Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Cell Mol Biol (Noisy-le-grand) ; 69(9): 67-74, 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37807332

ABSTRACT

The possibility that thyrotropin receptor (TSHR) expression in non-thyroid tissue is well-documented. However, there is insufficient data on the expression of TSHR in medulla oblongata regions, particularly when focusing on the background of encephalopathy associated with hyperthyroidism. In this study, we explored the expression of the functional TSHR in Graves' disease (GD) mouse cerebral vascular endothelial cells and the effects of thyrotropin receptor autoantibody (TRAb) on its expression. A mouse model of GD was constructed with an adenovirus overexpressing TSHR289. The location and expression of the TSHR gene and protein in vivo were determined via RT-qPCR, Western blot, and immunofluorescence techniques. The effect of TRAb on the expression of functional TSHR in vitro was investigated using bEnd.3 cells. Our results show that medulla oblongata vascular endothelial cells from GD mice expressed higher levels of TSHR compared to control mice. In an in vitro experiment, novel results demonstrated that after treatment with a monoclonal TSHR-specific agonistic antibody (M22), the expression of TSHR on the bEnd.3 cells increased at both the protein and mRNA levels. Furthermore, compared with bEnd.3 cells were treated with IBMX only, those treated with M22 showed increased cAMP production. This study suggested that TSHR is expressed and functionally active in the mouse medulla oblongata and in vitro-cultured bEnd.3 cells and TRAb (M22) increased the expression of TSHR on bEnd.3 cells.


Subject(s)
Graves Disease , Receptors, Thyrotropin , Animals , Mice , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Long-Acting Thyroid Stimulator/metabolism , Endothelial Cells/metabolism , Immunoglobulins, Thyroid-Stimulating/metabolism , Brain/metabolism
2.
Thyroid ; 32(4): 429-439, 2022 04.
Article in English | MEDLINE | ID: mdl-34927457

ABSTRACT

Background: Graves' eye disease, also called Graves' orbitopathy (GO), is a potentially debilitating autoimmune disease associated with retro-orbital inflammation and tissue expansion, involving both fibroblasts and adipocytes, resulting in periorbital edema, worsening proptosis, and muscle dysfunction with diplopia and may ultimately threaten sight. Accumulating evidence has indicated that autoantibodies to the thyrotropin receptor (TSHR), which induce the hyperthyroidism of Graves' disease, also help mediate the pathogenesis of the eye disease in susceptible individuals through TSHR expression on retro-orbital cells. Since it has long been known that the effects of insulin-like growth factor 1 (IGF-1) and thyrotropin are additive, recent clinical trials with a human monoclonal IGF-1 receptor blocking antibody (teprotumumab; IGF-1R-B-monoclonal antibody [mAb]) have demonstrated its ability to induce significant reductions in proptosis, diplopia, and clinical activity scores in patients with GO. However, the molecular mechanisms by which such an antibody achieves this result is unclear. Methods: We have used Li-Cor In-Cell Western, Western blot, and immunohistochemistry to define levels of different proteins in mouse and human fibroblast cells. Proteomic array was also used to define pathway signaling molecules. Using CCK-8 and BrdU cell proliferation ELISA, we have analyzed proliferative response of these cells to different antibodies. Results: We now show that a stimulating TSHR antibody was able to induce phosphorylation of the IGF-1R and initiate both TSHR and IGF-1R signaling in mouse and human fibroblasts. IGF-1R-B-mAb (1H7) inhibited all major IGF-1R signaling cascades and also reduced TSHR signaling. This resulted in the antibody-induced suppression of autophagy as shown by inhibition of multiple autophagy-related proteins (Beclin1, LC3a, LC3b, p62, and ULK1) and the induction of cell death by apoptosis as evidenced by activation of cleaved caspase 3, FADD, and caspase 8. Furthermore, this IGF-1R-blocking mAb suppressed serum-induced perkin and pink mitophagic proteins. Conclusions: Our observations clearly indicated that stimulating TSHR antibodies were able to enhance IGF-1R activity and contribute to retro-orbital cellular proliferation and inflammation. In contrast, an IGF-1R-B-mAb was capable of suppressing IGF-1R signaling leading to retro-orbital fibroblast/adipocyte death through the cell-extrinsic pathway of apoptosis. This is likely the major mechanism involved in proptosis reduction in patients with Graves' eye disease treated by IGF-1R inhibition.


Subject(s)
Graves Disease , Graves Ophthalmopathy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Apoptosis , Diplopia , Fibroblasts , Graves Ophthalmopathy/metabolism , Humans , Immunoglobulins, Thyroid-Stimulating/metabolism , Inflammation/metabolism , Insulin-Like Growth Factor I/metabolism , Mice , Proteomics , Receptor, IGF Type 1/metabolism , Receptors, Thyrotropin , Thyrotropin/metabolism
3.
J Korean Med Sci ; 35(23): e177, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32537950

ABSTRACT

BACKGROUND: High-dose intravenous steroids are the first-line treatment for patients with moderate-to-severe and active Graves' ophthalmopathy (GO). We aimed to investigate the response rate of methylprednisolone (MPD) treatment among Korean patients with active moderate-to-severe GO and to identify predictive factors of treatment response. METHODS: This is a retrospective observational study. We included 54 active moderate-to-severe GO patients treated with 4.5 g intravenous MPD over 12 weeks between November 2011 and November 2018. Response was defined as an improvement in at least two of five indicators (clinical activity score [CAS], soft-tissue involvement, exophthalmos, diplopia, and visual acuity) at immediate and 3 months after treatment completion. We examined predictive factors for response using logistic regression analysis. RESULTS: Twenty-four (44.4%) and 22 (40.7%) patients showed response at immediate and 3 months after intravenous (IV) steroid treatment. Of the five ophthalmic parameters, all patients in the responsive group (100.0%) showed a decrease in CAS and 90.9% showed less soft tissue involvement after IV steroid treatment. Among variables, the sum of extraocular muscle width was positively (odds ratio [OR], 1.163; 95% confidence interval [CI], 0.973-1.389; P = 0.096) associated with treatment response. While, the OR of age was 0.918 (95% CI, 0.856-0.985; P = 0.017) and thyrotropin binding inhibitory immunoglobulin (TBII) was 0.921 (95% CI, 0.864-0.982; P = 0.012). CONCLUSION: In Korean active moderate-to-severe GO patients, intravenous steroid treatment is not as effective as previously reported. Parameters associated with CAS and soft-tissue involvement were found to be influenced by IV MPD treatment. Extraocular muscle enlargement, younger age and lower TBII are predictive factors for a good steroid treatment response.


Subject(s)
Glucocorticoids/therapeutic use , Graves Ophthalmopathy/drug therapy , Administration, Intravenous , Adult , Digestive System Diseases/etiology , Diplopia/pathology , Female , Glucocorticoids/adverse effects , Graves Ophthalmopathy/pathology , Humans , Immunoglobulins, Thyroid-Stimulating/metabolism , Logistic Models , Male , Middle Aged , Odds Ratio , Republic of Korea , Retrospective Studies , Severity of Illness Index , Treatment Outcome , Visual Acuity
4.
Br J Ophthalmol ; 104(2): 254-259, 2020 02.
Article in English | MEDLINE | ID: mdl-31079052

ABSTRACT

AIMS: To evaluate the natural course of upper eyelid retraction (UER) in patients with thyroid eye disease (TED) and factors affecting its course. METHODS: Retrospective non-interventional cohort study in a single tertiary institution from March 2006 to March 2015 on patients with TED with (1) unilateral or bilateral UER within 6 months from initial presentation, and (2) no prior interventions nor surgical treatment for their UER. Main outcomes and measures were mean margin reflex distance 1 (MRD1) and factors associated with UER improvement. RESULTS: There were a total of 61 patients and 81 eyes (41 unilateral and 20 bilateral UER). Mean age was 42.3±15.1 years. Mean MRD1 decreased from 6.1 mm at presentation to 4.8 mm at 12 months, and 4.4 mm at 24 months. The proportion of eyes with normalisation of lid height increased from 0% at presentation to 22.2% at 6 months, 37.0% at 12 months and 49.4% at 24 months. Mean time to normalisation of MRD1 was 18.0±12.4 months. A positive family history of TED was found to be associated with a 6.2 times lower likelihood of normalisation. Change in exophthalmometry, clinical activity score and thyroid-stimulating immunoglobulin were significantly correlated to change in MRD1 (p<0.05). There was no correlation between change in MRD1 and thyroid-stimulating hormone receptor antibodies. CONCLUSION: An improved knowledge of the natural history of UER in TED will allow us to better decide and evaluate the optimal management for such patients.


Subject(s)
Eyelid Diseases/pathology , Graves Ophthalmopathy/pathology , Adult , Disease Progression , Female , Graves Ophthalmopathy/metabolism , Humans , Immunoglobulins, Thyroid-Stimulating/metabolism , Male , Middle Aged , Retrospective Studies , Risk Factors
6.
Thyroid ; 29(7): 1012-1017, 2019 07.
Article in English | MEDLINE | ID: mdl-31062662

ABSTRACT

Background: Exosomes or small extracellular vesicles secreted from cells are nanovesicles with a diameter of 40-150 nm, which play a number of roles in both physiologic and pathologic processes. In Graves' disease (GD), autoantibodies bind to the thyrotropin receptor (TSHR) on the surface of thyroid follicular epithelial cells and stimulate thyroid growth and thyroid hormone synthesis and secretion via cyclic adenosine monophosphate (cAMP) production. The present study aimed to confirm the existence of TSHR in exosomes secreted from thyroid cells and to define the role of TSHR exosomes in GD. Methods: Exosomes were isolated by differential centrifugation from the culture medium of the human thyroid follicular epithelial cell line (NTHY-ori 3-1) and thyroid carcinoma cell lines (8305C, 8505C, and FTC-133). TSHR expression in cell lysates and exosomes was evaluated by Western blot analysis. In order to study the function of TSHR exosomes, human embryonic kidney (HEK) 293 cells stably expressing TSHR (HEK/TSHR) were established. Using exosomes isolated from both HEK and HEK/TSHR cells, the binding capacity of the M22 human monoclonal autoantibody to TSHR exosomes and their effect on M22-mediated stimulation of cAMP production in HEK/TSHR cells were evaluated. As a positive control for the functional assay, human recombinant TSHR chimera protein capable of binding to TSH was used. Results: TSHR was detected in exosomes from cancer cells as well as normal epithelial cells. An in vitro binding assay showed that alkaline phosphatase-labeled M22 bound to TSHR exosomes in a dose-dependent manner. M22 dose-dependently stimulated intracellular cAMP production in HEK/TSHR cells. The addition of exosomes from HEK/TSHR cells but not those from parental HEK cells significantly ameliorated cAMP production stimulated by treatment with M22 in HEK/TSHR cells. A decoy effect similar to TSHR exosomes was observed for human recombinant TSHR chimera. Conclusions: The results suggest that exosomes expressing TSHR may be secreted from normal and cancerous thyroid cells. In the thyroid gland of patients with GD, TSHR exosomes may exert a decoy effect by sequestering autoantibody, thereby ameliorating autoantibody-mediated activation of thyroid function.


Subject(s)
Cyclic AMP/metabolism , Exosomes/metabolism , Graves Disease/metabolism , Immunoglobulins, Thyroid-Stimulating/metabolism , Receptors, Thyrotropin/metabolism , Thyroid Epithelial Cells/metabolism , Thyroid Neoplasms/metabolism , Autoantibodies/metabolism , Cell Line , Cell Line, Tumor , HEK293 Cells , Humans , In Vitro Techniques
7.
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
8.
Clin Perinatol ; 45(1): 31-40, 2018 03.
Article in English | MEDLINE | ID: mdl-29406005

ABSTRACT

Neonatal thyrotoxicosis (hyperthyroidism) is less prevalent than congenital hypothyroidism; however, it can lead to significant morbidity and mortality if not promptly recognized and adequately treated. Most cases are transient, secondary to maternal autoimmune hyperthyroidism (Graves disease [GD]). This article summarizes recommendations for screening and management of hyperthyroidism in both the fetal and neonatal periods, with a focus on neonatal thyrotoxicosis secondary to maternal GD. Early monitoring and treatment are crucial for optimizing short-term and long-term patient outcomes.


Subject(s)
Fetal Diseases/metabolism , Graves Disease/metabolism , Hyperthyroidism/metabolism , Infant, Newborn, Diseases/metabolism , Pregnancy Complications/metabolism , Thyrotoxicosis/metabolism , Adrenergic beta-Antagonists/therapeutic use , Antithyroid Agents/therapeutic use , Female , Fetal Diseases/etiology , Graves Disease/complications , Graves Disease/drug therapy , Humans , Hyperthyroidism/drug therapy , Immunoglobulins, Thyroid-Stimulating/metabolism , Infant, Newborn , Infant, Newborn, Diseases/drug therapy , Infant, Newborn, Diseases/etiology , Maternal-Fetal Exchange , Methimazole/therapeutic use , Pregnancy , Propranolol/therapeutic use , Thyroiditis, Autoimmune/complications , Thyrotoxicosis/drug therapy , Thyrotoxicosis/etiology
9.
Article in English | MEDLINE | ID: mdl-29468987

ABSTRACT

Backgroud and Objective: Antithyroid drugs (ATDs) [methylmercaptoimidazole (MMI) and propylthiouracil (PTU) ] are used to treat hyperthyroidism in Graves' disease. The effect of ATDs and reducing agents (mercaptoethanol, dithiothreitol and cysteine) on bovine (b) TSH binding to human (h) and porcine (p) TSH receptor (R) was examined. METHODS AND RESULTS: (1) ATDs was pre-incubated with hTSHR coated tube for 1- 4 h, washed free of ATDs, and then 125I-bTSH binding to hTSHR after 1 h incubation was examined. MMI (10-40 mM) decreased 125I-bTSH binding in a dose-dependent manner and binding decreased proportionally as preincubation time increased from 1 to 4 h. PTU (10mM) slightly decreased binding, When reducing agents were pre-incubated with hTSHR for 2 h, 125I-bTSH binding similarly decreased. (2) Porcine thyroid membrane was pre-incubated with both agents for 2 h. Then, the washed or unwashed membrane was incubated with 125I-bTSH for 1 h. 125I-bTSH binding in both methods decreased. (3) When the effect of ATDs or reducing agents on the biological activity of 125I-bTSH and thyroid stimulating antibody (TSAb) was examined after gel-filtration of 125I-bTSH- and TSAb- treated with both reagents for 1 h, no inactivation was observed. (4) ATDs showed similar reducing action as reducing agents because iodine (I+) was reduced to I- by ATDs. CONCLUSION: ATDs inactivate the TSH-binding site of TSHR by reduction, although ATDs do not inactivate bTSH and TSAb activity. This suggests that TSAb would not stimulate the thyroid due to the inactivation of the TSHR when ATDs are administered to patients with Graves' disease.


Subject(s)
Antithyroid Agents/pharmacology , Methimazole/pharmacology , Propylthiouracil/pharmacology , Receptors, Thyrotropin/antagonists & inhibitors , Thyroid Gland/drug effects , Thyrotropin/antagonists & inhibitors , Animals , Antithyroid Agents/metabolism , Binding Sites , Humans , Immunoglobulins, Thyroid-Stimulating/metabolism , Methimazole/metabolism , Oxidation-Reduction , Propylthiouracil/metabolism , Protein Binding , Receptors, Thyrotropin/metabolism , Sus scrofa , Thyroid Gland/metabolism , Thyrotropin/metabolism
10.
J Clin Endocrinol Metab ; 102(3): 776-785, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27768856

ABSTRACT

Context: The sources and biological impact of 3,3',5,5' tetraiodothyroacetic acid (TA4) are uncertain. CD34+ fibrocytes express several proteins involved in the production of thyroid hormones. They infiltrate the orbit in Graves disease (GD), an autoimmune process known as thyroid-associated ophthalmopathy. It appears that the thyrotropin receptor plays an important role in the pathogenesis of thyroid-associated ophthalmopathy. Objective: To quantify levels of TA4 in healthy participants and those with GD, determine whether fibrocytes generate this thyroid hormone analogue, and determine whether TA4 influences the actions of thyroid-stimulating hormone and thyroid-stimulating immunoglobulins in orbital fibroblasts. Design/Setting/Participants: Patients with GD and healthy donors in an academic medical center clinical practice were recruited. Main Outcome Measures: Liquid chromatography-tandem mass spectrometry, autoradiography, real-time polymerase chain reaction, hyaluronan immunoassay. Results: Serum levels of TA4 are elevated in GD. TA4 levels are positively correlated with those of thyroxine and negatively correlated with serum levels of triiodothyronine. Several cell types in culture generate TA4 from ambient thyroxine, including fibrocytes, HELA cells, human Müller stem cells, and retinal pigmented epithelial cells. Propylthiouracil inhibits TA4 generation. TA4 enhances the induction by thyrotropin and thyroid-stimulating immunoglobulins of several participants in the pathogenesis of thyroid-associated ophthalmopathy, including interleukin 6, hyaluronan synthase 1, prostaglandin endoperoxide H synthase 2, and haluronan production. Conclusion: TA4 may be ubiquitously generated in many tissues and enhances the biological impact of thyrotropin and thyroid-stimulating immunoglobulins in orbital connective tissue. These findings may identify a physiologically important determinant of extrathyroidal thyroid-stimulating hormone action.


Subject(s)
Graves Disease/blood , Graves Ophthalmopathy/blood , Thyroxine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Autoradiography , Case-Control Studies , Cells, Cultured , Chromatography, Liquid , Ependymoglial Cells/metabolism , Female , Fibroblasts/metabolism , Flow Cytometry , Glucuronosyltransferase/metabolism , Graves Disease/complications , Graves Ophthalmopathy/etiology , HeLa Cells , Humans , Hyaluronan Synthases , Hyaluronic Acid/metabolism , Immunoassay , Immunoglobulins, Thyroid-Stimulating/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Orbit , Prostaglandin-Endoperoxide Synthases/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Thyrotropin/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Sex Factors , Tandem Mass Spectrometry , Thyrotropin/metabolism , Thyroxine/blood , Triiodothyronine/blood , Young Adult
11.
Clin Nucl Med ; 39(4): 326-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24566408

ABSTRACT

PURPOSE OF THE REPORT: The incidence of postpartum thyrotoxicosis (PT) in Graves disease (GD) patients treated with antithyroid drugs (ATDs) is higher than in the general population, but the incidence of PT among GD patients who had been treated with radioiodine (RI) or by subtotal thyroidectomy before their pregnancy is not well known. SUBJECTS AND METHODS: We reviewed the cases of women with GD who had become pregnant, and we selected the 188 women who had undergone RI therapy before the pregnancy and the 148 women who had undergone subtotal thyroidectomy for GD before the pregnancy as the subjects of this study. The ATD subjects were 107 women with GD who had become pregnant after being treated with ATDs alone before their pregnancy and were in remission before and throughout the pregnancy. RESULTS: The overall incidence of PT was 2.1% (4/188) in the RI group, 23.6% (35/148) in the subtotal thyroidectomy group, and 55.1% (59/107) in the ATD group. There were no cases of permanent thyrotoxicosis in the RI group. CONCLUSIONS: The incidence of PT among women with GD who have undergone RI therapy before their pregnancy was significantly low compared to thyroidectomy group and ATD group. This finding is interesting because the incidence of PT in the RI group was lower than subtotal thyroidectomy group even though thyroid volume had been greatly reduced by thyroidectomy. RI treatment is recommended in the choice of treatment for childbearing-age women as regards the risk of postpartum recurrence.


Subject(s)
Antithyroid Agents/therapeutic use , Graves Disease/therapy , Postpartum Period , Thyroidectomy , Thyrotoxicosis/epidemiology , Adult , Female , Graves Disease/complications , Humans , Immunoglobulins, Thyroid-Stimulating/metabolism , Incidence , Iodine Radioisotopes/therapeutic use , Pregnancy , Thyrotoxicosis/complications , Thyrotoxicosis/metabolism
12.
J Immunol ; 190(11): 5373-81, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23630351

ABSTRACT

Graves' disease results from thyroid-stimulating Abs (TSAbs) activating the thyrotropin receptor (TSHR). How TSAbs arise from early precursor B cells has not been established. Genetic and environmental factors may contribute to pathogenesis, including the bacterium Yersinia enterocolitica. We developed two pathogenic monoclonal TSAbs from a single experimental mouse undergoing Graves' disease, which shared the same H and L chain germline gene rearrangements and then diversified by numerous somatic hypermutations. To address the Ag specificity of the shared germline precursor of the monoclonal TSAbs, we prepared rFab germline, which showed negligible binding to TSHR, indicating importance of somatic hypermutation in acquiring TSAb activity. Using rFab chimeras, we demonstrate the dominant role of the H chain V region in TSHR recognition. The role of microbial Ags was tested with Y. enterocolitica proteins. The monoclonal TSAbs recognize 37-kDa envelope proteins, also recognized by rFab germline. MALDI-TOF identified the proteins as outer membrane porin (Omp) A and OmpC. Using recombinant OmpA, OmpC, and related OmpF, we demonstrate cross-reactivity of monoclonal TSAbs with the heterogeneous porins. Importantly, rFab germline binds recombinant OmpA, OmpC, and OmpF confirming reactivity with Y. enterocolitica. A human monoclonal TSAb, M22 with similar properties to murine TSAbs, also binds recombinant porins, showing cross-reactivity of a spontaneously arising pathogenic Ab with Y. enterocolitica. The data provide a mechanistic framework for molecular mimicry in Graves' disease, where early precursor B cells are expanded by Y. enterocolitica porins to undergo somatic hypermutation to acquire a cross-reactive pathogenic response to TSHR.


Subject(s)
Germ-Line Mutation , Graves Disease/etiology , Immunoglobulins, Thyroid-Stimulating/genetics , Immunoglobulins, Thyroid-Stimulating/immunology , Yersinia enterocolitica/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Gene Expression , Graves Disease/genetics , Graves Disease/immunology , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulins, Thyroid-Stimulating/metabolism , Protein Binding/immunology , Protein Subunits/immunology , Protein Subunits/metabolism , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/metabolism , Recombinant Proteins
13.
J Clin Endocrinol Metab ; 98(5): 2153-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23482611

ABSTRACT

CONTEXT: Graves ophthalmopathy (GO) is an autoimmune disorder characterized by increased adipogenesis and hyaluronan (HA) production by orbital fibroblasts. Circulating autoantibodies (thyroid-stimulating antibodies [TSAbs]) directed at the thyrotropin receptor (TSHR) on these cells stimulate or augment these cellular processes. A recently developed drug-like small molecule inverse agonist of TSHR, NCGC00229600, termed 1, binds to TSHR and blocks basal and stimulated signal transduction. OBJECTIVE: The purpose of this article was to determine whether 1 might inhibit HA production and relevant signaling pathways in orbital fibroblasts cultured in the presence of monoclonal TSAbs or bovine TSH (bTSH). DESIGN: Primary cultures of undifferentiated GO orbital fibroblasts (n = 13) were untreated or treated with a TSAb (M22 or MS-1) or bTSH in serum-free medium, with or without 1 or a TSHR neutral antagonist, NCGC00242595, termed 2, which does not inhibit basal signaling but does inhibit stimulated signaling. MAIN OUTCOME MEASURES: cAMP production, Akt phosphorylation (Ser473pAkt in media and immunoblotting for pAkt/total Akt), and HA production were analyzed. RESULTS: Compound 1 inhibited basal cAMP, pAkt, and HA production and that stimulated by M22 in undifferentiated orbital fibroblasts. Inhibition of HA production was dose-dependent, with a half-maximal inhibitory dose of 830 nM. This compound also inhibited MS-1- and bTSH-stimulated cAMP, pAkt, and HA production. Compound 2 did not inhibit basal HA production but did inhibit M22-stimulated HA production. CONCLUSIONS: Because cAMP, pAkt, and HA production are fibroblast functions that are activated via TSHR signaling and are important in the pathogenesis of GO, small molecule TSHR antagonists may prove to be effective in the treatment or prevention of the disease in the future.


Subject(s)
Eye/drug effects , Fibroblasts/drug effects , Graves Ophthalmopathy/drug therapy , Immunoglobulins, Thyroid-Stimulating/metabolism , Pyridines/pharmacology , Quinazolinones/pharmacology , Receptors, Thyrotropin/antagonists & inhibitors , Signal Transduction/drug effects , Adipose Tissue, White/drug effects , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Antibodies, Monoclonal/metabolism , Cell Dedifferentiation , Cells, Cultured , Cyclic AMP/metabolism , Drug Inverse Agonism , Eye/immunology , Eye/metabolism , Eye/pathology , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Graves Ophthalmopathy/immunology , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/pathology , Humans , Hyaluronic Acid/metabolism , Osmolar Concentration , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Thyrotropin/agonists , Receptors, Thyrotropin/metabolism , Thyrotropin/agonists , Thyrotropin/antagonists & inhibitors , Thyrotropin/pharmacology
14.
Endocrinology ; 154(1): 562-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23183178

ABSTRACT

Despite elucidation of the crystal structure of M22, a human thyroid-stimulating autoantibody (TSAb) bound to the TSH receptor (TSHR) leucine-rich repeat domain (LRD), the mechanism by which TSAs activate the TSHR and cause Graves' disease remains unknown. A nonstimulatory murine monoclonal antibody, 3BD10, and TSAb interact with the LRD N-terminal cysteine cluster and reciprocally distinguish between two different LRD conformational forms. To study this remarkable phenomenon, we investigated properties of 3BD10, which has a linear epitopic component. By synthetic peptide ELISA, we identified 3BD10 binding to TSHR amino acids E34, E35, and D36 within TSHR cysteine-bonded loop 2 (C31-C41), which includes R38, the most N-terminal contact residue of TSAb M22. On flow cytometry, despite not contributing to the 3BD10 and M22 epitopes, chimeric substitution (but not deletion) of TSHR cysteine-bonded loop 1 (C24-C29) eliminated 3BD10 binding to the TSHR ectodomain (ECD) expressed on the cell surface, as found previously for TSAb including M22. Furthermore, 3BD10 did not recognize all cell surface TSHR ECDs, consistent with recognition of only one conformational receptor form. Reversion to wild-type of small components of the loop 1 chimeric substitution partially restored 3BD10 binding to the TSHR-ECD but not to synthetic peptides tested by ELISA. Molecular modeling supports the concept that modification of TSHR C-bonded loop 1 influences loop 2 conformation as well as LRD residues further downstream. In conclusion, the present study with mouse monoclonal antibody 3BD10 confirms TSHR conformational heterogeneity and suggests that the N-terminal cysteine cluster may contribute to this structural variability.


Subject(s)
Antibodies, Monoclonal/metabolism , Immunoglobulins, Thyroid-Stimulating/metabolism , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/metabolism , Amino Acid Sequence , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Structure, Secondary
15.
Clin Exp Immunol ; 170(2): 115-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23039881

ABSTRACT

Hyperthyroidism, defined by overproduction of thyroid hormones, has a 2-3% prevalence in the population. The most common form of hyperthyroidism is Graves' disease. A diagnostic biomarker for Graves' disease is the presence of immunoglobulins which bind to, and stimulate, the thyroid stimulating hormone receptor (TSHR), a G-protein coupled receptor (GPCR). We hypothesized that the ectopically expressed TSHR gene in a thyroid stimulating immunoglobulin (TSI) assay could be engineered to increase the accumulation of the GPCR pathway second messenger, cyclic AMP (cAMP), the molecule measured in the assay as a marker for pathway activation. An ectopically expressing TSHR-mutant guanine nucleotide-binding protein, (GNAS) Chinese hamster ovary (CHO) cell clone was constructed using standard molecular biology techniques. After incubation of the new clone with sera containing various levels of TSI, GPCR pathway activation was then quantified by measuring cAMP accumulation in the clone. The clone, together with a NaCl-free cell assay buffer containing 5% polyethylene glycol (PEG)6000, was tested against 56 Graves' patients, 27 toxic thyroid nodule patients and 119 normal patients. Using receiver operating characteristic analysis, when comparing normal with Graves' sera, the assay yielded a sensitivity of 93%, a specificity of 99% and an efficiency of 98%. Total complex precision (within-run, across runs and across days), presented as a percentage coefficient of variation, was found to be 7·8, 8·7 and 7·6% for low, medium and high TSI responding serum, respectively. We conclude that the performance of the new TSI assay provides sensitive detection of TSI, allowing for accurate, early detection of Graves' disease.


Subject(s)
Biological Assay/methods , GTP-Binding Proteins/chemistry , Immunoglobulins, Thyroid-Stimulating/blood , Immunoglobulins, Thyroid-Stimulating/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, Thyrotropin/chemistry , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cyclic AMP/genetics , Cyclic AMP/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Graves Disease/blood , Graves Disease/genetics , Graves Disease/metabolism , Humans , Hyperthyroidism/blood , Hyperthyroidism/genetics , Hyperthyroidism/metabolism , Immunoglobulins, Thyroid-Stimulating/metabolism , Polyethylene Glycols/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Sensitivity and Specificity , Thyroid Gland/metabolism
16.
Endocr J ; 59(1): 73-9, 2012.
Article in English | MEDLINE | ID: mdl-22082835

ABSTRACT

Previously we reported neutralization and partial purification of TSAb and TBAb activity using heterophilic antibody (Ab) to animal IgG from Graves' disease. Thus, we examined immunological similarity of TSAb and TBAb with animal IgG using experimentally generated anti-animal IgG [dog (d), bovine (b), porcine (p) and rabbit (rb)] Abs. TBII activity of TSAb- and TBAb-positive serum was neutralized by these anti-animal IgG Abs. Applied TSAb- or TBAb-IgG protein (purified by Protein A) on these anti-animal IgG Abs-bound column was found mainly in the unbound fraction (UF) (>65%) and partially in the bound fraction(BF) (<35%). The TBII and TSAb activity of TSAb-IgG in the BF showed significantly higher than the UF. Thus, the ratio of TBII activity (U/L)/mg protein in the BF/UF was high. TBII activity of TBAb-IgG was similarly purified by this column. We examined immunological characteristics of TSAb-and TBAb-Fab or F(ab')2 using rabbit anti-bF(ab')2 Ab. TBII and TSAb activity of TSAb-Fab or- F(ab')2 and TBII activity of TBAb-Fab or -F(ab')2 were neutralized by anti-bF(ab')2 Ab. Partial purification of TSAb- or TBAb-Fab and -F(ab')2 by anti-bF(ab')2 Ab-bound column was also possible. Immunological similarity of TSAb- and TBAb-IgG with animal IgG such as d, b, p, rb by anti-animal IgG Ab, and TSAb- or TBAb-Fab and -F(ab')2 with bFab by anti-bF(ab')2 Ab were demonstrated. These fact suggest that both Fab and Fc portion of TSAb- and TBAb-IgG molecule have immunological similarity with animal IgG.


Subject(s)
Antibody Specificity , Graves Disease/immunology , Immunoglobulins, Thyroid-Stimulating/metabolism , Animals , Antibodies, Anti-Idiotypic/metabolism , Antibodies, Immobilized/metabolism , Antibodies, Neutralizing/metabolism , Cross Reactions , Graves Disease/blood , Humans , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fc Fragments/isolation & purification , Immunoglobulin Fc Fragments/metabolism , Immunoglobulins, Thyroid-Stimulating/isolation & purification , Species Specificity
17.
Endocr J ; 59(1): 65-72, 2012.
Article in English | MEDLINE | ID: mdl-22082836

ABSTRACT

There are several reports that sera from Graves' patients contain heterophilic antibody (Ab) to animal IgG such as human anti-mouse antibody (HAMA). We examined the binding of TSAb and TBAb with heterophilic Ab. The binding of animal IgG with patient's IgG was examined by the inhibition of animal IgG on the binding of labeled bovine (b) IgG with patient's IgG. The binding to labeled bIgG was detected in the serum of 5 patients (2.7 %) among 185 patients with Graves' disease. The binding of the labeled bIgG with patient's IgG was inhibited by animal serum or the crude IgG (45% ammonium sulfate fraction of serum)(such as dog, horse, bovine, porcine, goat, ovine, rabbit, guinea-pig, rat, mouse) except human, monkey and chick. This heterophilic Ab which had cross-reaction with mammalian IgG (except human, monkey) was used as human anti-animal IgG Ab. TBII and TSAb activity of TSAb-positive serum, and TBII activity of TBAb-positive serum were neutralized by incubation with this Ab-bound column. Partial purification of TSAb- or TBAb- IgG from Protein A-purified TSAb- or TBAb-IgG was possible using this Ab-bound column. TBII and TSAb activity of TSAb-IgG and TBII activity of TBAb-IgG were neutralized by incubation with rabbit anti-human (h) IgG Ab (having cross-reaction with animal IgG). Further purification of Protein A-purified TSAb-IgG or TBAb-IgG by rabbit anti-hIgG Ab-bound column was impossible. The binding of TSAb and TBAb with heterophlic Ab means that TSAb-and TBAb-specific IgG have immunological similarity with mammalian species IgG compared to human IgG.


Subject(s)
Antibodies, Bispecific/metabolism , Antibodies, Neutralizing/metabolism , Antibody Specificity , Graves Disease/immunology , Immunoglobulins, Thyroid-Stimulating/metabolism , Adult , Animals , Antibodies, Anti-Idiotypic/metabolism , Antibodies, Immobilized/metabolism , Autoantigens/metabolism , Cross Reactions , Female , Graves Disease/blood , Humans , Immunoglobulin G/metabolism , Immunoglobulins, Thyroid-Stimulating/isolation & purification , Male , Membrane Transport Proteins/metabolism , Species Specificity
18.
Thyroid ; 21(12): 1359-66, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22066476

ABSTRACT

BACKGROUND: Graves' disease, caused by stimulatory thyrotropin receptor (TSHR) autoantibodies, has not been observed in animals. In contrast, Hashimoto's thyroiditis develops in chickens, rats, mice, dogs, and marmosets. Attempts to induce an immune response in mice to the luteinizing-hormone receptor suggested that autoantigen glycosylation was one parameter involved in breaking self-tolerance. Over evolution, TSHR glycosylation increased from three asparagine-linked-glycans (N-glycans) in fish to six N-glycans in humans and great apes. All other placental mammals lack one N-glycan in the shed TSHR A-subunit, the primary Graves' disease autoantigen. We hypothesized that (a) lesser TSHR A-subunit glycosylation reduces immunogenicity, accounting for the absence of Graves' disease in most placental mammals; (b) due to human-like A-subunit glycosylation, Graves' disease might arise in great apes. Here, we review and analyze the literature on this subject and report the results of a survey of veterinarians at primate centers and zoos in North America. SUMMARY: Previous experimental data from induced TSHR antibodies in mice support a role for A-subunit glycosylation in breaking self-tolerance. An extensive search of the great-ape literature revealed five reports of noncongenital thyroid dysfunction, four with hypothyroidism and one with hyperthyroidism. The latter was a gorilla who was treated with anti-thyroid drugs but is now deceased. Neither serum nor thyroid tissue from this gorilla were available for analysis. The survey of veterinarians revealed that none of the 979 chimpanzees in primate research centers had a diagnosis of noncongenital thyroid dysfunction and among ∼1100 great apes (gorillas, orangutans, and chimpanzees) in U.S. zoos, only three were hypothyroid, and none were hyperthyroid. CONCLUSIONS: Graves' disease appears to be either very rare or does not occur in great apes based on the literature and a survey of veterinarians. Although the available data do not advance our hypothesis, there is a paucity of information regarding thyroid function tests and thyroid autoantibodies in the great apes In addition, these primates may be protected against TSHR autoimmunity by the absence of genetic polymorphisms and putative environmental triggers. Finally, larger numbers of great apes need to be followed, and tests of thyroid function and thyroid autoantibodies be performed, to confirm that spontaneous Graves' disease is restricted to humans.


Subject(s)
Ape Diseases/immunology , Graves Disease/veterinary , Hominidae/immunology , Immunoglobulins, Thyroid-Stimulating/metabolism , Receptors, Thyrotropin/immunology , Thyroid Gland/immunology , Amino Acid Sequence , Animals , Ape Diseases/diagnosis , Ape Diseases/physiopathology , Autoimmunity , Biological Evolution , Glycosylation , Graves Disease/diagnosis , Graves Disease/immunology , Graves Disease/physiopathology , Humans , Molecular Sequence Data , Protein Processing, Post-Translational , Receptors, Thyrotropin/chemistry , Self Tolerance , Species Specificity , Thyroid Function Tests , Thyroid Gland/physiopathology
19.
Thyroid ; 21(9): 1013-20, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21834684

ABSTRACT

BACKGROUND: Thyroid-stimulating autoantibodies (TSAb) bind to the thyrotropin receptor (TSHR) extracellular domain, or ectodomain (ECD), comprising a leucine-rich repeat domain (LRD) linked by a hinge region to the transmembrane domain (TMD). The LRD (residues 22-260; signal peptide 1-21) contains two disulfide-bonded loops at its N-terminus. In the crystal structure of the isolated LRD complexed with human TSAb monoclonal antibody (mAb) M22, N-terminal disulfide loop 1 (residues 22-30) could not be determined because of crystal disorder. Nevertheless, present crystal structure data are interpreted to exclude a role for the LRD N-terminal disulfide loops in the TSAb epitope(s), contradicting prior functional evidence of a role for these loops in TSAb function. MATERIALS AND METHODS: To re-examine this issue we studied two cell types expressing the TSHR with the extreme N-terminal loop 1 (residues 22-30) deleted: the TSHR ECD lacking the TMD and tethered to the plasma membrane by a glycosyl-phosphatidylinositol (GPI) anchor, and the TSH holoreceptor containing the TMD. Because TSAb including M22 "see" the holoreceptor poorly relative to the TSHR ECD-GPI, we used the latter to examine the effect of deleting residues 22-30 on M22 binding by flow cytometry and the holoreceptor to test the effect of this deletion on the functional response to M22. RESULTS: Deletion of TSHR N-terminal loop 1 (residues 22-30) reduced the number of TSHR-ECD-GPI recognized by M22 relative to two TSHR mAb with epitopes far downstream of the LRD N-terminal loops. Relative to control mAb 2C11, M22 recognized only 60.4% of cell surface receptors (p = 0.02). In contrast to M22 binding to TSHR-ECD-GPI, in functional studies with the TSH holoreceptor, M22 stimulation of cAMP generation was unaltered by the loop 1 deletion. CONCLUSIONS: Our data support the concept that TSAb interact with the cysteine-rich N-terminus of the TSHR. Comparison of crystal structures of the same TSHR LRD in complex with TSAb M22 or blocking antibody K1-70 helps reconcile contradictory viewpoints. A difference between M22 interaction with the identical TSHR N-terminus expressed on the TSHR-ECD-GPI and holoreceptor suggests that crystallization of the TSHR LRD-M22 complex may not provide a complete understanding of the functional TSAb epitope(s) in Graves' disease.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitopes , Immunoglobulins, Thyroid-Stimulating/chemistry , Mutagenesis, Site-Directed , Receptors, Thyrotropin/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , CHO Cells , Cricetinae , Cricetulus , Crystallography , Epitope Mapping , Flow Cytometry , Immunoglobulins, Thyroid-Stimulating/metabolism , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Structure-Activity Relationship , Transfection
20.
J Neurol Sci ; 302(1-2): 114-7, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21196016

ABSTRACT

Expression of thyroid-stimulating hormone receptor (TSH-R) has been demonstrated in adipocytes, lymphocytes, bone, kidney, heart, intestine and rat brain. Immuno-reactive TSH-R has been localised in rat brain and human embryonic cerebral cortex but not in adult human brain. We designed a pilot study to determine whether anti-thyroid auto-antibodies immuno-localise in normal adult human cerebral cortex. Forensic samples from the frontal, motor, sensory, occipital, cingulate and parieto-occipito-temporal association cortices were obtained from five individuals who had died of trauma. Although there were no head injuries, the prior psychiatric history of patients was unknown. The tissues were probed with commercial antibodies against both human TSH-R and human thyroglobulin (TG). Anti-TSH-R IgG immuno-localised to cell bodies and axons of large neurones in all 6 regions of all 5 brains. The intensity and percentage of neurones labelled were similar in all tissue sections. TSH-R immuno-label was also observed in vascular endothelial cells in the cingulate gyrus. Although also found in all 5 brains and all six cortical regions, TG localised exclusively in vascular smooth muscle cells and not on neurones. Although limited by the small sample size and number of brain areas examined, this is the first study describing the presence of antigenic targets for anti-TSH-R IgG on human cortical neurons, and anti-TG IgG in cerebral vasculature.


Subject(s)
Cerebral Cortex/immunology , Cerebral Cortex/pathology , Immunoglobulins, Thyroid-Stimulating/metabolism , Adult , Endothelial Cells/immunology , Female , Humans , Image Processing, Computer-Assisted , Immunochemistry , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulins, Thyroid-Stimulating/immunology , Immunohistochemistry , Male , Neurons/immunology , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/metabolism , Thyroglobulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...