Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Am J Physiol Endocrinol Metab ; 327(3): E328-E343, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39046281

ABSTRACT

One of the sensitive markers for autoimmune thyroid disease (AITD) clinical identification is thyroid-stimulating hormone receptor antibodies (TRAbs). To quickly distinguish TRAb with distinct antigenic epitopes, a straightforward and uncomplicated technique has not yet been created. The objective of this study is to search for molecular diagnostic targets for different types of AITD {Graves' disease (GD), Graves' orbitopathy (GO), GD with third-degree goiter [GD(3)], hypothyroidism combined with positive TRAb [HT(TRAb+)]} as molecular diagnostic targets. Following action on thyroid cells, differential genes (DEGs) generated by TRAb with distinct antigenic epitopes were detected and identified by RNA sequencing (RNA-Seq), bioinformatics analysis, and quantitative reverse transcription-polymerase chain reaction (RT-qPCR) in the serum of patients with AITD. Using the 5-ethynyl-2'-deoxyuridine (EdU) assay, the effect of coculturing thyroid cells with different antigenic TRAb epitopes on the cells' capacity to proliferate was investigated. Bioinformatics analysis and RT-qPCR validation identified one GD key gene alpha 2-HS glycoprotein (AHSG), two GO key genes [adrenoceptor alpha 1D (ADRA1D) and H2B clustered histone 18 (H2BC18)], two GD(3) key genes [suppressor of cytokine signaling 1 (SOCS1) and cytochrome b-245 beta (CYBB)], and one HT(TRAb+) key gene (MASP2). Correlation analysis and ROC curves showed that the abovementioned genes could be used as molecular diagnostic targets for different types of AITD. Finally, EdU results showed that TRAb inhibited thyroid cell proliferation in the HT(TRAb+) group compared with the normal control group, whereas the remaining three groups promoted thyroid cell proliferation, with a statistically significant difference (P < 0.05). We identified six key genes for different types of AITD, which have diagnostic value for different types of AITD. Meanwhile, we found that TRAbs with different antigenic epitopes in AITD have different biological functions.NEW & NOTEWORTHY We identified six molecular targets of different types of AITD [GD, GO, GD(3), and HT(TRAb+)], which have diagnostic value for different types of AITD. Meanwhile, we found that TRAb with different antigenic epitopes extracted from the sera of patients with AITD had different biological functions, which also provided a new idea for further research on the mechanism of action of TRAb with different antigenic epitopes in AITD.


Subject(s)
Epitopes , Graves Disease , Receptors, Thyrotropin , Humans , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/genetics , Epitopes/immunology , Graves Disease/immunology , Graves Disease/blood , Graves Disease/diagnosis , Graves Ophthalmopathy/immunology , Graves Ophthalmopathy/genetics , Graves Ophthalmopathy/blood , Autoantibodies/immunology , Autoantibodies/blood , Female , Male , Immunoglobulins, Thyroid-Stimulating/blood , Immunoglobulins, Thyroid-Stimulating/immunology , Thyroid Gland/immunology , Adult , Middle Aged , Cell Proliferation , Thyroiditis, Autoimmune/immunology , Thyroiditis, Autoimmune/genetics , Hypothyroidism/immunology
2.
Nature ; 609(7928): 854-859, 2022 09.
Article in English | MEDLINE | ID: mdl-35940204

ABSTRACT

Thyroid-stimulating hormone (TSH), through activation of its G-protein-coupled thyrotropin receptor (TSHR), controls the synthesis of thyroid hormone-an essential metabolic hormone1-3. Aberrant signalling of TSHR by autoantibodies causes Graves' disease (hyperthyroidism) and hypothyroidism, both of which affect millions of patients worldwide4. Here we report the active structures of TSHR with TSH and the activating autoantibody M225, both bound to the allosteric agonist ML-1096, as well as an inactivated TSHR structure with the inhibitory antibody K1-707. Both TSH and M22 push the extracellular domain (ECD) of TSHR into an upright active conformation. By contrast, K1-70 blocks TSH binding and cannot push the ECD into the upright conformation. Comparisons of the active and inactivated structures of TSHR with those of the luteinizing hormone/choriogonadotropin receptor (LHCGR) reveal a universal activation mechanism of glycoprotein hormone receptors, in which a conserved ten-residue fragment (P10) from the hinge C-terminal loop mediates ECD interactions with the TSHR transmembrane domain8. One notable feature is that there are more than 15 cholesterols surrounding TSHR, supporting its preferential location in lipid rafts9. These structures also highlight a similar ECD-push mechanism for TSH and autoantibody M22 to activate TSHR, therefore providing the molecular basis for Graves' disease.


Subject(s)
Immunoglobulins, Thyroid-Stimulating , Receptors, Thyrotropin , Thyrotropin , Graves Disease/immunology , Graves Disease/metabolism , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Membrane Microdomains , Receptors, LH , Receptors, Thyrotropin/agonists , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/metabolism , Thyrotropin/metabolism
3.
Nature ; 609(7928): 846-853, 2022 09.
Article in English | MEDLINE | ID: mdl-35940205

ABSTRACT

Thyroid hormones are vital in metabolism, growth and development1. Thyroid hormone synthesis is controlled by thyrotropin (TSH), which acts at the thyrotropin receptor (TSHR)2. In patients with Graves' disease, autoantibodies that activate the TSHR pathologically increase thyroid hormone activity3. How autoantibodies mimic thyrotropin function remains unclear. Here we determined cryo-electron microscopy structures of active and inactive TSHR. In inactive TSHR, the extracellular domain lies close to the membrane bilayer. Thyrotropin selects an upright orientation of the extracellular domain owing to steric clashes between a conserved hormone glycan and the membrane bilayer. An activating autoantibody from a patient with Graves' disease selects a similar upright orientation of the extracellular domain. Reorientation of the extracellular domain transduces a conformational change in the seven-transmembrane-segment domain via a conserved hinge domain, a tethered peptide agonist and a phospholipid that binds within the seven-transmembrane-segment domain. Rotation of the TSHR extracellular domain relative to the membrane bilayer is sufficient for receptor activation, revealing a shared mechanism for other glycoprotein hormone receptors that may also extend to other G-protein-coupled receptors with large extracellular domains.


Subject(s)
Cryoelectron Microscopy , Immunoglobulins, Thyroid-Stimulating , Receptors, Thyrotropin , Thyrotropin , Cell Membrane/metabolism , Graves Disease/immunology , Graves Disease/metabolism , Humans , Immunoglobulins, Thyroid-Stimulating/chemistry , Immunoglobulins, Thyroid-Stimulating/immunology , Immunoglobulins, Thyroid-Stimulating/pharmacology , Immunoglobulins, Thyroid-Stimulating/ultrastructure , Phospholipids/metabolism , Protein Domains , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/ultrastructure , Receptors, Thyrotropin/agonists , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/ultrastructure , Rotation , Thyrotropin/chemistry , Thyrotropin/metabolism , Thyrotropin/pharmacology
4.
J Clin Endocrinol Metab ; 107(3): 743-754, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34687316

ABSTRACT

CONTEXT: Remission rates in young people with Graves hyperthyroidism are less than 25% after 2 years of thionamide antithyroid drug (ATD). OBJECTIVE: We explored whether rituximab (RTX), a B-lymphocyte-depleting agent, would increase remission rates when administered with a short course of ATD. METHODS: This was an open-label, multicenter, single-arm, phase 2 trial in young people (ages, 12-20 years) with Graves hyperthyroidism. An A'Hern design was used to distinguish an encouraging remission rate (40%) from an unacceptable rate (20%). Participants presenting with Graves hyperthyroidism received 500 mg RTX and 12 months of ATD titrated according to thyroid function. ATDs were stopped after 12 months and primary outcome assessed at 24 months. Participants had relapsed at 24 months if thyrotropin was suppressed and free 3,5,3'-triiodothyronine was raised; they had received ATD between months 12 and 24; or they had thyroid surgery/radioiodine. RESULTS: A total of 27 participants were recruited and completed the trial with no serious side effects linked to treatment. Daily carbimazole dose at 12 months was less than 5 mg in 21 of 27 participants. Thirteen of 27 participants were in remission at 24 months (48%, 90% one-sided CI, 35%-100%); this exceeded the critical value (9) for the A'Hern design and provided evidence of a promising remission rate. B-lymphocyte count at 28 weeks, expressed as a percentage of baseline, was related to likelihood of remission. CONCLUSION: Adjuvant RTX, administered with a 12-month course of ATD, may increase the likelihood of remission in young people with Graves hyperthyroidism. A randomized trial of adjuvant RTX in young people with Graves hyperthyroidism is warranted.


Subject(s)
Antithyroid Agents/therapeutic use , Graves Disease/drug therapy , Immunologic Factors/therapeutic use , Propylthiouracil/therapeutic use , Rituximab/therapeutic use , Adolescent , Child , Drug Therapy, Combination/methods , Female , Graves Disease/blood , Graves Disease/diagnosis , Graves Disease/immunology , Humans , Immunoglobulins, Thyroid-Stimulating/blood , Immunoglobulins, Thyroid-Stimulating/immunology , Male , Recurrence , Treatment Outcome , Young Adult
5.
J Autoimmun ; 122: 102666, 2021 08.
Article in English | MEDLINE | ID: mdl-34144327

ABSTRACT

BACKGROUND: BALB/c mice which received long-term immunizations of adenovirus (Ad) expressing thyrotropin receptor A-subunits (TSHR) developed stable Graves' disease (GD). TSHR-derived cyclic peptide 19 (P19) was identified as effective therapy in this model. METHODS: In Ad-TSHR mice, we investigated shorter disease intervals up to 4 months for histological alterations of the orbits, fine tuning of anti-TSHR antibodies (Ab) and free thyroxine (fT4) hormone levels by using novel detection methods in an independent laboratory. Therapy (0.3 mg/kg P19 or vehicle) was given intravenously after the fourth Ad-TSHR immunization (week 11) and continued until week 19. RESULTS: Thyrotropin binding inhibitory immunoglobulins (TBII, bridge immunoassay), blocking (TBAb) and stimulating (TSAb) TSHR-Ab (both cell-based bioassays) and serum levels of fT4 were significantly elevated at week 11 in Ad-TSHR-immunized mice versus none in control mice. For the first time, TSAb, TBAb, and thyroperoxidase-Ab were detected in 17 of 19, 12/19 and 6/19 Ad-TSHR immunized mice, respectively at week 21. Also, for the first time, this study showed that P19 treatment markedly reduced serum TBII (p < 0.0001), serum fT4 (p = 0.02), and acidic mucins and collagen content in the orbital tissue of Ad-TSHR-immunized mice. CONCLUSION: P19 significantly improved thyroid function, confirming previous results in an independent second laboratory. A relevant shift of anti-TSHR antibody subpopulations in response to P19 therapy may help explain its immunological effects. Moreover, P19 exerted a beneficial effect on mucine and collagen content of orbital tissue. Hence, P19 offers a potential novel therapeutic approach for GD and associated orbitopathy.


Subject(s)
Graves Disease/drug therapy , Graves Ophthalmopathy/drug therapy , Peptides, Cyclic/pharmacology , Animals , Collagen/analysis , Disease Models, Animal , Female , Graves Disease/blood , Graves Disease/immunology , Graves Disease/physiopathology , Graves Ophthalmopathy/immunology , Graves Ophthalmopathy/pathology , Humans , Immunoglobulins, Thyroid-Stimulating/blood , Immunoglobulins, Thyroid-Stimulating/immunology , Mice , Mucins/analysis , Orbit/drug effects , Orbit/pathology , Peptides, Cyclic/genetics , Peptides, Cyclic/therapeutic use , Receptors, Thyrotropin/administration & dosage , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Thyroid Gland/drug effects , Thyroid Gland/immunology , Thyroid Gland/physiopathology
6.
Medicine (Baltimore) ; 100(16): e25554, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33879707

ABSTRACT

BACKGROUND: Thyroid autoimmune disease (TAI) has been verified to be related to multiple adverse pregnancy outcomes. A growing number of evidences highlight the protective roles of glucocorticoid on the treatments of TAI. This meta-analysis aimed to study whether it is beneficial to add glucocorticoid treatment in infertile women with TAI when they are undergoing assisted reproductive technology (ART). METHODS: We conducted a systematic search in PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure (CNKI), WanFang database, Weipu China Science and Technology Journal Databases (VIP database) up to September 10, 2020. The Revman 5.3 software was utilized for data statistics. We used a random-effects model to analyze data and the odds ratio (OR) combining with 95% confidence interval (95% CI) were employed to reveal the results. RESULTS: Three publications with 237 antithyroid antibody (ATA)-positive and 384 ATA-negative women were included in the final analysis. Overall, glucocorticoid therapy showed satisfying effects on improving clinical pregnancy rate (OR = 4.63, 95% CI [2.23, 9.58], I2 = 0.0%, P < .0001) and live birth rate (OR = 3.19, 95% CI [1.13, 9.04], I2 = 0.0%, P = .03) of ATA-positive women compared with control group. However, it seems that glucocorticoid showed no significant difference in the abortion rate (OR = 0.62, 95% CI [0.09, 4.32], I2 = 35%, P = .64) and oocyte recovery (OR = 2.26, 95% CI [-1.46, 5.99], I2 = 79%, P < .0001) between the 2 groups. CONCLUSIONS: Glucocorticoid may improve the pregnancy outcomes of ART women with ATA positive, but there is no significant reduction in the risk of miscarriage. Due to the limited enrolled references, glucocorticoid adjuvant therapy should be applied after more randomized controlled trials.


Subject(s)
Dietary Supplements , Glucocorticoids/administration & dosage , Infertility, Female/therapy , Reproductive Techniques, Assisted , Thyroiditis, Autoimmune/therapy , Adult , Autoantibodies/blood , Female , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Infertility, Female/blood , Infertility, Female/immunology , Pregnancy , Pregnancy Outcome , Prospective Studies , Retrospective Studies , Thyroiditis, Autoimmune/blood , Thyroiditis, Autoimmune/immunology , Treatment Outcome
7.
Front Endocrinol (Lausanne) ; 12: 788535, 2021.
Article in English | MEDLINE | ID: mdl-35069441

ABSTRACT

Graves' Ophthalmopathy (GO) is an organ-specific autoimmune disease that is often characterized by infiltration of orbital tissues and is considered as the most common extra-thyroid manifestation of Graves' disease (GD). Although genetic susceptibility has been found to be critical for the phenotype of GO, the associated risk alleles in a single gene are generally insufficient to cause the disease. Accruing evidence has shown that epigenetic disorders can act as the potentially missing link between genetic risk and clinically significant disease development. Abnormal epigenetic modifications can lead to pro-inflammatory cascades and activation of orbital fibroblasts (OFs) by promoting the various inflammatory response pathways and regulating the diverse signaling molecules that are involved in the fibrogenesis and adipogenesis, thereby leading to the significant expansion of orbital tissues, fibrosis and inflammation infiltration. Additionally, emerging evidence has shown that the gut microbiome can possibly drive the pathogenesis of GO by influencing the secretion of Thyrotropin receptor antibody (TRAb) and T-helper 17 (Th17)/regulatory T cells (Treg) imbalance. This paper describes the latest epigenetic research evidence and progress made in comprehending the mechanisms of GO development, such as DNA methylation, histone modification, non-coding RNAs, and the gut microbiome.


Subject(s)
Epigenesis, Genetic , Fibroblasts/metabolism , Gastrointestinal Microbiome , Graves Ophthalmopathy/genetics , Inflammation/genetics , Adipogenesis , DNA Methylation/genetics , Fibroblasts/immunology , Fibrosis , Graves Ophthalmopathy/immunology , Graves Ophthalmopathy/microbiology , Histone Code/genetics , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
8.
J Clin Endocrinol Metab ; 105(12)2020 12 01.
Article in English | MEDLINE | ID: mdl-32929476

ABSTRACT

CONTEXT: Invited update on the management of systemic autoimmune Graves disease (GD) and associated Graves orbitopathy (GO). EVIDENCE ACQUISITION: Guidelines, pertinent original articles, systemic reviews, and meta-analyses. EVIDENCE SYNTHESIS: Thyrotropin receptor antibodies (TSH-R-Abs), foremost the stimulatory TSH-R-Abs, are a specific biomarker for GD. Their measurement assists in the differential diagnosis of hyperthyroidism and offers accurate and rapid diagnosis of GD. Thyroid ultrasound is a sensitive imaging tool for GD. Worldwide, thionamides are the favored treatment (12-18 months) of newly diagnosed GD, with methimazole (MMI) as the preferred drug. Patients with persistently high TSH-R-Abs and/or persistent hyperthyroidism at 18 months, or with a relapse after completing a course of MMI, can opt for a definitive therapy with radioactive iodine (RAI) or total thyroidectomy (TX). Continued long-term, low-dose MMI administration is a valuable and safe alternative. Patient choice, both at initial presentation of GD and at recurrence, should be emphasized. Propylthiouracil is preferred to MMI during the first trimester of pregnancy. TX is best performed by a high-volume thyroid surgeon. RAI should be avoided in GD patients with active GO, especially in smokers. Recently, a promising therapy with an anti-insulin-like growth factor-1 monoclonal antibody for patients with active/severe GO was approved by the Food and Drug Administration. COVID-19 infection is a risk factor for poorly controlled hyperthyroidism, which contributes to the infection-related mortality risk. If GO is not severe, systemic steroid treatment should be postponed during COVID-19 while local treatment and preventive measures are offered. CONCLUSIONS: A clear trend towards serological diagnosis and medical treatment of GD has emerged.


Subject(s)
Disease Management , Graves Disease/diagnosis , Graves Ophthalmopathy/diagnosis , Hyperthyroidism/diagnosis , Immunoglobulins, Thyroid-Stimulating/blood , Antithyroid Agents/therapeutic use , Biomarkers/blood , Diagnosis, Differential , Female , Graves Disease/complications , Graves Disease/therapy , Graves Ophthalmopathy/etiology , Graves Ophthalmopathy/therapy , Humans , Hyperthyroidism/complications , Hyperthyroidism/therapy , Immunoglobulins, Thyroid-Stimulating/immunology , Iodine Radioisotopes/therapeutic use , Male , Methimazole/therapeutic use , Pregnancy , Pregnancy Complications/diagnosis , Pregnancy Complications/therapy , Receptors, Thyrotropin/immunology , Thyroid Gland/diagnostic imaging , Thyroid Gland/immunology , Thyroidectomy/methods , Ultrasonography
9.
Sci Rep ; 10(1): 5399, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32214213

ABSTRACT

Patients with acquired adult-onset strabismus mainly present with binocular diplopia. Although cranial nerve palsies are reportedly the most common cause of binocular diplopia in adults, thyroid disease can also cause diplopia. In patients with thyroid-associated ophthalmopathy, upper lid retraction and proptosis are the most common initial findings, but diplopia could be the first manifestation. So far, there has been little information on the diagnostic value of thyroid autoantibodies in patients with strabismus. Therefore, we examined adults with acquired binocular diplopia from 2008 to 2016 and evaluated the presence of thyroid autoantibodies and the relationship between thyroid autoantibody status and clinical characteristics in adults with acquired binocular diplopia. Thyroid autoantibody tests were performed for all patients, unless other causes of diplopia were identified. Fifty one (39%) of 132 patients were positive for thyroid autoantibodies. In the thyroid autoantibody-positive (TAb+) group, microsomal autoantibodies, thyroid-stimulating hormone receptor antibodies, thyroglobulin antibodies, and thyroid-stimulating antibodies were observed in 30, 27, 12, and 7 patients, respectively. The vertical deviation and grade of duction limitation were greater in the TAb+ group. The presence of ocular torsion was 15.5% and 39.5% in the TAb- and TAb+ groups, respectively. Thyroid autoantibody evaluation may be helpful in adults with idiopathic acquired binocular diplopia.


Subject(s)
Autoantibodies/immunology , Diplopia/immunology , Thyroid Gland/immunology , Vision, Binocular/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Exophthalmos/immunology , Female , Graves Ophthalmopathy/immunology , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Male , Middle Aged , Oculomotor Muscles/immunology , Retrospective Studies , Strabismus/immunology , Young Adult
10.
Ann Acad Med Singap ; 48(6): 181-187, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31377762

ABSTRACT

INTRODUCTION: Our study aimed to identify the factors associated with successful first-time radioactive iodine (RAI) treatment in patients with Graves' disease (GD). MATERIALS AND METHODS: This is a restrospective study of patients with GD who were treated with RAI. Treatment success was defined as onset of permanent hypothyroidism or euthyroidism after 1 dose of RAI at 1-year follow-up. RESULTS: There were 388 GD patients who underwent RAI treatment between January 2014 and December 2015. Of these, 74% achieved treatment success. Median time to achieve permanent hypothyroidism was 2 months. Male gender, smoking, higher antithyroid drug dosage, lower thyroid stimulating hormone (TSH) level, large goitre size and TSH receptor antibody (TRAb) titre at time of RAI were significantly associated with treatment failure. Multivariate analysis showed that larger goitre size and higher TRAb titre were associated with lower first-time RAI success. CONCLUSION: Larger goitre size and higher TRAb titre predict lower success of RAI therapy in GD patients. Treatment decisions and strategies should be customised for patients who present with these characteristics.


Subject(s)
Graves Disease/radiotherapy , Immunoglobulins, Thyroid-Stimulating/immunology , Iodine Radioisotopes/therapeutic use , Adult , Female , Goiter/pathology , Graves Disease/immunology , Graves Disease/pathology , Humans , Male , Middle Aged , Organ Size , Prognosis , Thyroid Gland/pathology , Treatment Failure
11.
Endocr J ; 66(11): 1001-1009, 2019 Nov 28.
Article in English | MEDLINE | ID: mdl-31308303

ABSTRACT

It is known that there is a relationship between some diseases and blood groups. The objective of our study is to investigate how often ABO and Rh blood groups are seen in benign thyroid diseases, especially in autoimmune-mediated thyroid diseases, and hence whether there is an association between blood groups and thyroid diseases. A total of 958 patients who were followed due to any benign thyroid disease were included in the study. The study population comprised 958 patients, 550 with Hashimoto's hypothyroidism, 160 with non-Hashimoto's hypothyroidism, 103 with iatrogenic hypothyroidism, 93 with central hypothyroidism, and 28 with Graves' and 24 with non-Graves' hyperthyroidism. Of the patients, 47.1% belonged to the O blood group, 30% to the A blood group, 15.2% to the B blood group, and 7.7% to the AB blood group while 90% were Rh-positive. The ratio of those with the O blood group was determined to be significantly higher in the Hashimoto's hypothyroidism group compared to the other disease groups. In the non-Hashimoto's hypothyroidism group, however, the ratio of the AB blood group was statistically significantly higher. While autoimmune diseases were more common in those with the O blood group, they were significantly lower in the AB blood group (p < 0.001). In our study, we determined that the ratio of the O blood group was significantly higher among patients with hypothyroidism due to Hashimoto's thyroiditis. These findings imply that there might be a relation between O blood group and Hashimoto's thyroiditis.


Subject(s)
ABO Blood-Group System , Graves Disease/blood , Hashimoto Disease/blood , Hypothyroidism/blood , Rh-Hr Blood-Group System , Adult , Aged , Autoantibodies/immunology , Female , Graves Disease/immunology , Hashimoto Disease/immunology , Humans , Hyperthyroidism/blood , Hyperthyroidism/immunology , Hypothyroidism/immunology , Immunoglobulins, Thyroid-Stimulating/immunology , Iodide Peroxidase/immunology , Male , Middle Aged , Turkey
12.
Horm Metab Res ; 51(6): 341-346, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31207654

ABSTRACT

A rapid and fully automated chemiluminescent immunoassay for the detection of thyrotropin receptor autoantibodies (TSHR-Ab) based on a bridge technology was compared with two bioassays that measure either stimulating (TSAb) or blocking (TBAb) antibodies for the detection and differentiation of TSHR-Ab. A total of 229 patients with various thyroid disorders [151 with Graves' disease (GD), 35 with Hashimoto's thyroiditis (HT), 32 with nodular goiter, and 11 with thyroid cancer] were included. The bridge immunoassay was performed according to the manufacturer's instructions (cut-off>0.55 IU/l). TSAb and TBAb were measured with reporter bioassays. Blocking activity was defined as percent inhibition of luciferase expression relative to induction with bovine TSH alone (cut-off>34% inhibition). TSAb was reported as percentage of specimen-to-reference ratio (> 140 SRR%). The 3 TSHR-Ab assays were negative in all patients with benign euthyroid nodular goiter and differentiated thyroid cancer. In contrast, in all patients with GD, irrespective of the disease duration, TSHR-Ab positivity was present in 127 of 151 (84%) and 140 (93%) for the bridge assay and TSAb bioassay, respectively (p<0.001). Fifteen of 151 (10%) GD samples were positive in the TSAb bioassay but negative in the bridge assay. The bridge assay and the TSAb bioassay correlated positively (r=0.39, p<0.0001) in patients with GD. Both assays detected TSHR-Ab in all ten untreated hyperthyroid patients with GD. In GD patients with a duration of less than six months, 27/29 (93%) and 28 (97%) were TSHR-Ab positive with the bridge and TSAb bioassay, respectively. In comparison, TSHR-Ab were present in two of 35 (6%) and five (14%) HT patients with the bridge and TSAb bio-assay, respectively. TSHR blocking antibodies were present in one (3%) patient with HT and in two (1%) patients with GD; these two GD patients were also bridge assay positive but TSAb bioassay negative. In conclusion, the bridge immunoassay and both bioassays are highly sensitive for the detection of TSHR-Ab. The bridge assay is, however, also positive in the presence of TSHR blocking antibodies detected in a TBAb bioassay.


Subject(s)
Antibodies/immunology , Graves Disease/diagnosis , Hashimoto Disease/diagnosis , Immunoassay/methods , Immunoglobulins, Thyroid-Stimulating/blood , Receptors, Thyrotropin/immunology , Thyroid Neoplasms/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cell Differentiation , Female , Goiter, Nodular/blood , Goiter, Nodular/diagnosis , Goiter, Nodular/immunology , Graves Disease/blood , Graves Disease/immunology , Hashimoto Disease/blood , Hashimoto Disease/immunology , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Male , Middle Aged , Prognosis , Thyroid Neoplasms/blood , Thyroid Neoplasms/immunology , Young Adult
13.
Thyroid ; 29(8): 1138-1146, 2019 08.
Article in English | MEDLINE | ID: mdl-31184281

ABSTRACT

Background: Graves' disease, caused by autoantibodies that activate the thyrotropin (TSH) receptor (TSHR), has only been reported in humans. Thyroiditis-prone NOD.H2h4 mice develop autoantibodies to thyroglobulin (Tg) and thyroid peroxidase (TPO) but not to the TSHR. Evidence supports the importance of the shed TSHR A-subunit in the initiation and/or amplification of the autoimmune response to the holoreceptor. Cells expressing the gene for the isolated A-subunit secrete A-subunit protein, a surrogate for holoreceptor A-subunit shedding. NOD.H2h4 mice with the human TSHR A-subunit targeted to the thyroid (a "self" antigen in such transgenic (Tgic) animals), unlike their wild-type (wt) siblings, spontaneously develop pathogenic TSHR antibodies to the human-TSH holoreceptor. These autoantibodies do not recognize the endogenous mouse-TSH holoreceptor and do not cause hyperthyroidism. Methods: We have now generated NOD.H2h4 mice with the mouse-TSHR A-subunit transgene targeted to the thyroid. Tgic mice and wt littermates were compared for intrathyroidal expression of the mouse A-subunit. Sera from six-month-old mice were tested for the presence of autoantibodies to Tg and TPO as well as for pathogenic TSHR antibodies (TSH binding inhibition, bioassay for thyroid stimulating antibodies) and nonpathogenic TSHR antibodies (ELISA). Results: Expression of the mouse TSHR A-subunit transgene in the thyroid was confirmed by real-time polymerase chain reaction in the Tgics and had no effect on the spontaneous development of autoantibodies to Tg or TPO. However, unlike the same NOD.H2h4 strain with the human-TSHR A-subunit target to the thyroid, mice expressing intrathyroidal mouse-TSHR A subunit failed to develop either pathogenic or nonpathogenic TSHR antibodies. The mouse TSHR A-subunit differs from the human TSHR A-subunit in terms of its amino acid sequence and has one less glycosylation site than the human TSHR A-subunit. Conclusions: Multiple genetic and environmental factors contribute to the pathogenesis of Graves' disease. The present study suggests that the TSHR A-subunit structure (possibly including posttranslational modification such as glycosylation) may explain, in part, why Graves' disease only develops in humans.


Subject(s)
Graves Disease/genetics , Immunoglobulins, Thyroid-Stimulating/immunology , Protein Subunits/genetics , Receptors, Thyrotropin/genetics , Animals , Autoantibodies/immunology , Glycosylation , Graves Disease/immunology , Humans , Iodide Peroxidase/immunology , Mice , Mice, Transgenic , Protein Subunits/immunology , Receptors, Thyrotropin/immunology , Thyroglobulin/immunology , Thyroiditis
14.
Recenti Prog Med ; 110(5): 255-258, 2019 05.
Article in Italian | MEDLINE | ID: mdl-31140459

ABSTRACT

We report the case of a 67-year-old man, with a past medical history of radiotherapy for nasopharyngeal carcinoma, who presented with the classical features of a hyperthyroidism (H), due to Graves' disease, with a high TSH receptor antibodies (TRAb) titre. Thyrostatic therapy was started, with gradual improvement of the symptoms and of the thyroid function tests. Two years later, TRAb became negative and the therapy was stopped. In the following months a previously unknown anterior pituitary insufficiency became evident. Therapy with cortisone acetate, L-thyroxine and testosterone was started, resulting in prolonged normalization of the clinical picture. Six years later a short relapse of H was observed, simultaneously to a new increase of TRAb titre, requiring the transitory interruption of the L-thyroxine therapy. In a few months span H disappeared and central hypothyroidism manifested again, so that the patient is still taking replacement therapy. This case illustrates how H and hypopituitarism are not mutually exclusive diagnoses and how, even if rarely, central hypothyroidism and H could alternate in the clinical history of the same patient.


Subject(s)
Hyperthyroidism/diagnosis , Hypopituitarism/diagnosis , Immunoglobulins, Thyroid-Stimulating/immunology , Aged , Cortisone/administration & dosage , Graves Disease/diagnosis , Humans , Male , Testosterone/administration & dosage , Thyroxine/administration & dosage
16.
Endocrinology ; 160(5): 1008-1020, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30822352

ABSTRACT

The TSH receptor (TSHR) is the major autoantigen in Graves' disease (GD). Bioinformatic analyses predict the existence of several human TSHR isoforms from alternative splicing, which can lead to the coexpression of multiple receptor forms. The most abundant of these is TSHRv1.3. In silico modeling of TSHRv1.3 demonstrated the structural integrity of this truncated receptor isoform and its potential binding of TSH. Tissue profiling revealed wide expression of TSHRv1.3, with a predominant presence in thyroid, bone marrow, thymus, and adipose tissue. To gain insight into the role of this v1.3 receptor isoform in thyroid pathophysiology, we cloned the entire open reading frame into a mammalian expression vector. Immunoprecipitation studies demonstrated that both TSHR-stimulating antibody and human TSH could bind v1.3. Furthermore, TSHRv1.3 inhibited the stimulatory effect of TSH and TSHR-Ab MS-1 antibody on TSHR-induced cAMP generation in a dose-dependent manner. To confirm the antigenicity of v1.3, we used a peptide ELISA against two different epitopes. Of 13 GD samples, 11 (84.6%) were positive for a carboxy terminal peptide and 10 (76.9%) were positive with a junction region peptide. To demonstrate that intracellular v1.3 could serve as an autoantigen and modulate disease, we used double-transfected Chinese hamster ovary cells that expressed both green fluorescent protein (GFP)-tagged TSHRv1.3 and full-length TSHR. We then induced cell stress and apoptosis using a TSHR monoclonal antibody and observed the culture supernatant contained v1.3-GFP protein, demonstrating the release of the intracellular receptor variant by this mechanism.


Subject(s)
Autoantibodies/immunology , Autoantigens/metabolism , Graves Disease/metabolism , Immunoglobulins, Thyroid-Stimulating/metabolism , Receptors, Thyrotropin/metabolism , Amino Acid Sequence , Animals , Autoantibodies/metabolism , Base Sequence , CHO Cells , Cricetinae , Cricetulus , Graves Disease/genetics , Graves Disease/immunology , HEK293 Cells , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Molecular Dynamics Simulation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/immunology , Thyrotropin/chemistry , Thyrotropin/metabolism
17.
BMJ Open ; 9(1): e024705, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30670519

ABSTRACT

INTRODUCTION: Graves' disease (Graves' hyperthyroidism) is a challenging condition for the young person and their family. The excess thyroid hormone generated by autoimmune stimulation of the thyroid stimulating hormone receptor on the thyroid gland can have a profound impact on well-being. Managing the young person with Graves' hyperthyroidism is more difficult than in older people because the side effects of conventional treatment are more significant in this age group and because the disease tends not to resolve spontaneously in the short to medium term. New immunomodulatory agents are available and the anti-B cell monoclonal antibody rituximab is of particular interest because it targets cells that manufacture the antibodies that stimulate the thyroid gland in Graves'. METHODS AND ANALYSIS: The trial aims to establish whether the combination of a single dose of rituximab (500 mg) and a 12-month course of antithyroid drug (usually carbimazole) can result in a meaningful increase in the proportion of patients in remission at 2 years, the primary endpoint. A single-stage, phase II A'Hern design is used. 27 patients aged 12-20 years with newly presenting Graves' hyperthyroidism will be recruited. Markers of immune function, including lymphocyte numbers and antibody levels (total and specific), will be collected regularly throughout the trial. DISCUSSION: The trial will determine whether the immunomodulatory medication, rituximab, will facilitate remission above and beyond that observed with antithyroid drug alone. A meaningful increase in the expected proportion of young patients entering remission when managed according to the trial protocol will justify consideration of a phase III trial.Ethics and dissemination The trial has received a favourable ethical opinion (North East - Tyne and Wear South Research Ethics Committee, reference 16/NE/0253, EudraCT number 2016-000209-35). The results of this trial will be distributed at international endocrine meetings, in the peer-reviewed literature and via patient support groups. TRIAL REGISTRATION NUMBER: ISRCTN20381716.


Subject(s)
Antithyroid Agents/therapeutic use , Graves Disease/drug therapy , Immunologic Factors/therapeutic use , Rituximab/therapeutic use , Adolescent , Child , Drug Therapy, Combination , Graves Disease/blood , Graves Disease/immunology , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Lymphocyte Count , Thyrotropin/blood , Thyroxine/blood , Treatment Outcome , Triiodothyronine/blood , Young Adult
18.
Front Biosci (Landmark Ed) ; 24(1): 35-47, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30468646

ABSTRACT

Graves' disease (GD) is the most common cause for hyperthyroidism in iodine-replete areas. The disease is caused by the appearance of stimulating TSH receptor autoantibodies (TRAb) leading to hyperthyroidism. Blocking and neutral TRAb have, however, also been described. TRAb can be measured either by competition assays, assays using a bridge technology or bioassays (for discriminating stimulating vs. blocking antibodies). Therapy of GD with antithyroid drugs belonging to the group of thionamides is the first-line treatment to be continued for 12 up to 18 months. In case of relapse, thyroid ablative therapy including radioiodine therapy or thyroidectomy, respectively, should be performed. Risk factors for relapse are a large thyroid volume, persistence of high TRAb serum titer, smoking, and others. Within this review, we will give insights into the pathogenesis of GD including the pathogenesis of Graves' ophthalmopathy. We also describe recent developments of TRAb measurement, which is used for the diagnosis of GD as well as for outcome prediction. Finally, we discuss therapy aspects as well as the important issue of GD and pregnancy.


Subject(s)
Autoantibodies/immunology , Graves Disease/immunology , Immunoglobulins, Thyroid-Stimulating/immunology , Receptors, Thyrotropin/immunology , Antithyroid Agents/therapeutic use , Graves Disease/pathology , Graves Disease/therapy , Humans , Iodine Radioisotopes/therapeutic use , Receptors, Thyrotropin/metabolism , Recurrence , Thyroidectomy
19.
J Clin Endocrinol Metab ; 104(5): 1378-1385, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30517711

ABSTRACT

CONTEXT: Thyrotropin receptor antibodies (TRAbs) play a crucial role in the pathogenesis of Graves disease (GD). However, factors that influence the association of TRAbs with thyroid hormones and relapse risk in GD remain unclear. OBJECTIVE: We investigated the associations of TRAbs at diagnosis with thyroid hormones and relapse risk and potential factors that can influence these associations in GD. DESIGN AND SETTING: A prospective study in an endocrine center in England. PATIENTS AND MAIN OUTCOME MEASURES: Three hundred eighty-four consecutive patients with GD who had measurements of TRAbs and thyroid hormones at diagnosis. The association of TRAbs with thyroid hormones and relapse risk was assessed through linear regression and Cox proportional hazard models, adjusted for confounders. RESULTS: TRAbs were nonlinearly associated with thyroid hormones, following a curve with an initial positive slope and a subsequent flattening (P < 0.0001). Higher TRAbs were associated with greater relapse risk [hazard ratio (HR), 1.05 (95% CI, 1.02 to 1.08) per 1-U/L increase]. These associations were modified by age, but not by sex, race, smoking, or thyroid peroxidase antibody levels. In younger participants, increasing TRAbs were associated with higher thyroid hormones and greater relapse risk [HR, 1.13 (95% CI, 1.04 to 1.23) per 1-U/L increase]. In older participants, TRAbs were not associated with thyroid hormones or relapse risk [HR, 0.99 (95% CI, 0.93 to 1.05) per 1-U/L increase. CONCLUSIONS: In GD, age can influence the effect of TRAbs on thyroid function and relapse risk. TRAbs at diagnosis have better predictive value in younger patients with GD.


Subject(s)
Biomarkers/blood , Graves Disease/pathology , Immunoglobulins, Thyroid-Stimulating/blood , Receptors, Thyrotropin/immunology , Thyroid Gland/physiopathology , Thyroid Hormones/metabolism , Adult , Age Factors , Cross-Sectional Studies , Female , Follow-Up Studies , Graves Disease/blood , Graves Disease/immunology , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Male , Middle Aged , Prognosis , Prospective Studies , Recurrence , Thyroid Function Tests
20.
Proc Nutr Soc ; 78(1): 34-44, 2019 02.
Article in English | MEDLINE | ID: mdl-30208979

ABSTRACT

Hashimoto's thyroiditis (HT) and Graves' disease (GD) are examples of autoimmune thyroid disease (AITD), the commonest autoimmune condition. Antibodies to thyroid peroxidase (TPO), the enzyme that catalyses thyroid-hormone production and antibodies to the receptor for the thyroid-stimulating hormone, are characteristic of HT and GD, respectively. It is presently accepted that genetic susceptibility, environmental factors, including nutritional factors and immune disorders contribute to the development of AITD. Aiming to investigate the effect of iodine, iron and selenium in the risk, pathogenesis and treatment of thyroid disease, PubMed and the Cochrane Library were searched for relevant publications to provide a narrative review. Iodine: chronic exposure to excess iodine intake induces autoimmune thyroiditis, partly because highly-iodinated thyroglobulin (Tg) is more immunogenic. The recent introduction of universal salt iodisation can have a similar, although transient, effect. Iron: iron deficiency impairs thyroid metabolism. TPO is a haem enzyme that becomes active only after binding haem. AITD patients are frequently iron-deficient since autoimmune gastritis, which reduces iron absorption and coeliac disease which causes iron loss, are frequent co-morbidities. In two-thirds of women with persistent symptoms of hypothyroidism despite appropriate levothyroxine therapy, restoration of serum ferritin above 100 µg/l ameliorated symptoms. Selenium: selenoproteins are essential to thyroid action. In particular, the glutathione peroxidases remove excessive hydrogen peroxide produced there for the iodination of Tg to form thyroid hormones. There is evidence from observational studies and randomised controlled trials that selenium, probably as selenoproteins, can reduce TPO-antibody concentration, hypothyroidism and postpartum thyroiditis. Appropriate status of iodine, iron and selenium is crucial to thyroid health.


Subject(s)
Iodine/immunology , Iron/immunology , Selenium/immunology , Thyroiditis, Autoimmune/immunology , Autoantigens/immunology , Graves Disease/immunology , Hashimoto Disease/immunology , Humans , Hypothyroidism/immunology , Immunoglobulins, Thyroid-Stimulating/immunology , Iodide Peroxidase/immunology , Iron-Binding Proteins/immunology , Nutritional Status , Risk Factors , Sodium Chloride, Dietary
SELECTION OF CITATIONS
SEARCH DETAIL