Endokrynologia Polska 4/2017-The role of selenium in thyroid gland pathophysiology

PRACE POGLĄDOWE/REVIEWS

The role of selenium in thyroid gland pathophysiology

Michał Stuss1, 2, Marta Michalska-Kasiczak1, Ewa Sewerynek1, 2

1Department of Endocrine Disorders and Bone Metabolism, Chair of Endocrinology, Medical University of Lodz, Poland

2The Outpatient Clinic of Endocrinology and Osteoporosis Therapy of the Regional Centre of Menopause and Osteoporosis of the Military Teaching Hospital in Lodz, Poland


Disclosure of conflicts of interest

All authors certify that they have no financial interests such as employment, stock ownership, honoraria, or paid expert testimony, as well as any personal relationships, academic competition, and intellectual passion, which may inappropriately influence their actions.

All funding sources supporting the work and all institutional or corporate affiliations are acknowledged in a footnote.

All authors have had full access to all the data in the study (if applicable) and thereby accept full responsibility for the integrity of the data and the accuracy of the data analysis.


Abstract

It is now assumed that proper functioning of the thyroid gland (TG), beside iodine, requires also a number of elements, including selenium, iron, zinc, copper, and calcium. In many cases, only an adequate supply of one of these microelements (e.g. iodine) may reveal symptoms resulting from deficits of other microelements (e.g. iron or selenium).

Selenium is accounted to the trace elements of key importance for homeostasis of the human system, in particular, for the proper functioning of the immune system and the TG. Results of epidemiological studies have demonstrated that selenium deficit may affect as many as one billion people in many countries all over the world. A proper sequence of particular supplementations is also worth emphasising for the significant correlations among the supplemented microelements. For example, it has been demonstrated that an excessive supplementation of selenium may enhance the effects of iodine deficit in endemic regions, while proper supplementation of selenium in studied animals may alleviate the consequences of iodine excess, preventing destructive-inflammatory lesions in the TG.

This paper is a summary of the current knowledge on the role of selenium in the functionality of the TG. (Endokrynol Pol 2017; 68 (4): 440–454)

Key words: selenium, thyroid gland, supplementation

Michał Stuss, M.D., Ph.D, Department of Endocrine Disorders and Bone Metabolism, Chair of Endocrinology , Medical University in Lodz Żeligowskiego St. 7/9, 90–752 Łódź, Poland, tel./fax: +48 42 63 93 127, e-mail: mstuss@wp.pl

Introduction

Selenium is accounted to the trace elements of key importance for homeostasis of the human system, in particular, for the proper functioning of the immune system [1, 2]. Results of epidemiological studies have demonstrated that selenium deficit may affect as many as one billion people in many countries all over the world [3].

Selenium doses and sources

Following the recommendations of the World Health Organisation (WHO), supplementation of this microelement should not exceed 70 μg/day, while its daily intake above 400–700 μg may exert toxic effects [4]. Following the guidelines developed by the Food and Nutrition Board of the National Academy of Science, men should consume 40–70 μg and women 45–55 μg (60–70 μg during gestation and lactation periods) daily [5, 6]. According to population studies, the mean daily intake of selenium at European countries is 20–70 μg [1, 5, 7–9] and 20–59 μg in Poland [10, 11]. In turn, following the 30-day observation of other researchers, a typical diet of an average American provides from 90 to 168 μg of this microelement daily [12]. It is estimated that, in some regions of China, selenium consumption reaches the level of 5 mg/day [10, 13]. In order to ensure an adequately broad safety margin, in some sources, we may come across the term reference selenium dose (RSD). According to Patterson and Levander the RSD is 350 μg/day, which corresponds to the supplementation level of approximately 5 μg/kg of human body weight. According to the authors’ assumption, the daily dose of 150 μg should be delivered with food, while the remaining 200 μg may be provided by supplements [14].

Thus, considering the application of selenium supplementation, one should be aware of its narrow therapeutic index and of the possibility of adverse effects, not only when diet supplements are used but also when selenium-rich foodstuffs are consumed [15]. It has been demonstrated that consumption of large volumes of fruit from certain plants, including, among others, the species of Lecythis ollaria, may cause hair loss, nausea, vomiting, and diarrhoea [16]. The selenium content may differ in the same foodstuffs but originating from different regions, which is associated with the climate, the volumes of this microelement in soil, and the ability of plants to accumulate it, which is directly translated into higher links of the food chains [17]. A high selenium content is, among others, characteristic for protein-rich foods, nuts, in particular Brazil nuts (more than 6 μg/g of the product), and fungi, including mushrooms and yeasts (selenium content in the latter amounts to as much as 3 mg/g). As a rule, fruits and vegetables contain small volumes of this microelement (often below 0.5 μg/g of the product), which is associated with the high water content and low protein content. However, some of them may be good sources of selenium – among others, garlic, broccoli, cabbage, cauliflower, and kohlrabi [5].

Selenium, in its natural substance, most often takes an organic form of selenomethionine, methylselenocysteine, or γ-glutamylmethylselenocysteine. In turn, enriched products and supplements, beside organic compounds (selenoamino acids), often contain inorganic salts, especially of selenium (IV) [18]. It is assumed that organic selenium is more easily digestible, while yeasts provide an excellent substrate for production of its supplements [19]. It has been demonstrated that, on average, 85–95% of the dose of organic selenium compounds undergo intestinal absorption, while it is only 10% in the case of inorganic compounds. After it permeates into blood, selenium binds to erythrocytes and to globulins and albumins in plasma, being, later on, transported in this way to various tissues and organs. Its highest volumes are found in the liver, the kidneys, the testes, the thyroid gland (TG), the pancreas, and in the pituitary – also in the hair and nails. The muscles are characterised by the highest selenium content (there may even be a half of the systemic selenium stock). It is possible to evaluate the degree of selenium supply to the organism by assaying the concentrations of these microelement-containing proteins, including, among others, P1 selenoproteins (SPP1) or glutathione peroxidase. Taking into account the significant role of selenoproteins in maintaining systemic homeostasis, it is recommended that the concentrations of these proteins are as high as possible. It has been determined that glutathione peroxidase 3 achieves the plateau status at concentration of approximately 125 μU/L, which reflects selenium concentration in serum at approximately 1 μM/L (approximately 79 μg/L), which, in turn, is ensured by the mean daily supplementation of 125 μg of this microelement [20–24]. The highest concentrations of P selenoprotein were obtained at serum selenium levels from 110 to 125 μg/L [25]. In the literature, one may come across various reference ranges of selenium concentration in serum, depending on the geographic region, the country, as well as the ethnic group, which is related to diet.

The issue of selenium deficit

The effects of selenium deficit may be observed in the geographic regions with low selenium soils, such as Siberia and a major part of China [16, 26, 27]. The symptoms of selenium deficit concern many organs and systems and are manifested by decreased activities, as well as by impaired functions and structures that are associated with this microelement, i.e. the so-called selenoproteins [28]. The adverse effects caused by selenium deficit and most frequently described in literature include dilated cardiomyopathy (Keshan disease) and endemic osteoarthropathy (Kashin-Beck disease) [28–30].

The incidence of Keshan disease is highest in women of reproductive age and in children aged up to 10 years [30]. In turn, Kashin-Beck disease is characterised by changes that resemble rheumatoid arthritis, including shortening of fingers and toes and growing disturbances. Enhanced oxidative processes bring about lesions and necrosis of cartilages and cause bone deformations. The disease affects most often children aged up to 13 years [7, 29]. Iodine deficit contributes to the occurrence/enhancement of Kashin-Beck disease symptoms [29, 31].

The following other significant effects of selenium deficit are worth mentioning: heart failure, arrhythmia, strokes, sudden infant death syndrome, infertility in men, prostate cancer, nephropathy, and exacerbation of symptoms of diseases, both associated with the immune system and autoimmunological; among others, thyroid disease [26, 32–34].

The issue of selenium excess

Analogously to selenium deficit, also its excess is most often observed in geographic areas with high-selenium content in soil; among others, in certain regions of India.

High selenium doses induce an excessive production of free radicals, causing DNA damage. In addition, selenium excess inactivates the proteins that are responsible for repair of damaged DNA, demonstrating high affinity to their thiol groups [35].

Excessive consumption of selenium causes classical symptoms of intoxication: systemic weakness, nausea, vomiting, and diarrhoea; moreover, neurological disorders may occur, e.g. ataxia [7, 36]. Chronically increased selenium supplementation induces a medical condition called selenosis, which is manifested by: liver damage, haematopoiesis disorders, hair loss, infertility, rash, nail fractures, and an unpleasant (resembling garlic) mouth odour, as well as various neurological disorders [7, 27, 37]. The negative consequences of selenium excess on the endocrine system should also be mentioned, including an impaired synthesis of thyroid hormones (TH), growth hormone, and insulin-like growth factor-1 (IGF-1) [7]. It has also been demonstrated that its excessive supplementation may increase the risk for diabetes mellitus of type 2 [38]. Moreover, inhalation of selenium compounds may contribute to the occurrence of chemical bronchitis and pneumonia (even pulmonary oedema), eye irritation, and headaches. The clinical data on doses that may cause adverse effects are rather inconclusive. It was shown in a study of the Chinese population that an increased incidence of selenosis is associated with doses of this microelement above 850 μg/day [26]. In turn, regarding patients with rheumatoid arthritis, daily supplementation with 600 μg of selenium from yeasts reduced arthritic pain but without any symptoms of intoxication with selenium [39]. In another study, patients receiving a daily dose of selenium of 700 μg tolerated it very well [40]. In the opinion of many authors, the major benefits from selenium supplementation are observed in persons with a decreased supplementation rate of this microelement. In other cases, one should take under consideration an increased risk of the occurrence of metabolic disorders [38, 41].

How selenium works

As already mentioned, selenium atoms are part of the so-called selenoproteins and antioxidative enzymes, including, among others: glutathione peroxidases (GPs), thioredoxin reductases (TRRs), iodothyronine deiodinases (DIOs), selenoprotein P1 (SPP1), and selenoprotein W. GP was the first described selenoprotein; however, eight isoforms of the enzyme have so far been identified, depending on their structure and localisation [42]. The main task of GP is the protection of lipid membranes against oxidative stress. They catalyse the reactions of hydrogen peroxide (H2O2) reduction and of organic peroxide (ROOH) reduction, with production of selenous acid, as an intermediate product, or of some appropriate alcohols [33].

Three types of DIOs are distinguished. They play a key role in the activation of the other forms of TH to the most active triiodothyronine (T3) (type 1 and 2) and, in addition, in the inactivation of thyroxin and T3 (type 3) [43–46]. The above-mentioned group of enzymes plays then a key role in the proper functioning of the TG, as well as in its development [47]. In the case of selenium deficit, iodine metabolism is impaired, which supports various disorders in the synthesis of TH. It may have some impact on the clinical state and the general wellbeing of affected patients [34].

TRRs are biochemically responsible for the reaction of oxidised thioredoxin reduction. The enzymes of this group constitute an electron donor for other important oxidoreductive enzymes, e.g. ribonucleotide reductase and thioredoxin peroxidase. The thioredoxins alone reveal properties of apoptosis inhibitors, growth factors, and hydroperoxidase reducers. In addition, TRRs may reduce oxidised glutathione, dehydroascorbic acid, vitamin K, lipid peroxides, and hydrogen peroxide [48].

SPP1, besides its role of selenium storage site and transporter along the system, acts as a heavy metal chelator. It creates non-toxic complexes with them and may also exert anti-tumour activity [49, 50]. Selenoprotein W is responsible for metabolism of the muscular system, and N for muscle tissue development [3, 51]. It has been shown that certain mutations in the gene for selenoprotein N can cause multiminicore myopathy [51]. Other selenoproteins (S, M) were also identified [47, 52, 53], but their role is not yet well understood.

It mentioned earlier, selenium can control the functions of the immune system [1, 2]. It has been demonstrated that the above-mentioned element can stimulate the synthesis of antibodies, especially of IgG and IgM, and activate lymphocytes T and macrophages [38]. It has also been proven that selenium-containing compounds may suppress the transformation of HIV infection to fully symptomatic AIDS [54]. Selenium also exerts antiviral and antibacterial effects and inhibits the progression of various forms of viral hepatitis. It may also prevent infections with RNAs of certain viruses, among others, of the Ebola virus [1, 2, 55, 56].

The postulated anti-tumour properties of selenium are largely based on its already mentioned antioxidative properties. The advantageous effects of selenium on the activation of NK lymphocytes should be mentioned as well [50]. Thus far, the positive properties of selenium have been described in the context of colonic, prostatic, and pulmonary carcinoma [52]. There are also reports on the role of selenium in the conductivity of impulses in the central nervous system (CNS) [1, 57]. Moreover, the above-mentioned microelement may prevent diabetes mellitus, cardiovascular diseases, and infertility [26, 32].

Selenium and the thyroid gland

Thyroid diseases are considered to be among the most frequent medical conditions in general, and, decisively, they are the most frequent problems of endocrine character with which patients visit their GPs [58–60], while L-thyroxin is in the top ten of the most frequently prescribed and taken medicinal products [58, 61].

It is now assumed that proper functioning of the TG, besides iodine, also requires a number of elements, including selenium, iron, zinc, copper, and calcium [44, 62–67]. In many cases, only an adequate supply of one of these microelements (e.g. iodine) can reveal symptoms resulting from deficits of other microelements (e.g. iron or selenium) [62, 63, 66]. A proper sequence of particular supplementations is also worth emphasising for the significant correlations among the supplemented microelements. For example, it has been demonstrated that excessive supplementation of selenium may enhance the effects of iodine deficit in endemic regions, while proper supplementation of selenium in studied animals may alleviated the consequences of iodine excess, preventing destructive-inflammatory lesions in the TG [44, 68–72]. The TG, similarly to the testes and the brain, and unlike other organs in the human body, is characterised by high selenium content, despite its deficit in the system [44]. Studies carried out in patients with mutations of selenocysteine insertion sequence (SECIS) binding protein 2, may provide good proof of the significant role of selenium on the thyroid metabolism. This protein plays a significant role in the synthesis of selenoproteins. It has been observed that various mutations of the gene for the above-mentioned protein are, among others, responsible for the characteristic hormonal configuration: increased concentrations of TSH and FT4 and a decreased concentration of FT3 plus sensorineural hearing loss. Moreover, other, more characteristic features may occur, such as impaired development of the osseous system, myopathy, disturbances in CNS development, regarding the motor abilities, hypersensitivity to UV, and enhanced insulin sensitivity [73–76].

As already mentioned, all the DIOs, as well as other proteins, including GPs and TRRs, contain selenium atoms. A high activity of these enzymes has been demonstrated in the thyroid tissue; however, in order to maintain iodine metabolism, catalysed by deiodinases, and at an appropriate level, low activity of these enzymes will be enough, much lower than of other selenoproteins in the pathways of the metabolic turnover of proteins, fats, and amino acids [34, 77, 78]. Therefore, neither the concentration of selenium nor the concentrations of selenium-containing proteins directly influence the selenium content in the thyroid and the activity of selenoproteins contained in the TG [69, 79]. There is no data in the available literature concerning the measurable parameters that would unequivocally and reliably reflect thyroid supplementation with selenium.

A number of significant clinical data on the role of selenium supplementation in TG homeostasis were provided by studies carried out in Central Africa during the 1980s. Symptoms of cretinism with myxoedema were observed in the population living within that area, including mental retardation, growth suppression, and impaired puberty. The studies further confirmed that all those conditions had been associated with iodine and selenium deficits on one hand, and with the consumption of large quantities of foods containing goitrogens on the other [44, 80]. Negative additive effects, resulting from deficits of either microelement, have also been confirmed by the results of studies on animals [80, 81]. Most probably, it is the transforming growth factor β (TGF β) which is responsible for thyroid tissue damage and fibrosis resulting from selenium deficits. TGF β mediates the stimulation of destruction, caused by oxidative stress and initiated by high TSH concentrations [71]. In turn, appropriate selenium supplementation protects from the described processes, even when iodine supplementation is high, following its long-term deficit [68, 71, 72, 80]. According to the results of studies carried out in Zair, selenium supplementation without previous compensation of iodine deficit may deteriorate the patient’s thyroid functioning; therefore, previous iodine assays and supplementation, if necessary, are required [80]. An increased activity of deiodinases and an increased metabolism of TH, with their enhanced deiodination and iodine loss with urine, are assumed to be the mechanism of this phenomenon [44]. The above described clinical situation concerned a severe deficit of both microelements.

There are many hypotheses concerning the protective role of selenium in thyroid diseases. At present, it is assumed that proper selenium supplementation may [41]:

  1. decrease the expression of HLA-DR antigens on thyrocyte surface,
  2. cause a decrease in the concentrations of antithyroid antigen antibodies,
  3. control the lymphocyte B-dependent immunological response,
  4. inhibit the production of proinflammatory cytokines,
  5. reduce the synthesis of leukotrienes and prostaglandins,
  6. protect the thyroid against oxidative stress,
  7. optimise the synthesis and transport of TH via induction of selenoprotein synthesis (among others, p15 and S) [82–84].

In recent years, several selenium-containing analogues of thyrostatics have been produced, as well as of compounds that imitate the action mode of selenoproteins [85–88]. The anti-thyroid drugs that are currently available on the market may cause a number of unpleasant adverse effects [89]; therefore, a possibility of the synergistic effect of action on the suppression of not only the biosynthesis but also of the activation of TH – or their activation alone – may lead to a new, valuable therapeutic option for patients with hyperthyroidism of different aetiologies and will be better tolerated than already available anti-thyroid medications [90].

One of the first interventional studies in which the supplementation of various vitamins and microelements was applied, demonstrated that too small supplementation of selenium influenced the growth of the TG only in women [91]. Analogous results were obtained when a Danish population was examined, also finding out a reverse correlation between selenium concentrations and the number of focal changes in the TG [92]. Changes in serum selenium levels were also observed in other benign and malignant diseases of the TG [93]. As is common knowledge, the incidence of thyroid diseases is higher in women than in men [94]; however, no causes of this phenomenon have yet been adequately understood. An influence of female sex hormones is postulated, as well as the protective activity of factors that depend on the presence of chromosome Y or on the different mechanisms by which the immune systems function in both sexes. It is also worth emphasising that the advantageous effects of selenium in cases of thyroid diseases have been demonstrated in women, while in men it has been shown in other organs [83, 95–98].

Data that confirm the hypothesis of the beneficial selenium supplementation effects in the case of thyroid cancer are rather scarce and apply mainly to its papillary form [99–101]. In many studies on animal models, as well as in epidemiological and interventional surveys, beneficial effects were confirmed of normal or even increased serum selenium concentrations on the initiation, progression, and even metastases of various cancer types but not of thyroid cancer [50, 102]. The relationship between selenium supplementation and the incidence of thyroid cancer has been verified in many clinical studies (Table I); however, it has not been unequivocally confirmed, especially in the Polish population [103–111]. The access to mainly retrospective data, the relatively small groups of patients, and the short observation periods, as well as only single assays of selenium concentration levels, were all significant limitations in the performed studies. One should also consider the fact that low selenium concentrations may be not a cause but a result of some concomitant systemic diseases, also neoplastic, even in the effects of sustained chronic inflammation, which may impair the production of SPP1 in the liver, which, as is known, reflects the body’s supply with this microelement [112–115].

Table I. Short characteristics of the most important clinical trials, concerning the relationship between selenium and thyroid cancer

Study Studied group Study type and duration Description of intervention-group The most important, evaluated parameters The most important results
Glattre et al. Int J Epidemiol (1989) [103] Population of 172 patients (124 women and 48 men) Cross-sectional study Control group (n = 129)
Study group with thyroid carcinoma (n = 43)
Differences in serum selenium concentrations and their association with the risk of thyroid carcinoma Lower selenium concentrations in patients with thyroid carcinoma
Relative risks (RR) for thyroid carcinoma depending on selenium concentrations:
RR = 1 for ≥ 1.65 μmol/L
RR = 6.1 for 1.26–1.64 μmol/L
RR = 7.7 for ≥ 1.25 μmol/L
Kucharzewski et al. Biol Trace Elem Res (2002) [104] Population of 87 subjects (85 women and 2 men) Cross-sectional study Patients with benign thyroid diseases (41 women with nodular goitre, 18 with Graves’ disease, 7 with CAT)
21 patients with thyroid carcinoma (19 women and 2 men)
Differences in the concentrations of selenium in serum and in thyroid tissue Insignificant differences among the groups in serum selenium concentrations
Significantly lower selenium concentration in thyroid tissue – in the group of patients with thyroid carcinoma
Moncayo et al. BMC Endocr Disord (2008) [105] 1401 patients (1186 adults and 215 children) with benign thyroid diseases and thyroid carcinoma Cross-sectional study Groups concerning selenium analysis:
Control (n = 687)
Benign thyroid diseases (n = 550; 465 adults, 85 children)
Thyroid carcinoma (n = 164); 42 with follicular, 73 with papillary and 3 with anaplastic thyroid carcinoma
Differences in serum selenium concentrations and an evaluation of the correlation of this concentration with other measurable parameters Selenium concentration was significantly lower in:
  1. The population of patients with thyroid diseases vs. the control
  2. The subgroups with subacute and silent thyroiditis
  3. In the group of patients with follicular and papillary carcinoma
No significant correlations between selenium concentration and patient’s age, gender, BMI, scintigraphic and sonographic pictures of the thyroid gland, hormone concentrations and anti-thyroid antigen antibodies
Przybylik-Mazurek et al. Biol Trace Elem Res (2011) [106] Women with CAT, follicular and papillary thyroid carcinoma Cross-sectional study Control (n = 20)
Women with CAT (n = 17)
Patients with papillary carcinoma (n = 25)
Patients with follicular carcinoma (n = 13)
Differences in the concentrations of microlements in serum, including, among others, selenium and glutathione peroxidase (PG) 3 No significant differences among the groups in selenium and PG3 concentrations
Jonklaas et al. Thyroid (2013) [107] 65 patients qualified to the procedure of thyroidectomy (46 women and 19 men) Cross-sectional study Patients qualified to procedure for goitre occurrence (n = 17)
Patients qualified to procedure for thyroid cancer (n = 48; in histopathological diagnosis: 35 cases of papillary carcinoma and 9 cases of its follicular variant and 4 cases of follicular carcinoma)
Comparative evaluation of preoperative selenium and 25(OH) vit. D3 concentrations in serum and their correlation with the stage of the diseases severity
  1. Insignificant concentration differences among the groups
  2. Reverse, significant correlation between selenium concentration and carcinoma stage
O’Grady et al. PLoS One(2014) [108] 566398 patients qualified to NIH-AARP study (National Institute of Health American Association of Retired Persons The prospective study (initiated in 1995) In the studied population, 592 cases of carcinoma were identified (257 men and 335 women):
406 diagnoses of papillary carcinoma (164 cases in men and 242 in women),
113 diagnoses of follicular carcinoma (57 in men and 56 in women)
Evaluation of the correlation between the consumption of selenium, as well as of other microelements and vitamins on one hand and the incidence rate of thyroid carcinoma on the other No significant correlation was found between the quintiles of lower selenium consumption and the increased incidence of thyroid cancer
Shen et al. Biol Trace Elem Res (2015) [109] Metaanalysis of data from 8 clinical studies (4 concerning selenium). The total number of patients: 1291 Metaanalysis Metanalysis of data from studies, carried out in the following populations: Norwegian, Austrian and Polish Differences in the concentrations of selenium, copper and magnesium in serum
  1. Significantly lower selenium and magnesium concentrations and higher copper concentrations in patients with thyroid carcinoma
  2. An analysis in the subgroups confirmed the presence of lower selenium concentrations in the Norwegian and Austrian populations but not in the Polish one
Baltaci et al. Biol Trace Elem Res (2016) [110] 50 patients of both sexes (30 subjects with histopathological diagnosis of papillary thyroid cancer) Cross-sectional study Women with post-operative diagnosis of thyroid cancer (n = 15)
Men with post-operative diagnosis of thyroid cancer (n = 15)
Women with benign changes in histopathological evaluation
Men with benign changes in histopathological evaluation
Comparison of selenium and zinc concentrations in serum before, just after and 15 days after surgical procedure and in the thyroid tissue after operation
  1. Pre- and postoperative zinc and selenium concentrations in Group 1 and 2 were significantly lower in serum and higher in the thyroid tissue vs. the corresponding values in the control groups
  2. After 15 days from the surgical procedure, no significant differences were observed among the groups
Chung et al. Biol Trace Elem Res (2016) [111] Ninety-two (92) Korean women, qualified to the procedure of thyroidectomy Cross-sectional study Ninety-two (92) Korean women, qualified to the procedure of thyroidectomy and with post-operative, histopathological identification of papillary thyroid cancer Evaluation of the concentrations/ contents of cadmium, selenium and zinc in post-operative materials and of their correlations with the stage of thyroid cancer progression Acc. to TNM, cadmium, selenium and zinc concentrations were significantly higher in stages III and IV

CAT – chronic autoimmune thyroiditis

Selenium in autoimmune diseases of the thyroid gland

It is known that selenium affects the process of differentiation of T lymphocytes in such a way that its increasing supplementation may induce increased production of regulatory T lymphocytes and a decrease of the synthesis of anti-thyroid antigen antibodies and of lymphocyte infiltrations [116], while its deficit may enhance the activity of Th2 lymphocytes [117]. In addition, it has been demonstrated on an animal model of chronic autoimmune thyroiditis (CAT) that selenium supplementation suppresses the Th1-dependent immune responses, thus inhibiting the inflammatory response and the occurrence of destructive lesions in the TG [118]. Moreover, in blood from healthy males, supplemented with a selenium-containing agent, a lower number of NK lymphocytes was observed [119]. The presented results suggest beneficial effects of selenium on the autoimmune diseases of the TG [83, 120]. In women at risk of postpartum thyroiditis, proper supplementation of the microelement may provide a prophylactic effect against the disease, although the data from clinical studies still require additional confirmation [121, 122].

In addition, lower selenium concentrations were found in serum from patients with Graves’ disease and with hypothyroidism in the course of CAT. However, the results of studies on the correlation between the concentrations of anti-TPO, anti-TG, and anti-TSHR antibodies and selenium supplementation rates are much less conclusive [123]. In many interventional studies, no relationship was found between the time of selenium intake and selenium dose on one hand, and changes in the concentrations of TSH and TH on the other; however, most of the studies were not carried out in cases of severe selenium deficit [44, 98, 124, 125]. Therefore, the results of those studies may have also been a consequence of good mechanisms of supplementation with selenium of the TG and of the anterior pituitary, despite maintained systemic selenium deficit.

A number of prospective studies have also been run to evaluate the severity of symptoms resulting from identified autoimmune thyroid disease, as well as to assess the quality of patient life and TH production rates and concentration levels. The results of meta-analyses of the studies are often divergent and non-unequivocal. In one of the meta-analyses [123] its authors evaluated the results of four clinical studies assessing the effect of supplementation with sodium selenate (IV) and with selenomethionine in a daily dose of 200 μg on various end-points in patients with diagnosed CAT. In each of the three studies, where selenomethionine had been used, a significant reduction was found in the concentration of anti-TPO antibodies. That effect was not significant in the population, which had been supplemented with selenate; however, improved quality of patient life was reported by patients from that population [123]. Despite all the encouraging beneficial and supplementation results, the authors of the above described statistical analysis recommend a precautionary approach to the issue for the high heterogeneity and the relatively small numbers of the studied populations, different observation time periods (7.5 months on average), and a high risk of bias [123]. While studying the effects of selenium on patients with Hashimoto’s disease, the authors of another meta-analysis [120] also found a significant decrease in the concentration of anti-TPO antibodies and some improvement in the quality of life of those patients already after three months of supplementation. Moreover, those authors observed a correlation between the strength of the response to supplementation and the initial concentrations of anti-TPO antibodies. Nine studies and a total of 787 patients were taken into account in another meta-analysis. All the patients received 200 μg of selenate or selenomethionine and, possibly, additional doses of L-thyroxin (in four clinical studies) and methimazole (in one clinical study). In a clear majority the patients were diagnosed with CAT, while in one study the efficacy of the therapy was evaluated in patients with diffuse toxic goitre. A six-month supplementation brought about a significant drop in the concentrations of anti-TPO antibodies. In turn, the results obtained after 12 months of therapy demonstrated a significant drop in the levels of anti-TG and also anti-TPO antibodies. In addition, based on the data from two studies, the beneficial effect of selenium supplementation was confirmed regarding the mood of the patients [126]. Brief characteristics of the most important prospective clinical studies [98, 122, 127–144] concerning the efficacy of supplementation with selenium in benign thyroid diseases, in particular autoimmune diseases, are presented in Table II.

Table II. Main characteristics of major clinical studies concerning the relationship between selenium and benign thyroid diseases

Study Studied group Study type and duration Description of intervention-group The most important, evaluated parameters The most important results
Gartner et al. JCEM (2002) [127] 70 female patients with CAT 3 months; RCS L-T4 + 200 μg/day of sodium selenate (n = 36)
L-T4 + placebo (n = 34)
The initial mean serum concentration of selenium: 0.87-0.91 μmol/L
TSH, fT3, fT4 concentration|
aTPO, aTG levels
Ultrasound thyroid image
Well-being of patients
  1. Significant aTPO decrease in the study group and no significant difference in the placebo group
  2. No significant differences among aTG concentrations in the study group and a significant drop in the control group
  3. In a higher number of patients in the study group, aTPO and aTG concentrations normalised, as well as thyroid echogenicity, and the patients’ well-being improved
Gartner et al. Biofactors (2003) [128] 47 female patients with CAT from the previous study (JCEM 2002) 6 months; NRCS The group, previously receiving selenium → continuation (Se-Se) (n = 13)
The group, previously receiving selenium → placebo (Se-0) (n = 9)
The placebo group → 200 μg/day of sodium selenate (0-Se) (n = 14)
The placebo group → placebo (0-0) (n = 11)
TSH, fT3, fT4 concentrations
aTPO, aTG levels
Ultrasound thyroid image
  1. aTPO concentrations significantly decreased in Groups 1 and 3
  2. In Group 2, aTPO concentration increased, while it did not change in any significant way in Group 4
  3. The concentrations of aTG did not undergo any significant change
  4. In three patients of Group 4 and two of Group 2, TSH increased above the normal value, despite continued L-thyroxin administration
  5. In three patients of Group 1 and two of Group 3, aTPO and aTG decreased to < 40 lU/mL and the thyroid echogenicity normalised
Duntas et al. Eur J Endocrinol (2003)[129] 65 female patients with CAT 6 months; RCS 65 patients (56 women and 9 men) with aTPO → 100 U/L and with subclinical hypothyroidism:
L-T4 + 200 μg of selenomethionine (n = 34)
L-T4 + placebo (n = 31)
The initial mean selenium concentration in serum – 75 jug/L
TSH, fT3, fT4, aTPO and aTG levels
Selenium concentration in serum
  1. The aTPO concentrations significantly decreased in both groups
  2. The differences between the groups were not significant
  3. No changes in the aTG concentrations were found
  4. TSH, fT3 and fT4 – within normal limits and without changes
Turkeret al. J Endocrinol (2006)[130] 88 female patients with CAT 9 months; RCS L-T4 + 200 μg of selenomethionine (n = 48)
Dose reduction to 100 μg after 3 months in some of the patients
Placebo
The initial mean concentration of selenium in serum – not assayed
TSH, fT3, fT4 levels
aTPO, aTG concentrations
  1. Decreased concentrations of the antibodies after selenium in dose of 200 μg/day
  2. The dose of 200 μg seems to be superior to the dose of 100 μg
  3. TSH, fT3 and fT4 – normal and without changes
Mazokopakis et ai. Thyroid (2007) [131] 80 female patients with CAT 12 months; NRCS 200 μg of selenomethionine for 6 months, then for subsequent 6 months:
Continuation of supplementation (n = 40)
Placebo (n = 40)
The initial mean concentration of selenium in serum – no data
TSH, fT3, fT4 concentrations
aTPO, aTG levels
  1. A significant drop in aTPO concentration in the first phase and further decrease in the supplementation continuing group (Group 1)
  2. A significant increase of aTPO concentration in the placebo-receiving group
  3. No relevant changes in aTG concentration
  4. TSH, fT3 and fT4 – within normal limits and without changes
Negro et al. JCEM (2007) [122] Pregnant euthyroid women: 169 with high concentrations ofaTPO and 85 with low concentrations of aTPO From the 12th week of pregnancy through the 12th month after birth RCS 200 μg of selenomethionine (n = 85)
Placebo (n = 84)
The aTPO control group (-) (n = 85)
The initial mean concentration of selenium in serum – 78.8 μg/L
TSH, fT4, aTPO levels
Selenium concentration in serum
Ultrasound thyroid image
  1. Lower incidence of the postpartum thyroiditis and of permanent hypothyroidism in the selenium-receiving group
  2. Without changes in the ultrasound image in selenium group. A significant deterioration of ultrasound image in the control group at the end of the observation period in comparison with the early pregnancy
Karanikas et al. Thyroid (2008) [132] 36 female patients with CAT 3 months; RCS L-T4 + 200 μg of sodium selenate (n = 18)
L-T4 + placebo (n = 18)
The initial mean concentration of selenium in serum – 75 jug/L
Thyroid hormones and aTPO
Well-being of patients
Lymphocytic cytokines levels
Selenium concentration in serum
  1. No significant differences among the groups, regarding the results of laboratory tests
  2. Improved well-being more often reported by patients from the selenium taking group. TSH, fT3 and fT4 – normal and without significant changes
Combs et al. Am J Clin Nutr (2009) [98] 28 healthy subjects (13 men) 28 months; NRCS All the patients received 200 μg of selenomethionine/day for 28 months
The initial mean concentration of selenium in serum – 1.64 mol/L for women and 1.78 μmol/L for men
TSH, T3,T4
Serum selenium concentration
A 5%, significant increase of T3 concentration / year, without accompanying changes of TSH or T4
Bonfig et al. Scientific World Journal (2010) [133] 49 children at the mean age of 12.2 ± 2.2 years (33 girls) with newly diagnosed CAT and hypothyroidism 12 months; RCS L-T4 (14 women and 4 men)
L-T4+ 100 μg/day of sodium selenate (9 women and 4 men)
L-T4+ 200 μg/day of sodium selenate (10 women and 8 men)
The initial mean concentration of selenium in serum – no data
TSH, fT3, fT4 concentrations
aTPO, aTG levels
  1. The aTPO concentrations were comparable in all the groups, both at study onset and after 12 months of therapy
  2. The concentrations of aTG significantly decreased in Group 1 and 3 after 12 months
Nacamulli et al. Clin Endocrinol (Oxf) (2010) [134]







76 patients (including 65 women) with CAT, in euthyreosis or with subclinical hypothyroidism 12 months; RCS Placebo (25 women and 5 men)
80 μg of sodium selenate for 6 months (40 women and 6 men)
The initial mean concentration of selenium in serum – no data
TSH, fT4 concentrations
aTPO, aTG levels
Ultrasound thyroid image
  1. No significant changes in TSH or fT4 levels, either in the groups or among them
  2. A significant drop in aTPO and aTG levels in Group 2 after 12 months
  3. A significant decrease of echogenicity in both groups after 6 months and its further drop after 12 months in the control group
Krysiak i Okopien JCEM (2011) [135] 170 euthyroid women with
CAT and 41 health subjects
6 months; RCS Female patients with newly diagnosed and untreated CAT:
L-T4 (42)
200 μg of selenomethionine (43)
L-T4 + 200 μg of selenomethionine (43)
Placebo (42)
The initial mean concentration of selenium in serum –
not assayed (57.5 μg/L at that region of Poland)
ATPO concentrations
Levels of lymphocytic and monocytic cytokines
Concentrations of acute phase parameters
The highest decrease in the concentrations of aTPO, hsCRP and secreted cytokines was noted In the group on combined therapy
Onal et al. J Pediatr Endocrinol Metab (2012) [136] Twenty-three (23) euthyroid children at the age of 12.3 ± 2.4 years with freshly diagnosed CAT (16 girls and 7 boys) 3 months; NRCS All the study participants received a daily dose of 50 μg of selenomethionine for 3 months
The initial mean concentration of selenium in serum – no access to data
TSH, fT3, fT4, aTPO, aTG concentrations
Ultrasound thyroid image
Serum selenium concentration
  1. The concentrations of aTPO and of aTG, as well as thyroid echogenicity did not change
  2. In 35% of the patients, the thyroid size decreased by ≥ 30%
Anastasllakls et al. Int J Clin Pract (2012) [137] 86 patients with CAT (including 33 men) 6 month of quasi-RCS 200 μg of selenomethionine for 3 months (n = 15)
200 μg of selenomethionine for 6 months (n = 46)
Placebo (n = 25)
The initial mean concentration of selenium in serum – 83 μg/L
TSH, fT3, fT4, aTPO and aTG levels
Serum selenium concentration
Ultrasound thyroid image
The number of lymphocytes in smear from fine-needle aspiration biopsy of the thyroid (the sub-group of 18 patients)
  1. No significant changes in aTPO concentrations and lympocyte numbers from biopsy in the selenium taking groups
  2. A significant decrease of aTG concentrations after 3 and 6 months in the selenium taking groups
  3. TSH, fT3 and fT4 – without any significant changes
Deng et al. Chinese Gen Practice (2013) [138] 94 patients with CAT (81 women and 13 men) 6 months; RCS 200 μg of selenium (n = 48; including 7 men)
Placebo (n = 46; including 6 men)
The initial mean concentration of selenium in serum – no data
TSH, fT3, fT4 levels
aTPO, aTG levels
Ultrasound thyroid image
In comparison with the control group: decreased levels of antibodies reduced goitre size and smaller focal changes
Zhang et al. Medical Innov. of China (2013) [139] 66 patients with CAT (61 women and 5 men) 3 months; RCS L-T4 + 200 μg of yeast selenium (n = 46; including 4 men)
L-T4 + placebo (n = 20; including 1 man)
The initial mean concentration of selenium in serum – no data
TSH, fT3, fT4 concentrations
aTPO, aTG levels
  1. Significant drop of aTPO concentrations in both groups, no difference between them
  2. No aTG concentration changes
Eskes et al. Clin Endocrinol (Oxf) (2014) [140] 61 women with CAT in euthyreosis and without treatment with L-T4 9 months; RCS 200 μg of sodium selenate for 6 months (n = 30)
Placebo for 6 months (n = 31)
The initial mean concentration of selenium in serum – 72.9 and 74.7 jug/L
TSH, fT4, aTPO levels
Selenium and SPP1 concentrations in serum
Quality of life
  1. Neither significant changes of TSH, ft4 and aTPO in the groups nor differences among them
  2. No differences among the groups, regarding the quality of life of the patients
Callssendorff et al. Eur Thyroid J (2015) [141] 38 patients with Graves’ disease (31 women) 9 months; RCS 30 mg of methimazole + 100 μg of L-T4 + 200 μg of selenomethionine/day (15 women and 4 men)
30 mg of methimazole + 100μg of L-T4/day (16 women and 3 men)
No selenium concentration assays, SPP concentration in serum – 47 ng/mL and
TSH, fT3, fT4 concentrations
aTSHR, aTPO levels SPP concentration in serum
Well-being studies (depression and anxiety indices)
  1. Results in Group 1:
    — significantly lower fT4 values after 18 and 36 weeks and higher TSH values after 18 weeks
    — negative correlation between depression index and fT3 and positive correlation between depression index and TSH
  2. No significant differences among the groups, regarding aTPO and aTSHR concentrations and well-being indices
Pilli i wsp. Eu r Thyroid J (2015) [142] 60 women with CAT, not treated with L-T4 and in euthyreosis 12 months; RCS Placebo (n = 20)
80 μg of selenomethionine (n = 20)
160 μg selenomethionine (n = 20)
The initial mean concentration of selenium in serum – 81.8 μg/L
TSH, fT3, fT4 concentrations
aTPO, aTG levels
Serum selenium concentration
Ultrasound thyroid image
Concentrations of selected cytokines and chemokines
Quality of life
  1. No significant changes in aTPO concentrations in selenium taking groups
  2. A significant drop of aTG concentrations in the group of patients, taking 160 μg of selenium and placebo after 12 months
  3. No significant changes in the sonographic picture of the thyroid gland and in the quality of life in and among the groups
  4. CXCL-9 and -10 chemokine concentrations decreased after 6 and 12 months of selenomethionine administration, whereas they did not change in the control group
Farias et al. J Endocrinol Invest (2015) [143] 55 patients with chronic autoimmune thyroiditis (CAT) (50 women and 5 men) 6 months; RCS 200 μg of selenomethionine for 3 months (26 women and 2 men)
Placebo for 3 months (24 women and 3 men)
The initial mean concentration of selenium in serum – no access to data
Selenium and GP1 concentrations in serum
aTPO levels
Ultrasound thyroid image
Significant decrease of aTPO concentration after 6 months of the observation, without any analogous changes in the placebo group
Wang et al. Horm Metab Res. (2016) [144] 41 patients with recurrent Graves’ disease (34 women and 7 men) 6 months + follow up; RCS or quasi-RCS Methimazole (18 women and 2 men)
Methimazole + 200 μg of sodium selenite (16 women and 5 men)
The initial mean concentration of selenium in serum – no access to data
TSH, fT3, fT4, aTSHR concentrations
The percent of remissions
Results in Group 2:
  • significantly lower fT3 and fT4 and higher TSH after 2 months of therapy
  • significantly higher aTSHR level after 6-month supplementation (assayed at follow-up visit)
  • significantly higher percent of aTSHR normalisation after 6 months (19% vs. 0%)
  • significantly higher chance to achieve remission in selenium taking patients

CAT – chronic autoimmune thyroiditis; RCS – randomised clinical study; NRCS – non-randomised clinical study – open label; GP1 – glutathione peroxidase 1; SPP – P selenoprotein

The role of selenium in thyroid gland pathophysiology

Marcocci et al. [145] studied the efficacy of supplementation with selenium in a population of 159 patients with mild Graves’ ophthalmopathy. The population was divided into three groups: the first group (control) received placebo, the second – 600 mg of pentoxifylline twice daily, and the third – 100 μg of sodium selenate (IV) twice daily. After six months of therapy with sodium selenate, the researchers observed a significant improvement in the quality of life of their patients, less intensity in the ophthalmological changes, and suppression of orbitopathy progression into its more severe forms. The intensity of ophthalmological changes, evaluated by the Clinical Activity Score (CAS), was reduced in all the groups, but it was significantly lower in the group receiving selenium. The results, obtained after 12 months, already after selenium withdrawal, were analogous to those obtained before [145]. The results, as presented above, have contributed to the inclusion of selenium supplementation in the recommendations of European Group on Graves’ orbitopathy (EUGOGO), issued in 2016, according to which, in benign forms of short-term thyroid orbitopathy, the use of sodium selenate is recommended twice daily in dose of 100 μg for six months [146], although its use was already encouraged before that [147]. Two interesting clinical trials are currently underway [148, 149], the results of which may bring us closer to a more conclusive answer to the question of whether supplementation with selenium significantly influences the state of patients with Graves’ disease [148] and the quality of life of patients with CAT [149].

Selenium plays a significant role in the maintenance of homeostasis of the human body. Selenoproteins protect the body against damage exerted by oxidative stress, including lesions in the TG, and are responsible for its physiological activity and adequate hormonal production. The issue of selenium supplementation for primary and secondary prophylactic purposes, regarding various structural and functional disorders of the TG, remains an open question and requires further studies. Taking into account the data from the presented clinical studies, one should consider the application of selenium supplementation in autoimmune diseases of the TG, including the widespread Hashimoto disease. Patients who are in the population with selenium deficit may especially benefit, as well as persons in whom selenium deficit is confirmed by selenium concentration assay in plasma. One should then consider the cost-effectiveness of such an approach and the possible implementation of such studies into diagnostic algorithms. However, the rather narrow therapeutic index of selenium should be taken into account, as well as the risk associated with acute and chronic overdose, in particular, the increased risk for development of diabetes mellitus type 2. In consideration of the increased demands for selenium in pregnancy, the benefits should also be considered, which could be achieved as a result of selenium supplementation in pregnant women and, possibly, appropriate recommendations should be defined, similar as those formulated for iodine [150]. At present, a six-month supplementation with 200 μg of selenium is recommended in mild forms of shortterm thyroid ophthalmopathy, taking into account its measurable clinical benefits.

References

  1. Rayman MP, Rayman MP. The importance of selenium to human health. Lancet. 2000; 356(9225): 233–241, doi: 10.1016/S0140-6736(00)02490-9, indexed in Pubmed: 10963212.
  2. Hoffmann PR, Berry MJ. The influence of selenium on immune responses. Mol Nutr Food Res. 2008; 52(11): 1273–1280, doi: 10.1002/ mnfr.200700330, indexed in Pubmed: 18384097.
  3. Holben DH, Smith AM. The diverse role of selenium within selenoproteins: a review. J Am Diet Assoc. 1999; 99(7): 836–843, doi: 10.1016/ S0002-8223(99)00198-4, indexed in Pubmed: 10405682.
  4. Kieliszek M, Błażejak S. Selenium: Significance, and outlook for supplementation. Nutrition. 2013; 29(5): 713–718, doi: 10.1016/j.nut.2012.11.012, indexed in Pubmed: 23422539.
  5. Slencu BG, Ciobanu C, Cuciureanu R. Selenium content in foodstuffs and its nutritional requirement forhumans. Clujul Med. ; 2012: 139–145.
  6. Dietary re, vitamin E. selenium, and carotenoids: A report of the Panel on Dietary Antioxidants and Related Compounds, Subcommittees on Upper Reference Levels of Nutrients and Interpretation and Uses of Dietary Reference Intakes, and the Standing Committee on the Scientific Evaluation of Dietary Reference Intakes, Food and Nutrition Board, Institute of Medicine. Washington, D.C. : National Academy Press. ; 2000.
  7. Navarro-Alarcon M, Cabrera-Vique C. Selenium in food and the human body: a review. Sci Total Environ. 2008; 400(1-3): 115–141, doi: 10.1016/j. scitotenv.2008.06.024, indexed in Pubmed: 18657851.
  8. Alfthan G, Eurola M, Ekholm P, et al. Selenium Working Group. Effects of nationwide addition of selenium to fertilizers on foods, and animal and human health in Finland: From deficiency to optimal selenium status of the population. J Trace Elem Med Biol. 2015; 31: 142–147, doi: 10.1016/j.jtemb.2014.04.009, indexed in Pubmed: 24908353.
  9. Fairweather-Tait SJ, Collings R, Hurst R. Selenium bioavailability: current knowledge and future research requirements. Am J Clin Nutr. 2010; 91(5): 1484S–1491S, doi: 10.3945/ajcn.2010.28674J, indexed in Pubmed: 20200264.
  10. Wasowicz W, Gromadzinska J, Rydzynski K, et al. Selenium status of low-selenium area residents: Polish experience. Toxicol Lett. 2003; 137(1-2): 95–101, indexed in Pubmed: 12505435.
  11. Jablonska E, Gromadzinska J, Klos A, et al. Original research article. Journal of Food Composition and Analysis. 2013; 31: 259–65.
  12. Schrauzer GN, White DA. Selenium in human nutrition: dietary intakes and effects of supplementation. Bioinorg Chem. 1978; 8(4): 303–318, indexed in Pubmed: 647060.
  13. Combs GF. Selenium in global food systems. Br J Nutr. 2001; 85(5): 517–547, indexed in Pubmed: 11348568.
  14. Patterson BH, Levander OA. Naturally occurring selenium compounds in cancer chemoprevention trials: a workshop summary. Cancer Epidemiol Biomarkers Prev. 1997; 6(1): 63–69, indexed in Pubmed: 8993799.
  15. Zagrodzki P, Laszczyk P. [Selenium and cardiovascular disease: selected issues]. Postepy Hig Med Dosw (Online). 2006; 60: 624–631, indexed in Pubmed: 17199104.
  16. Fordyce F. Selenium Geochemistry and Health. AMBIO: A Journal of the Human Environment. 2007; 36(1): 94–97, doi: 10.1579/0044-7447(2007)36[94:sgah]2.0.co;2.
  17. Dumont E, Vanhaecke F, Cornelis R. Selenium speciation from food source to metabolites: a critical review. Anal Bioanal Chem. 2006; 385(7): 1304–1323, doi: 10.1007/s00216-006-0529-8, indexed in Pubmed: 16830114.
  18. Davis TZ, Tiwary AK, Stegelmeier BL, et al. Comparative oral dose toxicokinetics of sodium selenite and selenomethionine. J Appl Toxicol. 2017; 37(2): 231–238, doi: 10.1002/jat.3350, indexed in Pubmed: 27283737.
  19. Pérez-Corona MT, Sánchez-Martínez M, Valderrama MJ, et al. Selenium biotransformation by Saccharomyces cerevisiae and Saccharomyces bayanus during white wine manufacture: Laboratory-scale experiments. Food Chemistry. 2011; 124(3): 1050–1055, doi: 10.1016/j.foodchem.2010.07.073.
  20. Burk RF, Norsworthy BK, Hill KE, et al. Effects of chemical form of selenium on plasma biomarkers in a high-dose human supplementation trial. Cancer Epidemiol Biomarkers Prev. 2006; 15(4): 804–810, doi: 10.1158/1055-9965.EPI-05-0950, indexed in Pubmed: 16614127.
  21. Combs GF, Watts JC, Jackson MI, et al. Determinants of selenium status in healthy adults. Nutr J. 2011; 10: 75, doi: 10.1186/1475-2891-10-75, indexed in Pubmed: 21767397.
  22. Hollenbach B, Morgenthaler NG, Struck J, et al. New assay for the measurement of selenoprotein P as a sepsis biomarker from serum. J Trace Elem Med Biol. 2008; 22(1): 24–32, doi: 10.1016/j.jtemb.2007.11.003, indexed in Pubmed: 18319137.
  23. Burk RF, Hill KE. Selenoprotein P: an extracellular protein with unique physical characteristics and a role in selenium homeostasis. Annu Rev Nutr. 2005; 25: 215–235, doi: 10.1146/annurev.nutr.24.012003.132120, indexed in Pubmed: 16011466.
  24. Hoeflich J, Hollenbach B, Behrends T, et al. The choice of biomarkers determines the selenium status in young German vegans and vegetarians. Br J Nutr. 2010; 104(11): 1601–1604, doi: 10.1017/S0007114510002618, indexed in Pubmed: 20637135.
  25. Hurst R, Armah CN, Dainty JR, et al. Establishing optimal selenium status: results of a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2010; 91(4): 923–931, doi: 10.3945/ajcn.2009.28169, indexed in Pubmed: 20181815.
  26. Mistry HD, Broughton Pipkin F, Redman CWG, et al. Selenium in reproductive health. Am J Obstet Gynecol. 2012; 206(1): 21–30, doi: 10.1016/j. ajog.2011.07.034, indexed in Pubmed: 21963101.
  27. Li S, Xiao T, Zheng B. Medical geology of arsenic, selenium and thallium in China. Sci Total Environ. 2012; 421-422: 31–40, doi: 10.1016/j. scitotenv.2011.02.040, indexed in Pubmed: 21440288.
  28. Pedrero Z, Madrid Y. Novel approaches for selenium speciation in foodstuffs and biological specimens: a review. Anal Chim Acta. 2009; 634(2): 135–152, doi: 10.1016/j.aca.2008.12.026, indexed in Pubmed: 19185112.
  29. Suetens C, Moreno-Reyes R, Chasseur C, et al. Epidemiological support for a multifactorial aetiology of Kashin-Beck disease in Tibet. Int Orthop. 2001; 25(3): 180–187, indexed in Pubmed: 11482537.
  30. Chen J. An original discovery: selenium deficiency and Keshan disease (an endemic heart disease). Asia Pac J Clin Nutr. 2012; 21(3): 320–326, indexed in Pubmed: 22705420.
  31. Yao Y, Pei F, Kang P. Selenium, iodine, and the relation with Kashin-Beck disease. Nutrition. 2011; 27(11-12): 1095–1100, doi: 10.1016/j. nut.2011.03.002, indexed in Pubmed: 21967994.
  32. Hendrickx W, Decock J, Mulholland F, et al. Selenium Biomarkers in Prostate Cancer Cell Lines and Influence of Selenium on Invasive Potential of PC3 Cells. Front Oncol. 2013; 3: 239, doi: 10.3389/fonc.2013.00239, indexed in Pubmed: 24066278.
  33. Ruseva B, Himcheva I, Nankova D. Importance of selenoproteins for the function of the thyroid gland. Medicine. 2013; 3: 60–64.
  34. Roman M, Jitaru P, Barbante C. Selenium biochemistry and its role for human health. Metallomics. 2014; 6(1): 25–54, doi: 10.1039/c3mt00185g, indexed in Pubmed: 24185753.
  35. Letavayová L, Vlasáková D, Spallholz J, et al. Toxicity and mutagenicity of selenium compounds in Saccharomyces cerevisiae. Mutation Research/ Fundamental and Molecular Mechanisms of Mutagenesis. 2008; 638(1-2): 1–10, doi: 10.1016/j.mrfmmm.2007.08.009.
  36. Fordyce F. Selenium Deficiency and Toxicity in the Environment. Essentials of Medical Geology. 2012: 375–416, doi: 10.1007/978-94-007-4375-5_16.
  37. Nazemi L, Nazmara S, Eshraghyan MR, et al. Selenium status in soil, water and essential crops of Iran. Iranian J Environ Health Sci Eng. 2012; 9(1): 11, doi: 10.1186/1735-2746-9-11, indexed in Pubmed: 23369199.
  38. Drutel A, Archambeaud F, Caron P. Selenium and the thyroid gland: more good news for clinicians. Clin Endocrinol (Oxf). 2013; 78(2): 155–164, doi: 10.1111/cen.12066, indexed in Pubmed: 23046013.
  39. Rayman MP. The use of high-selenium yeast to raise selenium status: how does it measure up? Br J Nutr. 2004; 92(4): 557–573, indexed in Pubmed: 15522125.
  40. Zwolak I, Zaporowska H. Selenium interactions and toxicity: a review. Selenium interactions and toxicity. Cell Biol Toxicol. 2012; 28(1): 31–46, doi: 10.1007/s10565-011-9203-9, indexed in Pubmed: 21913064.
  41. Dharmasena A. Selenium supplementation in thyroid associated ophthalmopathy: an update. Int J Ophthalmol. 2014; 7(2): 365–375, doi: 10.3980/j.issn.2222-3959.2014.02.31, indexed in Pubmed: 24790886.
  42. Muller FL, Lustgarten MS, Jang Y, et al. Trends in oxidative aging theories. Free Radic Biol Med. 2007; 43(4): 477–503, doi: 10.1016/j.freeradbiomed. 2007.03.034, indexed in Pubmed: 17640558.
  43. Berry MJ, Banu L, Chen YY, et al. Recognition of UGA as a selenocysteine codon in type I deiodinase requires sequences in the 3’ untranslated region. Nature. 1991; 353(6341): 273–276, doi: 10.1038/353273a0, indexed in Pubmed: 1832744.
  44. Köhrle J, Jakob F, Contempré B, et al. Selenium, the thyroid, and the endocrine system. Endocr Rev. 2005; 26(7): 944–984, doi: 10.1210/er.2001-0034, indexed in Pubmed: 16174820.
  45. Arthur JR, Nicol F, Beckett GJ. Hepatic iodothyronine 5_-deiodinase. The role of selenium. Biochemical Journal. 1990; 272(2): 537–540, doi: 10.1042/bj2720537.
  46. Behne D, Kyriakopoulos A, Meinhold H, et al. Identification of type I iodothyronine 5’-deiodinase as a selenoenzyme. Biochem Biophys Res Commun. 1990; 173(3): 1143–1149, indexed in Pubmed: 2268318.
  47. Rosen BP, Liu Z. Transport pathways for arsenic and selenium: a minireview. Environ Int. 2009; 35(3): 512–515, doi: 10.1016/j.envint.2008.07.023, indexed in Pubmed: 18789529.
  48. Tamura T, Stadtman TC. A new selenoprotein from human lung adenocarcinoma cells: purification, properties, and thioredoxin reductase activity. Proc Natl Acad Sci U S A. 1996; 93(3): 1006–1011, indexed in Pubmed: 8577704.
  49. Pappa EC, Pappas AC, Surai PF. Selenium content in selected foods from the Greek market and estimation of the daily intake. Sci Total Environ. 2006; 372(1): 100–108, doi: 10.1016/j.scitotenv.2006.08.008, indexed in Pubmed: 16959300.
  50. Rayman M. Selenium and human health. The Lancet. 2012; 379(9822): 1256–1268, doi: 10.1016/s0140-6736(11)61452-9.
  51. Cagliani R, Fruguglietti ME, Berardinelli A, et al. New molecular findings in congenital myopathies due to selenoprotein N gene mutations. J Neurol Sci. 2011; 300(1-2): 107–113, doi: 10.1016/j.jns.2010.09.011, indexed in Pubmed: 20937510.
  52. Brozmanová J, Mániková D, Vlčková V, et al. Selenium: a double-edged sword for defense and offence in cancer. Arch Toxicol. 2010; 84(12): 919–938, doi: 10.1007/s00204-010-0595-8, indexed in Pubmed: 20871980.
  53. Papp LV, Lu J, Holmgren A, et al. From selenium to selenoproteins: synthesis, identity, and their role in human health. Antioxid Redox Signal. 2007; 9(7): 775–806, doi: 10.1089/ars.2007.1528, indexed in Pubmed: 17508906.
  54. Kamwesiga J, Mutabazi V, Kayumba J, et al. Effect of selenium supplementation on CD4+ T-cell recovery, viral suppression and morbidity of HIV-infected patients in Rwanda. AIDS. 2015; 29(9): 1045–1052, doi: 10.1097/qad.0000000000000673.
  55. Lipinski B. Can Selenite be an Ultimate Inhibitor of Ebola and Other Viral Infections? British Journal of Medicine and Medical Research. 2015; 6(3): 319–324, doi: 10.9734/bjmmr/2015/14858.
  56. Kawai K, Meydani SN, Urassa W, et al. Micronutrient supplementation and T cell-mediated immune responses in patients with tuberculosis in Tanzania. Epidemiol Infect. 2014; 142(7): 1505–1509, doi: 10.1017/ S0950268813002495, indexed in Pubmed: 24093552.
  57. Solovyev ND. Importance of selenium and selenoprotein for brain function: From antioxidant protection to neuronal signalling. J Inorg Biochem. 2015; 153: 1–12, doi: 10.1016/j.jinorgbio.2015.09.003, indexed in Pubmed: 26398431.
  58. IMS Institute for Healthcare Informatics. Medicine use and shifting costs of healthcare: a review of the use of medicines in the United States in 2013. www.imshealth.com.
  59. Biron VL, Bang H, Farwell DG, et al. National Trends and Factors Associated with Hospital Costs Following Thyroid Surgery. Thyroid. 2015; 25(7): 823–829, doi: 10.1089/thy.2014.0495, indexed in Pubmed: 25873398.
  60. Nexo MA, Watt T, Pedersen J, et al. Increased risk of long-term sickness absence, lower rate of return to work, and higher risk of unemployment and disability pensioning for thyroid patients: a Danish register-based cohort study. J Clin Endocrinol Metab. 2014; 99(9): 3184–3192, doi: 10.1210/jc.2013-4468, indexed in Pubmed: 24937367.
  61. Cooper R, Pinkney J, Ayling RM. Appropriateness of prescribing thyroxine in primary care. Ann Clin Biochem. 2015; 52(Pt 4): 497–501, doi: 10.1177/0004563214568686, indexed in Pubmed: 25575699.
  62. Hess SY. The impact of common micronutrient deficiencies on iodine and thyroid metabolism: the evidence from human studies. Best Pract Res Clin Endocrinol Metab. 2010; 24(1): 117–132, doi: 10.1016/j. beem.2009.08.012, indexed in Pubmed: 20172476.
  63. Yu X, Shan Z, Li C, et al. Iron deficiency, an independent risk factor for isolated hypothyroxinemia in pregnant and nonpregnant women of childbearing age in China. J Clin Endocrinol Metab. 2015; 100(4): 1594–1601, doi: 10.1210/jc.2014-3887, indexed in Pubmed: 25599388.
  64. Knudsen N, Brix TH. Genetic and non-iodine-related factors in the aetiology of nodular goitre. Best Pract Res Clin Endocrinol Metab. 2014; 28(4): 495–506, doi: 10.1016/j.beem.2014.02.005, indexed in Pubmed: 25047201.
  65. Jain RB. Thyroid function and serum copper, selenium, and zinc in general U.S. population. Biol Trace Elem Res. 2014; 159(1-3): 87–98, doi: 10.1007/s12011-014-9992-9, indexed in Pubmed: 24789479.
  66. Zimmermann MB. The influence of iron status on iodine utilization and thyroid function. Annu Rev Nutr. 2006; 26: 367–389, doi: 10.1146/annurev. nutr.26.061505.111236, indexed in Pubmed: 16602928.
  67. Jain RB, Choi YS. Interacting effects of selected trace and toxic metals on thyroid function. Int J Environ Health Res. 2016; 26(1): 75–91, doi: 10.1080/09603123.2015.1020416, indexed in Pubmed: 25788177.
  68. Contempre B, Dumont JE, Denef JF, et al. Effects of selenium deficiency on thyroid necrosis, fibrosis and proliferation: a possible role in myxoedematous cretinism. Eur J Endocrinol. 1995; 133(1): 99–109, indexed in Pubmed: 7627345.
  69. Schomburg L, Köhrle J. On the importance of selenium and iodine metabolism for thyroid hormone biosynthesis and human health. Mol Nutr Food Res. 2008; 52(11): 1235–1246, doi: 10.1002/mnfr.200700465, indexed in Pubmed: 18686295.
  70. Liu Y, Huang H, Zeng J, et al. Thyroid volume, goiter prevalence, and selenium levels in an iodine-sufficient area: a cross-sectional study. BMC Public Health. 2013; 13: 1153, doi: 10.1186/1471-2458-13-1153, indexed in Pubmed: 24321191.
  71. Contempre B, Le Moine O, Dumont JE, et al. Selenium deficiency and thyroid fibrosis. A key role for macrophages and transforming growth factor beta (TGF-beta). Mol Cell Endocrinol. 1996; 124(1-2): 7–15, indexed in Pubmed: 9027319.
  72. Xu J, Liu XL, Yang XF, et al. Supplemental selenium alleviates the toxic effects of excessive iodine on thyroid. Biol Trace Elem Res. 2011; 141(1-3): 110–118, doi: 10.1007/s12011-010-8728-8, indexed in Pubmed: 20517655.
  73. Azevedo MF, Barra GB, Naves LA, et al. Selenoprotein-related disease in a young girl caused by nonsense mutations in the SBP2 gene. J Clin Endocrinol Metab. 2010; 95(8): 4066–4071, doi: 10.1210/jc.2009-2611, indexed in Pubmed: 20501692.
  74. Dumitrescu AM, Liao XH, Abdullah MSY, et al. Mutations in SECISBP2 result in abnormal thyroid hormone metabolism. Nat Genet. 2005; 37(11): 1247–1252, doi: 10.1038/ng1654, indexed in Pubmed: 16228000.
  75. Hamajima T, Mushimoto Y, Kobayashi H, et al. Novel compound heterozygous mutations in the SBP2 gene: characteristic clinical manifestations and the implications of GH and triiodothyronine in longitudinal bone growth and maturation. Eur J Endocrinol. 2012; 166(4): 757–764, doi: 10.1530/EJE-11-0812, indexed in Pubmed: 22247018.
  76. Schoenmakers E, Agostini M, Mitchell C, et al. Mutations in the selenocysteine insertion sequence-binding protein 2 gene lead to a multisystem selenoprotein deficiency disorder in humans. J Clin Invest. 2010; 120(12): 4220–4235, doi: 10.1172/JCI43653, indexed in Pubmed: 21084748.
  77. Hatfield DL, Tsuji PA, Carlson BA, et al. Selenium and selenocysteine: roles in cancer, health, and development. Trends Biochem Sci. 2014; 39(3): 112–120, doi: 10.1016/j.tibs.2013.12.007, indexed in Pubmed: 24485058.
  78. Labunskyy VM, Hatfield DL, Gladyshev VN. Selenoproteins: Molecular Pathways and Physiological Roles. Physiol Rev. 2014; 94(3): 739–777, doi: 10.1152/physrev.00039.2013.
  79. Köhrle J. Pathophysiological relevance of selenium. J Endocrinol Invest. 2013; 36(10 Suppl): 1–7, indexed in Pubmed: 24419053.
  80. Contempré B, Duale NL, Dumont JE, et al. Effect of selenium supplementation on thyroid hormone metabolism in an iodine and selenium deficient population. Clin Endocrinol (Oxf). 1992; 36(6): 579–583, indexed in Pubmed: 1424183.
  81. Contempré B, de Escobar GM, Denef JF, et al. Thiocyanate induces cell necrosis and fibrosis in selenium- and iodine-deficient rat thyroids: a potential experimental model for myxedematous endemic cretinism in central Africa. Endocrinology. 2004; 145(2): 994–1002, doi: 10.1210/ en.2003-0886, indexed in Pubmed: 14500569.
  82. Turanov AA, Shchedrina VA, Everley RA, et al. Selenoprotein S is involved in maintenance and transport of multiprotein complexes. Biochem J. 2014; 462(3): 555–565, doi: 10.1042/BJ20140076, indexed in Pubmed: 24897171.
  83. Schomburg L. Selenium, selenoproteins and the thyroid gland: interactions in health and disease. Nat Rev Endocrinol. 2011; 8(3): 160–171, doi: 10.1038/nrendo.2011.174, indexed in Pubmed: 22009156.
  84. Shchedrina VA, Zhang Y, Labunskyy VM, et al. Structure-function relations, physiological roles, and evolution of mammalian ER-resident selenoproteins. Antioxid Redox Signal. 2010; 12(7): 839–849, doi: 10.1089/ ars.2009.2865, indexed in Pubmed: 19747065.
  85. Rijntjes E, Scholz PM, Mugesh G, et al. Se- and s-based thiouracil and methimazole analogues exert different inhibitory mechanisms on type 1 and type 2 deiodinases. Eur Thyroid J. 2013; 2(4): 252–258, doi: 10.1159/000355288, indexed in Pubmed: 24783056.
  86. Bhowmick D, Srivastava S, D’Silva P, et al. Highly Efficient Glutathione Peroxidase and Peroxiredoxin Mimetics Protect Mammalian Cells against Oxidative Damage. Angew Chem Int Ed Engl. 2015; 54(29): 8449–8453, doi: 10.1002/anie.201502430, indexed in Pubmed: 26032473.
  87. Manna D, Roy G, Mugesh G. Antithyroid drugs and their analogues: synthesis, structure, and mechanism of action. Acc Chem Res. 2013; 46(11): 2706–2715, doi: 10.1021/ar4001229, indexed in Pubmed: 23883148.
  88. Raja K, Mugesh G. Remarkable effect of chalcogen substitution on an enzyme mimetic for deiodination of thyroid hormones. Angew Chem Int Ed Engl. 2015; 54(26): 7674–7678, doi: 10.1002/anie.201502762, indexed in Pubmed: 25968052.
  89. Weissel M. Propylthiouracil: clinical overview of its efficacy and its side effects more than 50 years after the introduction of its use in thyrostatic treatment. Exp Clin Endocrinol Diabetes. 2010; 118(2): 101–104, doi: 10.1055/s-0029-1215587, indexed in Pubmed: 19449281.
  90. Gershengorn MC, Neumann S. Update in TSH receptor agonists and antagonists. J Clin Endocrinol Metab. 2012; 97(12): 4287–4292, doi: 10.1210/jc.2012-3080, indexed in Pubmed: 23019348.
  91. Derumeaux H, Valeix P, Castetbon K, et al. Association of selenium with thyroid volume and echostructure in 35- to 60-year-old French adults. Eur J Endocrinol. 2003; 148(3): 309–315, indexed in Pubmed: 12611611.
  92. Rasmussen LB, Schomburg L, Köhrle J, et al. Selenium status, thyroid volume, and multiple nodule formation in an area with mild iodine deficiency. Eur J Endocrinol. 2011; 164(4): 585–590, doi: 10.1530/EJE-10-1026, indexed in Pubmed: 21242171.
  93. Bülow Pedersen I, Knudsen N, Carlé A, et al. Serum selenium is low in newly diagnosed Graves’ disease: a population-based study. Clin Endocrinol (Oxf). 2013; 79(4): 584–590, doi: 10.1111/cen.12185, indexed in Pubmed: 23448365.
  94. Li H, Li J. Thyroid disorders in women. Minerva Med. 2015; 106(2): 109–114, indexed in Pubmed: 25668600.
  95. Schomburg L, Schweizer U. Hierarchical regulation of selenoprotein expression and sex-specific effects of selenium. Biochim Biophys Acta. 2009; 1790(11): 1453–1462, doi: 10.1016/j.bbagen.2009.03.015, indexed in Pubmed: 19328222.
  96. Riese C, Michaelis M, Mentrup B, et al. Selenium-dependent pre- and posttranscriptional mechanisms are responsible for sexual dimorphic expression of selenoproteins in murine tissues. Endocrinology. 2006; 147(12): 5883–5892, doi: 10.1210/en.2006-0689, indexed in Pubmed: 16959840.
  97. Stoedter M, Renko K, Hög A, et al. Selenium controls the sex-specific immune response and selenoprotein expression during the acute-phase response in mice. Biochem J. 2010; 429(1): 43–51, doi: 10.1042/BJ20091868, indexed in Pubmed: 20370716.
  98. Combs GF, Midthune DN, Patterson KY, et al. Effects of selenomethionine supplementation on selenium status and thyroid hormone concentrations in healthy adults. Am J Clin Nutr. 2009; 89(6): 1808–1814, doi: 10.3945/ajcn.2008.27356, indexed in Pubmed: 19403637.
  99. Pellegriti G, Frasca F, Regalbuto C, et al. Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol. 2013; 2013: 965212, doi: 10.1155/2013/965212, indexed in Pubmed: 23737785.
  100. Colonna M, Uhry Z, Guizard AV, et al. FRANCIM network. Recent trends in incidence, geographical distribution, and survival of papillary thyroid cancer in France. Cancer Epidemiol. 2015; 39(4): 511–518, doi: 10.1016/j. canep.2015.04.015, indexed in Pubmed: 26003877.
  101. Davies L, Welch HG. Current thyroid cancer trends in the United States. JAMA Otolaryngol Head Neck Surg. 2014; 140(4): 317–322, doi: 10.1001/ jamaoto.2014.1, indexed in Pubmed: 24557566.
  102. Davis CD, Tsuji PA, Milner JA. Selenoproteins and cancer prevention. Annu Rev Nutr. 2012; 32: 73–95, doi: 10.1146/annurev-nutr-071811-150740, indexed in Pubmed: 22404120.
  103. Glattre E, Thomassen Y, Thoresen SO, et al. Prediagnostic serum selenium in a case-control study of thyroid cancer. Int J Epidemiol. 1989; 18(1): 45–49, indexed in Pubmed: 2722382.
  104. Kucharzewski M, Braziewicz J, Majewska U, et al. Concentration of selenium in the whole blood and the thyroid tissue of patients with various thyroid diseases. Biol Trace Elem Res. 2002; 88(1): 25–30, doi: 10.1385/BTER:88:1:25, indexed in Pubmed: 12117262.
  105. Moncayo R, Kroiss A, Oberwinkler M, et al. The role of selenium, vitamin C, and zinc in benign thyroid diseases and of selenium in malignant thyroid diseases: Low selenium levels are found in subacute and silent thyroiditis and in papillary and follicular carcinoma. BMC Endocr Disord. 2008; 8: 2, doi: 10.1186/1472-6823-8-2, indexed in Pubmed: 18221503.
  106. Przybylik-Mazurek E, Zagrodzki P, Kuźniarz-Rymarz S, et al. Thyroid disorders-assessments of trace elements, clinical, and laboratory parameters. Biol Trace Elem Res. 2011; 141(1-3): 65–75, doi: 10.1007/s12011-010-8719-9, indexed in Pubmed: 20455027.
  107. Jonklaas J, Danielsen M, Wang H. A pilot study of serum selenium, vitamin D, and thyrotropin concentrations in patients with thyroid cancer. Thyroid. 2013; 23(9): 1079–1086, doi: 10.1089/thy.2012.0548, indexed in Pubmed: 23350941.
  108. O’Grady TJ, Kitahara CM, DiRienzo AG, et al. The association between selenium and other micronutrients and thyroid cancer incidence in the NIH-AARP Diet and Health Study. PLoS One. 2014; 9(10): e110886, doi: 10.1371/journal.pone.0110886, indexed in Pubmed: 25329812.
  109. Shen F, Cai WS, Li JL, et al. The Association Between Serum Levels of Selenium, Copper, and Magnesium with Thyroid Cancer: a Meta-analysis. Biol Trace Elem Res. 2015; 167(2): 225–235, doi: 10.1007/s12011-015-0304-9, indexed in Pubmed: 25820485.
  110. Baltaci AK, Dundar TK, Aksoy F, et al. Changes in the Serum Levels of Trace Elements Before and After the Operation in Thyroid Cancer Patients. Biol Trace Elem Res. 2017; 175(1): 57–64, doi: 10.1007/s12011-016-0768-2, indexed in Pubmed: 27263537.
  111. Chung HK, Nam JiS, Ahn CW, et al. Some Elements in Thyroid Tissue are Associated with More Advanced Stage of Thyroid Cancer in Korean Women. Biol Trace Elem Res. 2016; 171(1): 54–62, doi: 10.1007/s12011-015-0502-5, indexed in Pubmed: 26419761.
  112. Becker NP, Martitz J, Renko K, et al. Hypoxia reduces and redirects selenoprotein biosynthesis. Metallomics. 2014; 6(5): 1079–1086, doi: 10.1039/c4mt00004h, indexed in Pubmed: 24700164.
  113. Hesse-Bähr K, Dreher I, Köhrle J. The influence of the cytokines Il-1beta and INFgamma on the expression of selenoproteins in the human hepatocarcinoma cell line HepG2. Biofactors. 2000; 11(1-2): 83–85, indexed in Pubmed: 10705969.
  114. Renko K, Hofmann PJ, Stoedter M, et al. Down-regulation of the hepatic selenoprotein biosynthesis machinery impairs selenium metabolism during the acute phase response in mice. FASEB J. 2009; 23(6): 1758–1765, doi: 10.1096/fj.08-119370, indexed in Pubmed: 19136613.
  115. Dreher I, Jakobs TC, Köhrle J. Cloning and characterization of the human selenoprotein P promoter. Response of selenoprotein P expression to cytokines in liver cells. J Biol Chem. 1997; 272(46): 29364–29371, indexed in Pubmed: 9361018.
  116. Xue H, Wang W, Li Y, et al. Selenium upregulates CD4(+)CD25(+) regulatory T cells in iodine-induced autoimmune thyroiditis model of NOD.H-2(h4) mice. Endocr J. 2010; 57(7): 595–601, indexed in Pubmed: 20453397.
  117. Huang Z, Rose AH, Hoffmann PR. The role of selenium in inflammation and immunity: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 2012; 16(7): 705–743, doi: 10.1089/ ars.2011.4145, indexed in Pubmed: 21955027.
  118. Tan L, Sang ZNa, Shen J, et al. Selenium supplementation alleviates autoimmune thyroiditis by regulating expression of TH1/TH2 cytokines. Biomed Environ Sci. 2013; 26(11): 920–925, doi: 10.3967/bes2013.022, indexed in Pubmed: 24331538.
  119. Hawkes WC, Richter D, Alkan Z. Dietary selenium supplementation and whole blood gene expression in healthy North American men. Biol Trace Elem Res. 2013; 155(2): 201–208, doi: 10.1007/s12011-013-9786-5, indexed in Pubmed: 23955482.
  120. Toulis KA, Anastasilakis AD, Tzellos TG, et al. Selenium supplementation in the treatment of Hashimoto’s thyroiditis: a systematic review and a meta-analysis. Thyroid. 2010; 20(10): 1163–1173, doi: 10.1089/ thy.2009.0351, indexed in Pubmed: 20883174.
  121. Mao J, Pop VJ, Bath SC, et al. Effect of low-dose selenium on thyroid autoimmunity and thyroid function in UK pregnant women with mild to moderate iodine deficiency. Eur J Nutr. 2016; 55(1): 55–61, doi: 10.1007/ s00394-014-0822-9, indexed in Pubmed: 25524327.
  122. Negro R, Greco G, Mangieri T, et al. The influence of selenium supplementation on postpartum thyroid status in pregnant women with thyroid peroxidase autoantibodies. J Clin Endocrinol Metab. 2007; 92(4): 1263–1268, doi: 10.1210/jc.2006-1821, indexed in Pubmed: 17284630.
  123. van Zuuren EJ, Albusta AY, Fedorowicz Z, et al. Selenium supplementation for Hashimoto’s thyroiditis. Cochrane Database Syst Rev. 2013(6): CD010223, doi: 10.1002/14651858.CD010223.pub2, indexed in Pubmed: 23744563.
  124. Köhrle J, Gärtner R. Selenium and thyroid. Best Pract Res Clin Endocrinol Metab. 2009; 23(6): 815–827, doi: 10.1016/j.beem.2009.08.002, indexed in Pubmed: 19942156.
  125. Rayman MP, Thompson AJ, Bekaert B, et al. Randomized controlled trial of the effect of selenium supplementation on thyroid function in the elderly in the United Kingdom. Am J Clin Nutr. 2008; 87(2): 370–378, indexed in Pubmed: 18258627.
  126. Fan Y, Xu S, Zhang H, et al. Selenium supplementation for autoimmune thyroiditis: a systematic review and meta-analysis. Int J Endocrinol. 2014; 2014: 904573, doi: 10.1155/2014/904573, indexed in Pubmed: 25574167.
  127. Gärtner R, Gasnier BCH, Dietrich JW, et al. Selenium supplementation in patients with autoimmune thyroiditis decreases thyroid peroxidase antibodies concentrations. J Clin Endocrinol Metab. 2002; 87(4): 1687–1691, doi: 10.1210/jcem.87.4.8421, indexed in Pubmed: 11932302.
  128. Gärtner R, Gasnier B. Selenium in the treatment of autoimmune thyroiditis. BioFactors. 2003; 19(3-4): 165–170, doi: 10.1002/biof.5520190309.
  129. Duntas LH, Mantzou E, Koutras DA. Effects of a six month treatment with selenomethionine in patients with autoimmune thyroiditis. Eur J Endocrinol. 2003; 148(4): 389–393, indexed in Pubmed: 12656658.
  130. Turker O, Kumanlioglu K, Karapolat I, et al. Selenium treatment in autoimmune thyroiditis: 9-month follow-up with variable doses. J Endocrinol. 2006; 190(1): 151–156, doi: 10.1677/joe.1.06661, indexed in Pubmed: 16837619.
  131. Mazokopakis EE, Papadakis JA, Papadomanolaki MG, et al. Effects of 12 months treatment with L-selenomethionine on serum anti-TPO Levels in Patients with Hashimoto’s thyroiditis. Thyroid. 2007; 17(7): 609–612, doi: 10.1089/thy.2007.0040, indexed in Pubmed: 17696828.
  132. Karanikas G, Schuetz M, Kontur S, et al. No immunological benefit of selenium in consecutive patients with autoimmune thyroiditis. Thyroid. 2008; 18(1): 7–12, doi: 10.1089/thy.2007.0127, indexed in Pubmed: 18302514.
  133. Bonfig W, Gärtner R, Schmidt H. Selenium supplementation does not decrease thyroid peroxidase antibody concentration in children and adolescents with autoimmune thyroiditis. ScientificWorldJournal. 2010; 10: 990–996, doi: 10.1100/tsw.2010.91, indexed in Pubmed: 20526530.
  134. Nacamulli D, Mian C, Petricca D, et al. Influence of physiological dietary selenium supplementation on the natural course of autoimmune thyroiditis. Clin Endocrinol (Oxf). 2010; 73(4): 535–539, doi: 10.1111/j.1365-2265.2009.03758.x, indexed in Pubmed: 20039895.
  135. Krysiak R, Okopien B. The effect of levothyroxine and selenomethionine on lymphocyte and monocyte cytokine release in women with Hashimoto’s thyroiditis. J Clin Endocrinol Metab. 2011; 96(7): 2206–2215, doi: 10.1210/jc.2010-2986, indexed in Pubmed: 21508145.
  136. Onal H, Keskindemirci G, Adal E, et al. Effects of selenium supplementation in the early stage of autoimmune thyroiditis in childhood: an openlabel pilot study. J Pediatr Endocrinol Metab. 2012; 25(7-8): 639–644, doi: 10.1515/jpem-2012-0078, indexed in Pubmed: 23155687.
  137. Anastasilakis AD, Toulis KA, Nisianakis P, et al. Selenomethionine treatment in patients with autoimmune thyroiditis: a prospective, quasirandomised trial. Int J Clin Pract. 2012; 66(4): 378–383, doi: 10.1111/j.1742-1241.2011.02879.x, indexed in Pubmed: 22356267.
  138. Deng SY, Chen XY, Wu LY, et al. Influence of seleniumon Hashimoto thyroiditis with normal thyroid function. Chinese General Practice. 2013; 70: 2483–2485.
  139. Zhang W, Wang JF, Li JL, et al. The effect of seleniumon thyroid peroxidase antibody and thyroglobulin antibody in Hashimoto thyroiditis. Medical Innovation of China. 2013; 10: 13–14.
  140. Eskes SA, Endert E, Fliers E, et al. Selenite supplementation in euthyroid subjects with thyroid peroxidase antibodies. Clin Endocrinol (Oxf). 2014; 80(3): 444–451, doi: 10.1111/cen.12284, indexed in Pubmed: 23844613.
  141. Calissendorff J, Mikulski E, Larsen EH, et al. A Prospective Investigation of Graves’ Disease and Selenium: Thyroid Hormones, Auto-Antibodies and Self-Rated Symptoms. Eur Thyroid J. 2015; 4(2): 93–98, doi: 10.1159/000381768, indexed in Pubmed: 26279994.
  142. Pilli T, Cantara S, Schomburg L, et al. IFNγ-Inducible Chemokines Decrease upon Selenomethionine Supplementation in Women with Euthyroid Autoimmune Thyroiditis: Comparison between Two Doses of Selenomethionine (80 or 160 μg) versus Placebo. Eur Thyroid J. 2015; 4(4): 226–233, doi: 10.1159/000439589, indexed in Pubmed: 26835425.
  143. de Farias CR, Cardoso BR, de Oliveira GMB, et al. A randomized-controlled, double-blind study of the impact of selenium supplementation on thyroid autoimmunity and inflammation with focus on the GPx1 genotypes. J Endocrinol Invest. 2015; 38(10): 1065–1074, doi: 10.1007/ s40618-015-0285-8, indexed in Pubmed: 25894865.
  144. Wang L, Wang B, Chen SR, et al. Effect of Selenium Supplementation on Recurrent Hyperthyroidism Caused by Graves’ Disease: A Prospective Pilot Study. Horm Metab Res. 2016; 48(9): 559–564, doi: 10.1055/s-0042-110491, indexed in Pubmed: 27392116.
  145. Marcocci C, Kahaly GJ, Krassas GE, et al. European Group on Graves’ Orbitopathy. Selenium and the course of mild Graves’ orbitopathy. N Engl J Med. 2011; 364(20): 1920–1931, doi: 10.1056/NEJMoa1012985, indexed in Pubmed: 21591944.
  146. Bartalena L, Baldeschi L, Boboridis K, et al. European Group on Graves’ Orbitopathy (EUGOGO). The 2016 European Thyroid Association/ European Group on Graves’ Orbitopathy Guidelines for the Management of Graves’ Orbitopathy. Eur Thyroid J. 2016; 5(1): 9–26, doi: 10.1159/000443828, indexed in Pubmed: 27099835.
  147. Ruchała M, Hernik A, Zybek A. Orbital radiotherapy in the management of Graves’ orbitopathy – current state of knowledge. Endokrynol Pol. 2014; 65(5): 388–396, doi: 10.5603/EP.2014.0054, indexed in Pubmed: 25301490.
  148. Watt T, Cramon P, Bjorner JB, et al. Selenium supplementation for patients with Graves’ hyperthyroidism (the GRASS trial): study protocol for a randomized controlled trial. Trials. 2013; 14: 119, doi: 10.1186/17456215-14-119, indexed in Pubmed: 23782950.
  149. Winther KH, Watt T, Bjørner JB, et al. The chronic autoimmune thyroiditis quality of life selenium trial (CATALYST): study protocol for a randomized controlled trial. Trials. 2014; 15: 115, doi: 10.1186/17456215-15-115, indexed in Pubmed: 24716668.
  150. Zygmunt A, Adamczewski Z, Zygmunt A, et al. An assessment of the effectiveness of iodine prophylaxis in pregnant women – analysis in one of reference gynaecological-obstetric centres in Poland. Endokrynol Pol. 2015; 66(5): 404–411, doi: 10.5603/EP.2015.0050, indexed in Pubmed: 26457494.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl