Hashimoto's thyroiditis disorders. Although the exact etiology has not been fully elucidated, Hashimoto's thyroiditis is related to an
Autoimmunity interaction among genetic elements, environmental factors and epigenetic influences. Cellular and humoral
Hypothyroidism immunity play a key role in the development of the disease; thus, a T and B cells inflammatory infiltration is
Autoimmune thyroid disorders
frequently found. Histopathologic features of the disease include lymphoplasmacytic infiltration, lymphoid
follicle formation with germinal centers, and parenchymal atrophy. Moreover, the occurrence of large follicular
cells and oxyphilic or Askanazy cells is frequently associated to Hashimoto's thyroiditis. Clinically, Hashimoto's
thyroiditis is characterized mainly by systemic manifestations due to the damage of the thyroid gland, devel-
oping a primary hypothyroidism. Diagnosis of Hashimoto's thyroiditis is clinical and based on clinical char-
acteristics, positivity to serum antibodies against thyroid antigens (thyroid peroxidase and thyroglobulin), and
lymphocytic infiltration on cytological examination. The mainstream of treatment is based on the management
of the hypothyroidism with a substitution therapy. A relationship between Hashimoto's thyroiditis and a possible
malignant transformation has been proposed in several studies and involves immunological/hormonal patho-
genic links although specific correlation is still debated and needs to be further investigated with prospective
studies.

1. Introduction a specific clinical entity until 1931, when Allen Graham [8] described
the condition as an autonomous pathology. In 1956, Rose and Witebsky
Hashimoto thyroiditis (HT), also called chronic lymphocytic or au- [9,10] demonstrated that rabbit immunization with extracts of rabbit
toimmune thyroiditis, is an autoimmune thyroid disease characterized thyroid induced histologic modification on thyroid tissue similar to HT,
by increased thyroid volume, lymphocyte infiltration of parenchyma, identifying anti-thyroglobulin antibodies in the serum. In the same
and the presence of antibodies specific to thyroid antigens. HT is con- year, Roitt, Doniach et al. [11] isolated anti-thyroglobulin antibodies
sidered, together with Graves' Ddisease (GD), an autoimmune thyroid from the serum of patients with HT and stated that patients with HT
disorder (AITD) whose frequency has increased considerably in the may have an immunological reaction to thyroglobulin, concluding that
recent years [1–3]. HT is currently the leading cause of hypothyroidism Hashimoto's goiter should be considered an autoimmune disease of the
[1,4]; moreover, patients with HT are more likely to be affected by thyroid gland.
cardiovascular diseases and malignant neoplasms [5,6].
HT was first described by a Japanese physician, Haraku Hashimoto, 2. Epidemiology
in 1912 [7]. He found that thyroid tissue was infiltrated by lymphocytes
with increased volume of the gland, naming this disease as “struma Incidence of HT rapidly increased in the last 3 decades [12]. Cur-
lymphomatosa”. Hashimoto's struma lymphomatosa was not considered rently, HT is one of the most common thyroid diseases and its incidence


Corresponding author at: Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy.
E-mail addresses: massimo.ralli@uniroma1.it (M. Ralli), diletta.angeletti@uniroma1.it (D. Angeletti), marco.fiore@cnr.it (M. Fiore),
alessandro.lambiase@uniroma1.it (A. Lambiase), marco.artico@uniroma1.it (M. Artico), marco.devincentiis@uniroma1.it (M. de Vincentiis),
antonio.greco@uniroma1.it (A. Greco).

https://doi.org/10.1016/j.autrev.2020.102649
Received 15 March 2020; Accepted 21 March 2020
Available online 15 August 2020
1568-9972/ © 2020 Elsevier B.V. All rights reserved.
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

is 0.3–1.5 cases per 1000 people [13]. More than 10% of women display Eastern Asia population [32].
positive antibody and around 2% show clinical manifestations [14]; Studies have focused on possible polymorphism of the group of
men present one-tenth of this prevalence [15]. The white race shows a genes that encode for the cytokines involved in HT pathogenesis and
higher incidence than black, while HT is rare in Pacific Islanders [16]. progression; however, no significant results have been reported so far
Disease prevalence increases with age [16]. [33,34]. A major study of 202 Tunisians patients with HT revealed the
Although the reasons of female gender prevalence are still un- association with an IL1RN VNTR polymorphism, [33] while another
known, possible explanations could be found in the role of female sex study of 182 Chinese patients with HT showed the importance of the
hormones, as demonstrated in animal models of many autoimmune rs763780 polymorphism in IL17F [34].
diseases [17,18], or following the inactivation of chromosome X and Studies regarding GD have focused the attention on the lack of
fetal microchimerism [19]. In a recent study on 490 patients affected by regulatory T cells (Treg) that may induce a thyroid lymphocyte in-
HT, no significant differences in chromosome X inactivation were ob- filtration, associated with hypothyroidism in an animal model of GD
served compared to normal subjects, but four other meta-analysis [35,36]. This observation suggests that Treg cells in humans may de-
showed that chromosome X inactivation may be relevant [20]. If mi- termine a natural progression from the hyperthyroidism of GD to the
crochimerism played a significant role, HT should have been more hypothyroidism of HT [37]. Regulatory B cells (Breg) also seem to be
frequent in women with multiple pregnancies. However, a study of 4.6 involved in HT, although their role has not been completely understood
million Danes revealed only a irrelevant increase of autoimmune dis- and further studies are needed [38].
eases in women with a previous pregnancy, compared to those who did Chemokines also play a role in HT and other autoimmune thyroid
not have children [21]. The pathogenesis of HT has also been associated disorders. In a recent study, Ferrari et al. investigated the modulation of
with climate conditions, since Siberian women have higher thyroid the secretion of chemokines CXCL8 and CXCL10 in cell cultures of
peroxidase (TPO) antibody title than general population [22]. thyroid follicular cells in GD, concluding that CXCL10 could be asso-
Last, HT may have a higher prevalence in some conditions such as ciated with the initial phase of GD, while CXCL8 could be associated
myasthenia gravis (MG) and systemic sclerosis. A recent meta-analysis with a later chronic phase of the disease [39].
from Song et al. on 39 studies showed reliable evidence that HT, along Furthermore, selenoproteins (SEP) are important for the thyroid
with other thyroid disorders, have a higher prevalence in patients with hormone deiodination and selenium deficiency could be considered a
MG and identified MG as a risk factor for thyroid autoimmunity [23]. In predisposing factor as dietary environmental element [40,41], as it will
a meta-analysis conducted on 46 studies by Yao et al., the authors found be further discussed in the paragraph on environmental triggers. In a
a considerably high prevalence of autoimmune thyroid conditions in study on a Portuguese population including 481 subjects, HT suscept-
subjects with systemic sclerosis, and that this condition is associated ibility has been associated to a polymorphism in the promoter region of
with increased risks of thyroid pathologies [24]. Although the patho- the selenoprotein S gene (SEPS1) [42].
genic mechanisms of the associations between thyroid autoimmunity
and these conditions are not clear, it has been hypothesized that im- 3.2. Environmental triggers
mune defects, hormones, genetic and environmental factors may play a
central role in poly-autoimmunity [25]. Environmental factors could play an important role, as demonstrate
recent epidemiological changes, and development of HT may not be
3. Etiology only due to an innate predisposition but may be a consequence of en-
vironmental factors that have changed rapidly. Moreover, as demon-
Although still largely unknown, pathogenesis of HT is related to strated in homozygous twins, the disease only clinically manifests
genetic influences, environmental triggers and epigenetic effects [26]. manifests in about 50% of the subjects, thus in genetically predisposed
subjects some environmental factors could cause an autoimmune re-
3.1. Genetic susceptibility action against the thyroid.
As observed in several autoimmune disorders, the presence of more
A genetic susceptibility to HT disease has been shown in epide- hygienic environment without microbial agents may be associated with
miological studies that focused on familial predisposition [27]. Brix an high incidence of allergic and autoimmune diseases, including HT
et al. showed in Danish twins that monozygotic twins exhibited a [43]. Many studies have shown that the excess of iodine in the diet may
concordance rate of more than 50%, while dizygotic twins showed determine the onset of HT in predisposed individuals. A study of io-
absence of any concordance [28]. Moreover, data from the same study doprophylaxis on volunteers in a region of Italy with iodine deficiency
regarding thyroid autoantibodies showed an high concordance rate for revealed a doubled occurrence of anti-thyroid antibodies with a quad-
monozygotic twins that was nearly 80%, when compared to dizygotic rupled incidence of HT during the observation period [44].
ones (40%) [28]. Insufficient selenium intake with diet may result in a worsening of
Several genes have been shown to be involved in HT pathogenesis, HT. Selenium intake in Europe showed a decrease of 50% in the last
including genes of immune response and thyroid function. Among the 30 years. Nevertheless, the administration of selenium supplements did
genes that control the immune response, a relevant role is played by not show an improvement in thyroid morphology but only a reduction
those coded in the Human Leukocyte Antigen (HLA) complex; thus, it in levels of TPO autoantibodies, as demonstrated in recent meta-ana-
has been showed that the HLAeB* 46:01 gene is associated with the lysis [45].
development of HT, as demonstrated in Chinese children is a case- Another dietary component that could have a role in HT is vitamin
control and family-based study [29]. In another study of 444 Japanese D, whose serum levels are related to exposure to the sun. Although
patients with HT, some genes (HLA-A* 02:07 and HLA-DRB4) were lower serum levels of vitamin D were observed in subject with HT, these
shown to increase the chance of illness while others favored protection might be related to metabolic changes in hypothyroidism, especially
[30]. because thyroid dysfunction is inversely related to the severity of vi-
Current literature established the involvement of many other im- tamin D levels [46]. A recent study confirmed that it should be taken
munoregulatory genes that control the immune response, in addition to into account in future research [47].
those in the HLA complex. Single nucleotide polymorphisms (SNPs) Novel anticancer regimens such as interferon-α and tyrosine kinase
regarding the CTLA-4, PTPN22, CD14, CD40 and IL2R genes have been inhibitors have been associated to thyroid diseases including HT [48].
associated to the development of HT [31]. A meta-analysis by Ji et al. The role of smoking and alcohol in the etiopathogenesis of HT is still
regarding a specific polymorphism involving A49G in CTLA-4 gene has controversial and no clear evidence is available so far, although it seems
demonstrated an increased risk of HT for white race subjects and for that a moderate consumption of alcohol may be protective against HT

2
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

and other autoimmune diseases [49,50]. It has been hypothesized that in HT there is an alteration in the function
The observation that Hepatitis C virus may cause a worsening of HT of T cell suppressors against specific antigens of thyroid cells [69].
has suggested a possible mechanism of molecular mimicry between Some of the functions of T suppressor cells seem to be carried out by the
viral and self-antigens. However, so far, all attempts to find viruses in T regulator cells (Treg) [70]. These cells can attenuate the immune
thyroid patients with HT did not give reliable results. Human response by direct or indirect contact with the production of cytokines
Herpesvirus 6 has been shown to be active in some studies in HT pa- such as growth factor (TGF)-Beta and Interleukin 10 [70,71]. Some
tients and demonstrated a strong tropism for thyroid follicular cells studies have demonstrated an alteration in the number and function of
(TFCs). However, these studies only included a relatively small number Treg cells in HT [72,73].
of patients and require further validation [51,52]. The role of Treg cells has been focused in a recent study [36] that
analyzed the rate and the expressions of Helios and PD-1 in HT patients,
3.3. Epigenetics factors exploring the relationship of these with thyroid function and specific
autoantibodies in peripheral blood mononuclear cells of HT patients
Current research has shown that genetic and environmental factors and healthy patients. In particular, Helios is considered as an important
act synergically in determining HT through modulation of epigenetic mediator for Treg cells since it can upregulate Foxp3 expression. It has
factors [53–57]. Epigenetic factors may regulate gene expression and been acknowledged that Treg-coexpressing Foxp3 and Helios pheno-
phenotype, resulting in the onset of disease in absence of DNA structure type exhibited a superior suppressive abilities than CD4 + CD25+ Treg
alterations [58]. [74]. Moreover, PD-1 delivers inhibitory signal to prevent immune
Epigenetic factors involved in the onset of the disease are numerous, damage when binding to its ligands PD-L1 [75]. Helios and PD-1 may
although methylation, histone modifications, and RNA interference also exert vital function in regulating Treg cell peripheral tolerance and
through non-coding RNAs are the most frequent [58]. HT is char- autoimmunity. Interestingly, a recent study based on flow cytometry
acterized by lymphocytic infiltration in the thyroid, followed by the analysis detected the percentage of Treg cells was remarkably lower in
infiltration of T and B cells into the thyroid gland. It has been hy- HT patients with an inverse correlation to thyroid function when
pothesized that autoantibodies and B cell dysfunction represent the compared with healthy controls. The levels of Treg, aTreg, and Helios-
primary immune reactions in autoimmune thyroid disorders, and expressing aTreg cells were all negatively correlated with antithyroid
aberrant functions of T cell subsets also play important roles in breaking antibodies, confirming that the deficiency of Treg frequency and
the immune homeostasis and starting the autoimmune cascade against aberrant expressions of Helios and PD-1 may possibly contribute to the
thyroid tissues (Fig. 1) [55]. immune damage to the thyroid in HT [36,76] (Fig. 2).
The methylation of DNA can determine the inactivation of certain
genes, while some histone alterations induce the activation of other 4.2. Humoral immunity
genes; however, the action of these epigenetic mechanisms can be
variable and influenced by environmental factors [58,59]. Moreover, The production of specific antibodies to thyroid tissue is one of the
non-coding RNAs including microRNAs can also control the expression most important features of HT. In the great majority of HT patients,
of specific genes [60,61]. there are specific autoantibodies for TG and TPO [77]. In addition, the
Evidence suggests that female preponderance in HT may be due to X anti-TPO antibody assay is useful in predicting a condition of hy-
chromosome inactivation, considered a major epigenetic feature in pothyroidism [78]. Recently, a variant of HT called IG4 thyroiditis has
which one X chromosome is silenced [55]. This suggests that the been identified and is part of a systemic autoimmune disease char-
functioning of the genes can be altered by epigenetic mechanisms and acterized by the presence of IG4-positive cells [79].
result in the onset of autoimmune disorders; therefore, epigenetic fac- Some studies have found an increase of serum levels of Th1 cells
tors could play a fundamental role together with genetic and environ- [80,81] and IL-17 and IL-22 cytokines in HT [82]. IL-12 cytokine has
mental factors in determining autoimmune diseases [54,55,62]. been shown to be increased in 56% of patients with HT [83].
This growing evidence suggests that environmental factors can in- A recent study [84] suggested that circulating exosomes play an
duce epigenetic modifications that, in genetically susceptible in- active role in the pathogenesis of HT. Exosomes have the capability to
dividuals, may produce autoimmunity thyroid diseases including HT transfer bioactive molecules into other cells thus affecting biological
[63,64]. activity and are involved in several cellular process as antigen pre-
sentation, inflammatory activation, autoimmune disorders and tumor
4. Pathogenic mechanisms metastasis (5,6). Specific HT-exosomes might present antigens to den-
dritic cells (DC) and bind TLR2/3, causing DC activation via the NFκB
Pathogenesis of HT is strictly related to autoantibodies with a re- signaling pathway, leading to an imbalance in CD4+ T lymphocyte
levant lymphocytic infiltrate, including of B and T cells in the thyroid differentiation and potentially contributing to HT onset. [85] A similar
tissue [65,66]. It is believed that one of the first events in HT patho- process has been demonstrated in systemic lupus erythematosus;
genesis is a functional alteration of B cells with formation of auto- however, further studies are required to confirm this hypothesis in
antibodies. In addition, T cell dysfunction is associated with the patients with HT.
breakdown of immune homeostasis against thyroid tissue [65,66]. HT is often associated with other autoimmune disorders, indicating
Therefore, it could be hypothesized that cellular and humoral immunity a possible common poly-autoimmune etiology. In a large prospective
is associated to the pathogenesis of HT. study on 3209 patients with GD, Ferrari et al. evaluated the association
of this thyroid disorder with other autoimmune conditions. A sig-
4.1. Cellular immunity nificant percentage (16.7%) of GD patients had another associated
autoimmune disease such as vitiligo, autoimmune gastritis, rheumatoid
In HT patients, CD8+ T cells against thyroglobulin and TPO were arthritis, polymyalgia rheumatica, multiple sclerosis, and celiac disease.
found [67]. However, only a small number (2–3%) of CD8+ cells are In some patients, three or more associated autoimmune disorders were
specific to TG/TPO, so most of them are not specific to thyroid antigens. diagnosed [86].
Moreover recent studies established that cell death in autoimmune
thyroiditis is not only due to cytotoxicity but also to apoptosis processes 5. Histopathology
[68].
A specific population-specialized CD8+ cells, termed “Suppressor T Histopathological characteristics of HT include lymphoplasmacytic
cells” have been considered able to inhibit harmful immune responses. infiltration, fibrotic tissue presence, lymphatic follicular formation,

3
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

Fig. 1. Loss of immune tolerance results in autoimmunity during the development of autoimmune thyroid diseases. Naive CD4+ T cells can be activated by dendritic
cells (DC) or other antigen-presenting cells and they can differentiate into various subsets which are characterized by different cytokines and specific transcription
factors. The balance of those immune cells is necessary for the maintenance of immune homeostasis. Under normal conditions, T cell subsets have normal functions,
and there is immune homeostasis in human body, which can maintain the immune tolerance and avoid unwarranted immune attacks to thyroid tissues. Some genetic
factors and environmental factors can result in the dysfunctions of these T cell subsets, B cells, and antigen-presenting cells, which may break up the immune
homeostasis and cause thyroid autoimmunity. From Wang B, Shao X, Song R, Xu D, Zhang JA. The Emerging Role of Epigenetics in Autoimmune Thyroid Diseases. Front
Immunol. 2017 Apr 7;8:396 [55].

parenchyma atrophy and presence in lymphoid follicles of large cells disease (IG4-RD) [97].
with eosinophilic granule in the cytoplasm called Hurtle cells [87]. The growing interests on IG4-related disease provided recently a
Hypothyroidism is due to the destruction of thyroid cells [88]. simplified classification [98], where HT is divided into IgG4-positive
Histopathological features of HT are not unique, although several and IgG4-negative groups, based on immunohistochemistry for IgG4
different variants of HT have been identified through clinical and his- and IgG. Patients in the IgG4-positive group were significantly younger
tologic features, such as fibrotic and atrophic [89,90], Riedel thyroiditis than those in the IgG4-negative HT group, and displayed a significantly
[91,92] and IG4 thyroiditis [93,94]. higher degree of fibrosis of thyroid parenchyma. Immunohistochemical
In the fibrous variant, thyroid tissue is completely replaced by fi- expression score for TGF-b1 was higher in IgG4-positive group than in
brous tissue. However, fibrosis never extends beyond the thyroid cap- IgG4-negative, suggesting that this new classification might have re-
sule as is the case for malignant thyroid neoplasms [15]. levant clinical implications for the management of HT [99].
In the fibrous atrophy variant, the thyroid gland is reduced in volume
with large quantities of fibrous tissue and atrophy of the thyroid tissue
[88,95]. TSH antibody receptors can be detected in a small percentage 6. Symptomatology
of cases [96].
Riedel's thyroiditis was so named by the name of its discoverer Symptomatology of HT is characterized by local and systemic
Bernhard Riedel [92], and was later considered as a local manifestation manifestations. Local symptoms are due to the compression of the
of a systemic disease called multifocal fibrosclerosis [91]. The main anatomical structures of the neck, including dysphonia following the
histological features are the presence of abundant fibrotic tissue ex- involvement of the recurrent laryngeal nerve, dyspnea due to com-
tending beyond the thyroid capsule in the absence of neoplastic cells pression of the trachea, and dysphagia consequent to compression of
[15]. the esophagus.
IgG4 thyroiditis has been proposed recently as another variant of HT Systemic symptoms are more common and are due to primary hy-
[94]. This variant is characterized by high concentrations in thyroid pothyroidism that occurs almost always in HT and involves most organs
tissue and serum of IG4. Probably, this variant represents a local and tissues with significant variability [100], although often preceded
manifestation of a systemic disease called Immunoglobulin G4-related by subclinical manifestations [101,102].
Non-specific rheumatic manifestations associated with

4
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

Fig. 2. Thyroid autoimmunity produces two opposite pathogenetic processes and clinical outcomes. A: During Hashimoto's thyroiditis, self-reactive CD4+ T lym-
phocytes recruit B cells and CD8+ T cells into the thyroid. Disease progression leads to the death of thyroid cells and hypothyroidism. Both autoantibodies and
thyroid-specific cytotoxic T lymphocytes (CTLs) have been proposed to be responsible for autoimmune thyrocyte depletion. B: In Graves' disease, activated CD4+ T
cells induce B cells to secrete thyroid-stimulating immunoglobulins (TSI) against the thyroid-stimulating hormone receptor (TSHR), resulting in unrestrained thyroid
hormone production and hyperthyroidism. From: Stassi G, De Maria R. Autoimmune thyroid disease: new models of cell death in autoimmunity. Nat Rev Immunol 2002,
2(3):195–204 [76].

undifferentiated inflammatory arthropathy have been recently ob- 7. Diagnosis


served in patients with HT [103,104].
Several clinical variants of HT have been described, and include The diagnosis of HT is based on clinical symptoms, anti-thyroid
painless thyroiditis, painful thyroiditis, postpartum thyroiditis, and antibodies and histological features.
Hashimoto's encephalopathy. Serum anti-TPO antibodies are considered the most important fea-
Painless thyroiditis is so named because the patient does not feel any ture of HT and are present in about 95% of patients [100]. Instead, anti-
pain in the neck and its main feature is represented by a transient phase thyroglobulin antibodies are present in a lower (60–80%) percentage of
of hypothyroidism that return to euthyroidism [105]. cases and therefore are less reliable for diagnosis [116]. It appears that
Painful Thyroiditis is so called because of the progressive, acute and anti-thyroglobulin antibodies may be the expression of an initial im-
intolerable pain in one lobe or the whole thyroid. It is a rare variant of mune response, whereas anti-TPO antibodies may be the result of a
HT that mostly affect women with a sex ratio of 10 to 11:1 [106]. later immune response as if there was an immune escalation [117].
Postpartum thyroiditis is so called because it manifests six months Cytological examination is not routinely performed, but only when
after delivery and is clinically identical to the painless thyroiditis. The a thyroid nodule is present with a suspicion of malignant transforma-
favorable course of these two clinical forms is probably due to transient tion. Moreover, HT ultrasonographic characteristics could make diffi-
autoimmune disorders [105]. cult the nodule identification and aspiration, although not associated
Hashimoto's encephalopathy is one of the most relevant clinical var- with non-diagnostic and indeterminate cytological rates [118]. The
iants of HT [107] due to its association with encephalopathy as first presence of lymphocytes in contact with thyroid cells is considered the
described by Brain in 1966 [108]. Later, many other clinical cases were most important element to make a differential diagnosis between HT
identified [109]. The most frequent symptoms include subacute cog- and thyroid tumors [119] (Fig. 3).
nitive deterioration, myoclonus, change in behavior, and seizures Radiologic evaluation of HT includes mainly sonographic ex-
[110]. Clinically, this condition responds well to corticosteroid therapy, amination, where specific features are considered a decreased echo-
and seems to be supported by alpha enolase autoantigens genicity, heterogeneity hypervascularity and presence of hypoechoic
[107,111–114]. The search for anti-alpha enolase antibodies is useful micronodules with echogenic rim [120].
for the diagnosis and treatment of Hashimoto's encephalopathy but
does not allow a connection between thyroid and encephalic disease
[115].

5
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

Fig. 3. (A) Histologic section of papillary thyroid carcinoma (left) and Hashimoto thyroiditis background (right). (B) Hashimoto thyroiditis showing effacement of
thyroid architecture by diffuse lymphocyte infiltration and residual thyroid follicles. From: Nam YJ, Kim BH, Lee SK, Jeon YK, Kim SS, Jung WJ, Kahng DH, Kim IJ. Co-
occurrence of papillary thyroid carcinoma and mucosa-associated lymphoid tissue lymphoma in a patient with long-standing hashimoto thyroiditis. Endocrinol Metab 2013,
28(4):341–5 [142].

8. Treatment The role of surgery for HT is limited for cases with a compression of
cervical anatomical structures or in the presence of a nodule with
The main purpose of HT treatment is the control of hypothyroidism malignant transformation characteristics [13,134]. However, thyr-
and consists of oral administration of a synthetic hormone, the Levo- oidectomy performed in HT patients is burdened with a greater number
Thyroxine 4 (L-T4) [121], at a dosage of 1.6–1.8 micrograms per kilo. of complications compared to other thyroid disorders [135]. As a future
The substitution therapy must be prolonged for a lifetime to achieve perspective, thyroid gland transplantation has also been proposed to
normal circulating thyrotropin (TSH) levels. In selected cases, L-T4 correct hypothyroidism but needs to be validated by further studies
treatment may be not required and only clinical observation is needed. [136].
The role of glucocorticoids been questioned, as they may regulate
the thyroiditis and acutely improve thyroid function, although the risks 9. Malignant transformation
associated with the high dose and long treatment are considered to
outweigh the benefit [122]. However, a short-term use of prednisolone Occurrence of HT is frequently found in thyroid glands resected for
may have a longer-term benefit in IgG4-disease subgroup [123]. a neoplastic process. The association of HT and papillary thyroid cancer
The additional role of a specific diet for the management of HT has (PTC), firstly reported by Dailey in 1955 [137], has been widely de-
been questioned in recent years [124]. It has been postulated that an bated. Controversy exists whether HT predisposes patients to the de-
excessive iodine intake induces thyroid autoimmunity by increasing the velopment of PTC, since this association may be considered a chance
immunogenicity of thyroglobulin in genetically predisposed subjects occurrence of two relatively common diseases or may be indicative of a
[125]. Thus, an high iodine supplementation in HT should be dis- cause and effect relationship, or at least a predisposing factor. This
couraged, as possibly dangerous, although an appropriate supple- relationship has been postulated since chronic inflammation may lead
mentation should be proposed in pregnancy to a total intake of 250 g/ to the development of a neoplastic transformation, as demonstrated for
day [122]. other tissues. Moreover, the prolonged elevated levels of TSH in HT
Selenium plays a key role in human thyroid hormone homeostasis, patients may stimulate follicular epithelial proliferation, thereby pro-
as several selenoproteins are involved in thyroid function, although moting the development of PTC [138–140] (Fig. 4).
efficacy of selenium supplementation in HT patients is debated [126]. Several studies have investigated this association; however, results
Oral administration of selenium in the form of seleno-methionine would are dissimilar among studies. Interestingly, ambiguous data regarding
be beneficial in HT patients with selenium deficiency and should pro- this association may be due to a potential selection bias, as the majority
tect the thyroid gland from the autoimmune reaction [127]. However, of the publications in the past decades were retrospective analyses of
the literature is discordant [128]. A systematic review and meta-ana- patients undergoing thyroidectomy [138]. In the last years, the re-
lysis by Wichman et al. [129] showed a reduction of serum TPO-Ab lationship between HT and PTC has been achieved by fine-needle as-
levels and serum Tg-Ab; however, no significant correlation between piration cytology (FNAC) data, that are more representative of a typical
the baseline selenium levels and the decrease in serum TPO-Ab level population of patients with HT [138]. Data obtained from FNAC
was demonstrated. A more recent systematic review and meta-analysis showed that the average prevalence of PTC in patients with HT was
regarding the efficacy of oral administration of selenium revealed no 1.20%, with an average risk ratio of 0.69, compared with the studies
effect of selenium supplementation on TSH levels, health-related based on thyroidectomy data, where the average prevalence was about
quality of life [130] or thyroid ultrasound, in levothyroxine substitu- 27% and risk ratio was and 1.59 [138,141]. These data confirmed a
tion-untreated individuals, and sporadic evaluation of clinically re- higher risk reported in studies of thyroidectomy specimens, compared
levant outcomes in levothyroxine substitution-treated patients [131]. to studies of patients undergoing FNAC. Studies regarding FNAC show
The association between vitamin D deficiency, HT pathogenesis and no significant increase of PTC in patients with HT, although FNAC
thyroid hypofunction has been demonstrated in several studies specimens may be limited by a lack of definitive histological pathology.
[126,132,133]. Since the low cost and the minimal side-effect of oral To date, evidence favoring a causal relationship between HT and PTC is
vitamin D supplementation, screening for vitamin D deficiency and inconsistent, and a validated criterion to identify HT patients at a
supplementation may be recommended for patients with HT, with higher risk of developing PTC is required, although a causative re-
monthly monitoring of calcium and 25[OH]D levels, when clinically lationship between HT and PTC could not be excluded.
required [126]. Despite PTC, occurrence of Non-Hodgkin primary thyroid

6
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

Fig. 4. The relationships between Hashimoto thyroiditis (HT) and papillary thyroid carcinoma (PTC) with their immunological/hormonal pathogenic links. The left
part shows the relevance of environmental factors and genetic background in thyroid carcinogenesis. Radiation may cause PTC development by RET-PTC oncogene
rearrangements and more rarely with point BRAF mutations. Increased TSH levels due to nonautoimmune thyroid failure induced by radiation play an important role
both in promotion and progression of PTC. The right part displays the complex relation between HT and PTC. Multiple factors related to thyroid autoimmunity
(serum anti-thyroid autoantibodies, inflammatory molecules, free radicals with secondary RET/PTC rearrangements), genetic/environmental conditions, and high
TSH values (consequence of autoimmune thyroid failure) are involved. From: Boi F, Pani F, Mariotti S. Thyroid Autoimmunity and Thyroid Cancer: Review Focused on
Cytological Studies. Eur Thyroid J. 2017 Jul; 6(4): 178–186 [140].

lymphoma has been strongly associated with HT, as considered the only References
recognized risk factor, with a risk of about 60 times higher in patients
HT [90]. Thyroid lymphoma accounts approximately 5% of all thyroid [1] Antonelli A, Ferrari SM, Corrado A, Di Domenicantonio A, Fallahi P. Autoimmune
neoplasms, with a female predominance and histologically are mainly thyroid disorders. Autoimmun Rev 2015;14:174–80.
[2] Leese GP, Flynn RV, Jung RT, Macdonald TM, Murphy MJ, Morris AD. Increasing
characterized by B cell phenotype, although a T-cell phenotype may prevalence and incidence of thyroid disease in Tayside, Scotland: the Thyroid
also occur [90]. Epidemiology Audit and Research Study (TEARS). Clin Endocrinol (Oxf)
2008;68:311–6.
[3] McLeod DS, Cooper DS. The incidence and prevalence of thyroid autoimmunity.
10. Conclusions Endocrine 2012;42:252–65.
[4] De Leo S, Lee SY, Braverman LE. Hyperthyroidism. Lancet 2016;388:906–18.
[5] Chen WH, Chen YK, Lin CL, Yeh JH, Kao CH. Hashimoto’s thyroiditis, risk of
Although many genetic and environmental factors that could trigger coronary heart disease, and L-thyroxine treatment: a nationwide cohort study. J
an autoimmune response have been identified, the exact pathogenic Clin Endocrinol Metab 2015;100:109–14.
[6] Chen YK, Lin CL, Cheng FT, Sung FC, Kao CH. Cancer risk in patients with
mechanisms of HT are still unknown. Recently, the significance of Hashimoto’s thyroiditis: a nationwide cohort study. Br J Cancer
epigenetic factors in HT etiopathogenesis has been highlighted, but 2013;109:2496–501.
further studies are necessary to accurately understand their role. The [7] Zur HH. Kenntniss der lymphomatösen Veränderung der Schilddrüse (Struma
lymphomatosa). Arch Klin Chir 1912:219–48.
treatment of HT, which currently focuses on clinical symptoms of the
[8] Graham A. Riedel’s struma in contrast to struma lymphomatosa (Hashimoto). West
disease, should in the future act on the autoimmune mechanism that J Surg 1931;39:681–9.
causes the destruction of the thyroid parenchyma and the consequent [9] Rose WE. NR, Studies on organ specificity. V. Changes in the thyroid glands of
rabbits following active immunization with rabbit thyroid extracts. J Immunol
hypothyroidism. A better understanding of epigenetic modifications
1956;76:417–27.
and autoimmune pathogenic mechanisms could contribute to a more [10] Witebsky RN, Paine JR, Egan RW. Thyroid-specific autoantibodies. Ann N Y Acad
accurate diagnosis of HT, a more adequate choice of treatment ap- Sci 1957;69:669–77.
proach, and a more precise prediction of treatment outcomes. [11] Roitt IM, Doniach D, Campbell PN, Hudson RV. Auto-antibodies in Hashimoto’s
disease (lymphadenoid goitre). Lancet 1956;271:820–1.
[12] Ott J, Meusel M, Schultheis A, Promberger R, Pallikunnel SJ, Neuhold N, et al. The
incidence of lymphocytic thyroid infiltration and Hashimoto’s thyroiditis increased
Funding source in patients operated for benign goiter over a 31-year period. Virchows Arch
2011;459:277–81.
the authors report no funding sources for this article. [13] Caturegli P, De Remigis A, Chuang K, Dembele M, Iwama A, Iwama S. Hashimoto’s
thyroiditis: celebrating the centennial through the lens of the Johns Hopkins
hospital surgical pathology records. Thyroid 2013;23:142–50.
[14] Tunbridge WM, Brewis M, French JM, Appleton D, Bird T, Clark F, et al. Natural
Declaration of Competing Interest history of autoimmune thyroiditis. Br Med J (Clin Res Ed) 1981;282:258–62.
[15] Hiromatsu Y, Satoh H, Amino N. Hashimoto’s thyroiditis: history and future out-
look. Hormones (Athens) 2013;12:12–8.
the authors report no conflict of interest.

7
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

[16] McLeod DS, Caturegli P, Cooper DS, Matos PG, Hutfless S. Variation in rates of supplementation in the treatment of Hashimoto’s thyroiditis: a systematic review
autoimmune thyroid disease by race/ethnicity in US military personnel. JAMA and a meta-analysis. Thyroid 2010;20:1163–73.
2014;311:1563–5. [46] Tamer G, Arik S, Tamer I, Coksert D. Relative vitamin D insufficiency in
[17] Lee TP, Chiang BL. Sex differences in spontaneous versus induced animal models Hashimoto’s thyroiditis. Thyroid 2011;21:891–6.
of autoimmunity. Autoimmun Rev 2012;11:A422–9. [47] Anaraki PV, Aminorroaya A, Amini M, Feizi A, Iraj B, Tabatabaei A. Effects of
[18] Shetty A, Chowdappa V. Cytomorphological spectrum of Hashimoto's thyroiditis Vitamin D deficiency treatment on metabolic markers in Hashimoto thyroiditis
and its correlation with hormonal profile and hematological parameters. J Cytol patients. J Res Med Sci 2017;22:5.
2019;36:137–41. [48] Torino F, Barnabei A, Paragliola R, Baldelli R, Appetecchia M, Corsello SM.
[19] Brix TH, Hansen PS, Kyvik KO, Hegedus L. Aggregation of thyroid autoantibodies Thyroid dysfunction as an unintended side effect of anticancer drugs. Thyroid
in twins from opposite-sex pairs suggests that microchimerism may play a role in 2013;23:1345–66.
the early stages of thyroid autoimmunity. J Clin Endocrinol Metab [49] Carle A, Pedersen IB, Knudsen N, Perrild H, Ovesen L, Rasmussen LB, et al.
2009;94:4439–43. Moderate alcohol consumption may protect against overt autoimmune hypothyr-
[20] Simmonds MJ, Kavvoura FK, Brand OJ, Newby PR, Jackson LE, Hargreaves CE, oidism: a population-based case-control study. Eur J Endocrinol 2012;167:483–90.
et al. Skewed X chromosome inactivation and female preponderance in auto- [50] Effraimidis G, Tijssen JG, Wiersinga WM. Alcohol consumption as a risk factor for
immune thyroid disease: an association study and meta-analysis. J Clin Endocrinol autoimmune thyroid disease: a prospective study. Eur Thyroid J 2012;1:99–104.
Metab 2014;99:E127–31. [51] Caselli E, Zatelli MC, Rizzo R, Benedetti S, Martorelli D, Trasforini G, et al.
[21] Jorgensen KT, Pedersen BV, Nielsen NM, Jacobsen S, Frisch M. Childbirths and risk Virologic and immunologic evidence supporting an association between HHV-6
of female predominant and other autoimmune diseases in a population-based and Hashimoto's thyroiditis. PLoS Pathog 2012;8:e1002951.
Danish cohort. J Autoimmun 2012;38:J81–7. [52] Therese Weider SJR, Morgan Noel G, Paulsen Trond H, Dahl-Jørgensen Knut,
[22] Weetman AP. The immunopathogenesis of chronic autoimmune thyroiditis one Hammerstad Sara Salehi. Upregulation of HLA class I and Antiviral Tissue
century after hashimoto. Eur Thyroid J 2013;1:243–50. Responses in Hashimoto’s Thyroiditis. 30(3). 2019. p. 432–42. (Mar 2020).
[23] Song RH, Yao QM, Wang B, Li Q, Jia X, Zhang JA. Thyroid disorders in patients [53] Marsit CJ. Influence of environmental exposure on human epigenetic regulation. J
with myasthenia gravis: a systematic review and meta-analysis. Autoimmun Rev Exp Biol 2015;218:71–9.
2019;18:102368. [54] Canas CA, Canas F, Bonilla-Abadia F, Ospina FE, Tobon GJ. Epigenetics changes
[24] Yao Q, Song Z, Wang B, Zhang JA, Mu K. Thyroid disorders in patients with sys- associated to environmental triggers in autoimmunity. Autoimmunity
temic sclerosis: a systematic review and meta-analysis. Autoimmun Rev 2016;49:1–11.
2019;18:634–6. [55] Wang B, Shao X, Song R, Xu D, Zhang JA. The emerging role of epigenetics in
[25] Lazurova I, Benhatchi K. Autoimmune thyroid diseases and nonorganspecific au- autoimmune thyroid diseases. Front Immunol 2017;8:396.
toimmunity. Pol Arch Med Wewn 2012;122(Suppl. 1):55–9. [56] Mazzone R, Zwergel C, Artico M, Taurone S, Ralli M, Greco A, et al. The emerging
[26] Hasham A, Tomer Y. Genetic and epigenetic mechanisms in thyroid autoimmunity. role of epigenetics in human autoimmune disorders. Clin Epigenetics 2019;11:34.
Immunol Res 2012;54:204–13. [57] Ralli M, De Virgilio A, Artico M, Longo L, de Vincentiis M, Greco A. New insights
[27] Kust D, Matesa N. The impact of familial predisposition on the development of into the etiopathogenesis of Hashimoto's thyroiditis: the role of genetics and epi-
Hashimoto's thyroiditis. Acta Clin Belg 2018:1–5. genetics. Autoimmun Rev 2018;17:1065–6.
[28] Brix TH, Kyvik KO, Hegedus L. A population-based study of chronic autoimmune [58] Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and im-
hypothyroidism in Danish twins. J Clin Endocrinol Metab 2000;85:536–9. plications. Nat Rev Genet 2012;13:97–109.
[29] Huang CY, Chang TY, Chu CC, Lo FS, Ting WH, Lin CH, et al. The HLA-B gene and [59] Dik S, Scheepers PT, Godderis L. Effects of environmental stressors on histone
Hashimoto disease in Han Chinese children: a case-control and family-based study. modifications and their relevance to carcinogenesis: a systematic review. Crit Rev
Tissue Antigens 2012;80:431–6. Toxicol 2012;42:491–500.
[30] Ueda S, Oryoji D, Yamamoto K, Noh JY, Okamura K, Noda M, et al. Identification [60] Turner M, Galloway A, Vigorito E. Noncoding RNA and its associated proteins as
of independent susceptible and protective HLA alleles in Japanese autoimmune regulatory elements of the immune system. Nat Immunol 2014;15:484–91.
thyroid disease and their epistasis. J Clin Endocrinol Metab 2014;99:E379–83. [61] Ranzani V, Rossetti G, Panzeri I, Arrigoni A, Bonnal RJ, Curti S, et al. The long
[31] Jia X, Wang B, Yao Q, Li Q, Zhang J. Variations in CD14 gene are associated with intergenic noncoding RNA landscape of human lymphocytes highlights the reg-
autoimmune thyroid diseases in the chinese population. Front Endocrinol ulation of T cell differentiation by linc-MAF-4. Nat Immunol 2015;16:318–25.
(Lausanne) 2018;9:811. [62] Hewagama A, Richardson B. The genetics and epigenetics of autoimmune diseases.
[32] Ji R, Feng Y, Zhan WW. Updated analysis of studies on the cytotoxic T-lymphocyte- J Autoimmun 2009;33:3–11.
associated antigen-4 gene A49G polymorphism and Hashimoto’s thyroiditis risk. [63] Tomer Y. Mechanisms of autoimmune thyroid diseases: from genetics to epige-
Genet Mol Res 2013;12:1421–30. netics. Annu Rev Pathol 2014;9:147–56.
[33] Zaaber I, Mestiri S, Marmouch H, Mahjoub S, Abid N, Hassine M, et al. [64] Limbach M, Saare M, Tserel L, Kisand K, Eglit T, Sauer S, et al. Epigenetic profiling
Polymorphisms in TSHR and IL1RN genes and the risk and prognosis of in CD4+ and CD8+ T cells from Graves' disease patients reveals changes in genes
Hashimoto’s thyroiditis. Autoimmunity 2014;47:113–8. associated with T cell receptor signaling. J Autoimmun 2016;67:46–56.
[34] Yan N, Yu YL, Yang J, Qin Q, Zhu YF, Wang X, et al. Association of interleukin-17A [65] McLachlan SM, Rapoport B. Breaking tolerance to thyroid antigens: changing
and -17F gene single-nucleotide polymorphisms with autoimmune thyroid dis- concepts in thyroid autoimmunity. Endocr Rev 2014;35:59–105.
eases. Autoimmunity 2012;45:533–9. [66] Smith TJ, Hegedus L. Graves' disease. N Engl J Med 2016;375:1552–65.
[35] Saitoh O, Nagayama Y. Regulation of Graves’ hyperthyroidism with naturally oc- [67] Ehlers M, Thiel A, Bernecker C, Porwol D, Papewalis C, Willenberg HS, et al.
curring CD4+CD25+ regulatory T cells in a mouse model. Endocrinology Evidence of a combined cytotoxic thyroglobulin and thyroperoxidase epitope-
2006;147:2417–22. specific cellular immunity in Hashimoto’s thyroiditis. J Clin Endocrinol Metab
[36] Hu Y, Zhang L, Chen H, Liu X, Zheng X, Shi H, et al. Analysis of regulatory T cell 2012;97:1347–54.
subsets and their expression of Helios and PD-1 in patients with Hashimoto [68] Kotani T, Aratake Y, Hirai K, Fukazawa Y, Sato H, Ohtaki S. Apoptosis in thyroid
thyroiditis. Int J Endocrinol 2019;2019:5368473. tissue from patients with Hashimoto’s thyroiditis. Autoimmunity 1995;20:231–6.
[37] Gonzalez-Aguilera B, Betea D, Lutteri L, Cavalier E, Geenen V, Beckers A, et al. [69] Roldan J Cardenas, Amaya-Amaya J, la Hoz J Castellanos-de, Giraldo-Villamil J,
Conversion to Graves disease from Hashimoto thyroiditis: a study of 24 patients. Montoya-Ortiz G, Cruz-Tapias P, et al. Autoimmune thyroid disease in rheumatoid
Arch Endocrinol Metab 2018;62:609–14. arthritis: a global perspective. Arthritis 2012;2012:864907.
[38] Santaguida MG, Gatto I, Mangino G, Virili C, Stramazzo I, Fallahi P, et al. BREG [70] MacDonald TT. Suppressor T cells, rebranded as regulatory T cells, emerge from
cells in Hashimoto's thyroiditis isolated or associated to further organ-specific the wilderness bearing surface markers. Gut 2002;51:311–2.
autoimmune diseases. Clin Immunol 2017;184:42–7. [71] Shevach EM. Suppressor T cells: rebirth, function and homeostasis. Curr Biol
[39] Ferrari SM, Ragusa F, Paparo SR, Nasini F, Nardi M, Franceschini SS, et al. 2000;10:R572–5.
Differential modulation of CXCL8 versus CXCL10, by cytokines, PPAR-gamma, or [72] Pan D, Shin YH, Gopalakrishnan G, Hennessey J, De Groot LJ. Regulatory T cells in
PPAR-alpha agonists, in primary cells from Graves' disease and ophthalmopathy. Graves’ disease. Clin Endocrinol (Oxf) 2009;71:587–93.
Autoimmun Rev 2019;18:673–8. [73] Mao C, Wang S, Xiao Y, Xu J, Jiang Q, Jin M, et al. Impairment of regulatory
[40] Pirola I, Rotondi M, Cristiano A, Maffezzoni F, Pasquali D, Marini F, et al. Selenium capacity of CD4+CD25+ regulatory T cells mediated by dendritic cell polariza-
supplementation in patients with subclinical hypothyroidism affected by auto- tion and hyperthyroidism in Graves’ disease. J Immunol 2011;186:4734–43.
immune thyroiditis: results of the SETI study. Endocrinol Diabetes Nutr [74] Gottschalk RA, Corse E, Allison JP. Expression of Helios in peripherally induced
2020;67(1):28–35. Foxp3+ regulatory T cells. J Immunol 2012;188:976–80.
[41] Krysiak R, Kowalcze K, Okopien B. Selenomethionine potentiates the impact of [75] Dai S, Jia R, Zhang X, Fang Q, Huang L. The PD-1/PD-Ls pathway and autoimmune
vitamin D on thyroid autoimmunity in euthyroid women with Hashimoto's thyr- diseases. Cell Immunol 2014;290:72–9.
oiditis and low vitamin D status. Pharmacol Rep 2019;71:367–73. [76] Stassi G, De Maria R. Autoimmune thyroid disease: new models of cell death in
[42] Santos LR, Duraes C, Mendes A, Prazeres H, Alvelos MI, Moreira CS, et al. A autoimmunity. Nat Rev Immunol 2002;2:195–204.
polymorphism in the promoter region of the selenoprotein S gene (SEPS1) con- [77] Weetman AP. Cellular immune responses in autoimmune thyroid disease. Clin
tributes to Hashimoto’s thyroiditis susceptibility. J Clin Endocrinol Metab Endocrinol (Oxf) 2004;61:405–13.
2014;99:E719–23. [78] Walsh JP, Bremner AP, Feddema P, Leedman PJ, Brown SJ, O'Leary P. Thyrotropin
[43] Wiersinga WM. clinical relevance of environmental factors in the pathogenesis of and thyroid antibodies as predictors of hypothyroidism: a 13-year, longitudinal
autoimmune thyroid disease. Endocrinol Metab (Seoul) 2016;31:213–22. study of a community-based cohort using current immunoassay techniques. J Clin
[44] Aghini Lombardi F, Fiore E, Tonacchera M, Antonangeli L, Rago T, Frigeri M, et al. Endocrinol Metab 2010;95:1095–104.
The effect of voluntary iodine prophylaxis in a small rural community: the [79] Hamano H, Kawa S, Horiuchi A, Unno H, Furuya N, Akamatsu T, et al. High serum
Pescopagano survey 15 years later. J Clin Endocrinol Metab 2013;98:1031–9. IgG4 concentrations in patients with sclerosing pancreatitis. N Engl J Med
[45] Toulis KA, Anastasilakis AD, Tzellos TG, Goulis DG, Kouvelas D. Selenium 2001;344:732–8.

8
M. Ralli, et al. Autoimmunity Reviews 19 (2020) 102649

[80] Ajjan RA, Weetman AP. Cytokines in thyroid autoimmunity. Autoimmunity encephalitis. Clin Microbiol Infect 2019;25:431–6.
2003;36:351–9. [113] Esposito S, Principi N, Calabresi P, Rigante D. An evolving redefinition of auto-
[81] Trifari S, Kaplan CD, Tran EH, Crellin NK, Spits H. Identification of a human helper immune encephalitis. Autoimmun Rev 2019;18:155–63.
T cell population that has abundant production of interleukin 22 and is distinct [114] Oblitas RL Caparo. Autoimmune encephalitis. New diagnosis for an old condition.
from T(H)-17, T(H)1 and T(H)2 cells. Nat Immunol 2009;10:864–71. Medicina (B Aires) 2018;78(Suppl. 2):88–93.
[82] Figueroa-Vega N, Alfonso-Perez M, Benedicto I, Sanchez-Madrid F, Gonzalez- [115] Yoneda M, Fujii A, Ito A, Yokoyama H, Nakagawa H, Kuriyama M. High pre-
Amaro R, Marazuela M. Increased circulating pro-inflammatory cytokines and valence of serum autoantibodies against the amino terminal of alpha-enolase in
Th17 lymphocytes in Hashimoto’s thyroiditis. J Clin Endocrinol Metab Hashimoto's encephalopathy. J Neuroimmunol 2007;185:195–200.
2010;95:953–62. [116] McLachlan SM, Rapoport B. Why measure thyroglobulin autoantibodies rather
[83] Ruggeri RM, Saitta S, Cristani M, Giovinazzo S, Tigano V, Trimarchi F, et al. Serum than thyroid peroxidase autoantibodies? Thyroid 2004;14:510–20.
interleukin-23 (IL-23) is increased in Hashimoto’s thyroiditis. Endocr J [117] Rose NR. Prediction and prevention of autoimmune disease: a personal perspec-
2014;61:359–63. tive. Ann N Y Acad Sci 2007;1109:117–28.
[84] Cui X, Liu Y, Wang S, Zhao N, Qin J, Li Y, et al. Circulating exosomes activate [118] Hu ZY Fengqiu, Ma Buyun, Jiang Yong, Huang Hui. The impact of concurrent
dendritic cells and induce unbalanced CD4+ T cell differentiation in Hashimoto Hashimoto thyroiditis on thyroid nodule cytopathology assessed by ultrasound-
thyroiditis. J Clin Endocrinol Metab 2019;104(10):4607–18. guided fine-needle aspiration cytology. Postgrad Med 2020:1–6. (2020 Mar
[85] Ragusa PF Francesca, Elia Giusy, Gonnella Debora, Paparo Sabrina Rosaria, Giusti 12[Online ahead of print]).
Claudia, Churilov Leonid P, et al. Hashimotos’ thyroiditis: epidemiology, patho- [119] Harvey AM, Truong LD, Mody DR. Diagnostic pitfalls of Hashimoto’s/lymphocytic
genesis, clinic and therapy. Best Pract. Res. Clinical Endocrinol. Metabol. thyroiditis on fine-needle aspirations and strategies to avoid overdiagnosis. Acta
2019;13(6):612–32. (Aug 2018). Cytol 2012;56:352–60.
[86] Ferrari SM, Fallahi P, Ruffilli I, Elia G, Ragusa F, Benvenga S, et al. The association [120] Anderson L, Middleton WD, Teefey SA, Reading CC, Langer JE, Desser T, et al.
of other autoimmune diseases in patients with Graves' disease (with or without Hashimoto thyroiditis: Part 2, sonographic analysis of benign and malignant no-
ophthalmopathy): review of the literature and report of a large series. Autoimmun dules in patients with diffuse Hashimoto thyroiditis. Am J Roentgenol
Rev 2019;18:287–92. 2010;195:216–22.
[87] Akamizu T, Amino N. Hashimoto's thyroiditis. In: Feingold KR, Anawalt B, Boyce [121] Wiersinga WM. Thyroid hormone replacement therapy. Horm Res 2001;56(Suppl.
A, Chrousos G, Dungan K, Grossman A, ... Wilson DP, editors. Endotext, South 1):74–81.
Dartmouth (MA). 2000. [122] Topliss DJ. Clinical update in aspects of the management of autoimmune thyroid
[88] Giordano C, Stassi G, De Maria R, Todaro M, Richiusa P, Papoff G, et al. Potential diseases. Endocrinol Metab (Seoul) 2016;31:493–9.
involvement of Fas and its ligand in the pathogenesis of Hashimoto’s thyroiditis. [123] Watanabe T, Maruyama M, Ito T, Fujinaga Y, Ozaki Y, Maruyama M, et al. Clinical
Science 1997;275:960–3. features of a new disease concept, IgG4-related thyroiditis. Scand J Rheumatol
[89] Katz SM, Vickery Jr. AL. The fibrous variant of Hashimoto’s thyroiditis. Hum 2013;42:325–30.
Pathol 1974;5:161–70. [124] Abbott RD, Sadowski A, Alt AG. Efficacy of the autoimmune protocol diet as part
[90] Ahmed R, Al-Shaikh S, Akhtar M. Hashimoto thyroiditis: a century later. Adv Anat of a multi-disciplinary, supported lifestyle intervention for Hashimoto's thyroiditis.
Pathol 2012;19:181–6. Cureus 2019;11:e4556.
[91] Hennessey JV. Clinical review: Riedel’s thyroiditis: a clinical review. J Clin [125] Carayanniotis G. Recognition of thyroglobulin by T cells: the role of iodine.
Endocrinol Metab 2011;96:3031–41. Thyroid 2007;17:963–73.
[92] BM R. Die chronische zur Bildung eisenharter Tumoren führende Entzündung der [126] Liontiris MI, Mazokopakis EE. A concise review of Hashimoto thyroiditis (HT) and
Schilddruese. Verh Ges Chir 1896;25:101–5. the importance of iodine, selenium, vitamin D and gluten on the autoimmunity
[93] Hay ID. Thyroiditis: a clinical update. Mayo Clin Proc 1985;60:836–43. and dietary management of HT patients. Points that need more investigation. Hell
[94] Stone JH, Khosroshahi A, Deshpande V, Chan JK, Heathcote JG, Aalberse R, et al. J Nucl Med 2017;20:51–6.
Recommendations for the nomenclature of IgG4-related disease and its individual [127] Kohrle J. Selenium and the thyroid. Curr Opin Endocrinol Diabetes Obes
organ system manifestations. Arthritis Rheum 2012;64:3061–7. 2013;20:441–8.
[95] Zaletel K, Gaberscek S. Hashimoto’s thyroiditis: from genes to the disease. Curr [128] Eskes SA, Endert E, Fliers E, Birnie E, Hollenbach B, Schomburg L, et al. Selenite
Genom 2011;12:576–88. supplementation in euthyroid subjects with thyroid peroxidase antibodies. Clin
[96] Tamaki H, Amino N, Kimura M, Hidaka Y, Takeoka K, Miyai K. Low prevalence of Endocrinol (Oxf) 2014;80:444–51.
thyrotropin receptor antibody in primary hypothyroidism in Japan. J Clin [129] Wichman J, Winther KH, Bonnema SJ, Hegedus L. Selenium supplementation
Endocrinol Metab 1990;71:1382–6. significantly reduces thyroid autoantibody levels in patients with chronic auto-
[97] Kakudo K, Li Y, Taniguchi E, Mori I, Ozaki T, Nishihara E, et al. IgG4-related immune thyroiditis: a systematic review and meta-analysis. Thyroid
disease of the thyroid glands. Endocr J 2012;59:273–81. 2016;26:1681–92.
[98] Zhang J, Zhao L, Gao Y, Liu M, Li T, Huang Y, et al. A classification of Hashimoto’s [130] Bruscolini A, Sacchetti M, La Cava M, Nebbioso M, Iannitelli A, Quartini A, et al.
thyroiditis based on immunohistochemistry for IgG4 and IgG. Thyroid Quality of life and neuropsychiatric disorders in patients with Graves' Orbitopathy:
2014;24:364–70. current concepts. Autoimmun Rev 2018;17:639–43.
[99] Li XW Yaqiong, Liu Zhiyan, Ma Jiwei, Xiaoyan, Lin YQ, Nishihara Eijun, et al. [131] Winther KH, Wichman JE, Bonnema SJ, Hegedus L. Insufficient documentation for
Hashimoto’s THYROIDITIS WITH INCREASed IgG4‐positive plasma cells: using clinical efficacy of selenium supplementation in chronic autoimmune thyroiditis,
thyroid‐specific diagnostic criteria may identify early‐phase IgG4 thyroiditis. 30 based on a systematic review and meta-analysis. Endocrine 2017;55:376–85.
(2). 2019. p. 251–61. (Feb 2020). [132] Roehlen N, Doering C, Hansmann ML, Gruenwald F, Vorlaender C, Bechstein WO,
[100] Caturegli P, De Remigis A, Rose NR. Hashimoto thyroiditis: clinical and diagnostic et al. Vitamin D, FOXO3a, and Sirtuin1 in Hashimoto's thyroiditis and differ-
criteria. Autoimmun Rev 2014;13:391–7. entiated thyroid cancer. Front Endocrinol (Lausanne) 2018;9:527.
[101] Biondi B, Cappola AR, Cooper DS. Subclinical hypothyroidism: a review. JAMA [133] Chao YZ Guanqun, Fang Lizheng. Correlation between Hashimoto’s thyr-
2019;322:153–60. oiditis–related thyroid hormone levels and 25-hydroxyvitamin D. Front
[102] Livingston EH. Subclinical hypothyroidism. JAMA 2019;322:180. Endocrinol (Lausanne) 2020:11. (4 2020 Feb 14).
[103] Luis EG-C, Valderrama-Hinds M, Hernández Evelyn, Agostini María I, Reyes- [134] Gan T, Randle RW. The role of surgery in autoimmune conditions of the thyroid.
Morales Omar R, Fung Liliana, et al. Prevalence of undifferentiated inflammatory Surg Clin North Am 2019;99:633–48.
arthropathy in patients with Hashimoto’s thyroiditis in an endocrinology clinic. 22 [135] Mack LA, Pasieka JL. An evidence-based approach to the treatment of thyroid
(11). 2019. p. 1985–9. (Nov 2019). lymphoma. World J Surg 2007;31:978–86.
[104] Giuffrida GBG, Campennì A, Giovinazzo S, Keller KP, Alibrandi A, Roberts WN, [136] Davies TF. Is thyroid transplantation on the distant horizon? Thyroid
et al. Non-specific rheumatic manifestations in patients with Hashimoto’s thyr- 2013;23:139–41.
oiditis: a pilot cross-sectional study. J Endocrinol Invest 2020;43(1):87–94. (Jan [137] Dailey ME, Lindsay S, Skahen R. Relation of thyroid neoplasms to Hashimoto
2020). disease of the thyroid gland. AMA Arch Surg 1955;70:291–7.
[105] Amino N, Tada H, Hidaka Y. Postpartum autoimmune thyroid syndrome: a model [138] Jankovic B, Le KT, Hershman JM. Clinical Review: Hashimoto’s thyroiditis and
of aggravation of autoimmune disease. Thyroid 1999;9:705–13. papillary thyroid carcinoma: is there a correlation? J Clin Endocrinol Metab
[106] Peng RH-EC Carol Chiung-Hui, Pennant Majorie, Huang Huei-Kai, Munir KM. A 2013;98:474–82.
literature review of painful hashimoto thyroiditis: 70 published cases in the past [139] Mukasa K, Noh JY, Kunii Y, Matsumoto M, Sato S, Yasuda S, et al. Prevalence of
70 years. Endocr Soc 2019;4(2):bvz008. malignant tumors and adenomatous lesions detected by ultrasonographic
[107] Pinedo-Torres I, Paz-Ibarra JL. Current knowledge on Hashimoto's encephalo- screening in patients with autoimmune thyroid diseases. Thyroid 2011;21:37–41.
pathy: a literature review. Medwave 2018;18:e7298. [140] Boi F, Pani F, Mariotti S. Thyroid autoimmunity and thyroid cancer: review fo-
[108] Brain L, Jellinek EH, Ball K. Hashimoto’s disease and encephalopathy. Lancet cused on cytological studies. Eur Thyroid J 2017;6:178–86.
1966;2:512–4. [141] Sulaieva OS Oksana, Shapochka Dmytro, Belemets Nataliia, Nechay Oleksandr,
[109] Schiess N, Pardo CA. Hashimoto’s encephalopathy. Ann N Y Acad Sci Chereshneva Yelizaveta, Tsomartova Dibakhan, et al. Hashimoto's thyroiditis at-
2008;1142:254–65. tenuates progression of papillary thyroid carcinoma: deciphering immunological
[110] Mattozzi LS Simone, Escudero Domingo, Ariño Helena, Armangue Thais, links. Eliyon 2020;6(1):e03077. (2020 Jan 8).
Simabukuro Mateus, Iizuka Takahiro, et al. Hashimoto encephalopathy in the 21st [142] Nam YJ, Kim BH, Lee SK, Jeon YK, Kim SS, Jung WJ, et al. Co-occurrence of
century. 94 (2). 2020. p. e217–24. (2020 Jan 14). papillary thyroid carcinoma and mucosa-associated lymphoid tissue lymphoma in
[111] Ercoli T, Defazio G, Muroni A. Cerebellar syndrome associated with thyroid dis- a patient with long-standing hashimoto thyroiditis. Endocrinol Metab (Seoul)
orders. Cerebellum 2019;18(5):932–40. 2013;28:341–5.
[112] Lascano AM, Vargas MI, Lalive PH. Diagnostic tools for immune causes of

You might also like