Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Horm Metab Res ; 53(7): 435-443, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34282595

RESUMO

The aim of the study was to investigate the use of serial measurements of TSH-receptor autoantibodies (TRAb) with the newest available assay technology to predict the course of Graves' Orbitopathy (GO) during the first 24 months from disease onset. Serial serum samples from patients with GO (103 mild/135 severe) were collected between 2007 and 2017 and retrospectively analyzed. The course of GO were classified into mild/severe 12 months after manifestation (severe: NOSPECS≥5; mild<5). TRAb were measured with automated binding immunoassays (IU/l): TRAb Elecsys (Cobas, Roche), TRAb bridge assay (IMMULITE, Siemens), and a cell-based bioassay (percent of specimen to reference ratio - SRR%) (Thyretain, Quidel). Variable cut off levels of measured TRAb were calculated at specificity of 90% from receiver operator curve (ROC) analysis for several timepoints during the course of GO. To select one: 5-8 months after first GO symptoms, which is the timepoint for usual referals for treatment mild course could be predicted at cut offs of 1.5 IU/l (Elecsys), 0.8 IU/l (Immulite) and 402% SRR (Thyretain) and the risc of severe course has to be anticipated if TRAb are above 11.6 IU/l (Elecsys), 6.5 IU/l (Thyretain), and 714% SRR (Thyretain). The Thyretain bioassay showed the highest diagnostic sensitivity (using the commercial cut off's) over the entire follow up period. TRAb measurements during the 24-month follow up of GO provide added value to the GO clinical activity and severity scores and should be used especially in the event of an unclear decision-taking situation with regard to therapy.


Assuntos
Autoanticorpos/sangue , Biomarcadores/sangue , Oftalmopatia de Graves/patologia , Imunoensaio/métodos , Receptores da Tireotropina/imunologia , Adulto , Idoso , Autoanticorpos/imunologia , Feminino , Seguimentos , Oftalmopatia de Graves/sangue , Oftalmopatia de Graves/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Adulto Jovem
2.
Horm Metab Res ; 53(4): 235-244, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33618407

RESUMO

The aim of this study was to investigate the potential of the new TSH-receptor antibody (TRAb) assays to predict remission or relapse of hyperthyroidism in patients with Graves' disease (GD) and Graves' orbitopathy (GO). TRAbs were measured retrospectively in sera from a cohort of GD patients with GO (n=117; remission n=38 and relapse n=79-Essen GO biobank) with automated binding immunoassays: TRAb Elecsys (Cobas Roche) and TRAb bridge assay (IMMULITE, Siemens), and the TSAb (thyroid stimulating Ab) cell-based bioassay (Thyretain, Quidel Corp.). To identify relapse risk/remission of hyperthyroidism patients were followed up at least 10 months after the end of antithyroid drug therapy (ATD) therapy. ROC plot analysis was performed to calculate cut-off levels of TRAb and TSAb for prediction of relapse and remission of hyperthyroidism. Cut-off serum levels are provided for timepoints around 3, 6, 10, and 15 months after the beginning of ATD. Repeated measurements of TRAb increase the rate of relapses predictions to 60% (Elecsys), 70% (IMMULITE), and 55% (Thyretain). Patients with remission have consistently TRAb levels below the cut off for relapse in repeated measurements. The cell-based bioassay was the most sensitive - and continued to be positive during follow up [at 15 months: 90% vs. 70% (IMMULITE) and 65% (Elecsys)]. Identification of relapsing hyperthyroidism is possible with automated immunoassays and cell-based bioassay especially with serial TRAb measurements during the course of ATD therapy. Patient who need eye surgery may profit from an early decision towards definitive treatment.


Assuntos
Antitireóideos/uso terapêutico , Autoanticorpos/sangue , Oftalmopatia de Graves/tratamento farmacológico , Receptores da Tireotropina/imunologia , Adulto , Idoso , Oftalmopatia de Graves/sangue , Oftalmopatia de Graves/imunologia , Oftalmopatia de Graves/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Receptores da Tireotropina/genética , Recidiva , Estudos Retrospectivos , Adulto Jovem
3.
Horm Metab Res ; 51(12): 765-769, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31826271

RESUMO

Thyroid peroxidase (TPO) is an enzyme that participates in thyroid hormone biosynthesis. TPO is also a major autoantigen in autoimmune thyroid diseases (AITD). In this review, we summarize the latest developments in the field of TPO research. We present the current understanding of immunodominant serologic determinants, frequency of TPO-specific autoantibodies in the population, as well as genetic and environmental factors contributing to their development. Moreover, we report recent progress in the clinical utilities of TPO autoantibody testing, including thyroid dysfunctions and extra-thyroidal disorders.


Assuntos
Iodeto Peroxidase/imunologia , Doenças da Glândula Tireoide/imunologia , Animais , Autoanticorpos/imunologia , Autoantígenos/genética , Autoantígenos/imunologia , Humanos , Iodeto Peroxidase/genética , Doenças da Glândula Tireoide/genética
4.
J Biol Chem ; 292(37): 15434-15444, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28743746

RESUMO

The thyroid gland secretes primarily tetraiodothyronine (T4), and some triiodothyronine (T3). Under normal physiological circumstances, only one-fifth of circulating T3 is directly released by the thyroid, but in states of hyperactivation of thyroid-stimulating hormone receptors (TSHRs), patients develop a syndrome of relative T3 toxicosis. Thyroidal T4 production results from iodination of thyroglobulin (TG) at residues Tyr5 and Tyr130, whereas thyroidal T3 production may originate in several different ways. In this study, the data demonstrate that within the carboxyl-terminal portion of mouse TG, T3 is formed de novo independently of deiodination from T4 We found that upon iodination in vitro, de novo T3 formation in TG was decreased in mice lacking TSHRs. Conversely, de novo T3 that can be formed upon iodination of TG secreted from PCCL3 (rat thyrocyte) cells was augmented from cells previously exposed to increased TSH, a TSHR agonist, a cAMP analog, or a TSHR-stimulating antibody. We present data suggesting that TSH-stimulated TG phosphorylation contributes to enhanced de novo T3 formation. These effects were reversed within a few days after removal of the hyperstimulating conditions. Indeed, direct exposure of PCCL3 cells to human serum from two patients with Graves' disease, but not control sera, led to secretion of TG with an increased intrinsic ability to form T3 upon in vitro iodination. Furthermore, TG secreted from human thyrocyte cultures hyperstimulated with TSH also showed an increased intrinsic ability to form T3 Our data support the hypothesis that TG processing in the secretory pathway of TSHR-hyperstimulated thyrocytes alters the structure of the iodination substrate in a way that enhances de novo T3 formation, contributing to the relative T3 toxicosis of Graves' disease.


Assuntos
Processamento de Proteína Pós-Traducional , Receptores da Tireotropina/agonistas , Transdução de Sinais , Tireoglobulina/metabolismo , Células Epiteliais da Tireoide/metabolismo , Tireotropina/metabolismo , Tri-Iodotironina/biossíntese , Animais , Proteínas de Ligação ao Cálcio/agonistas , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Caseína Quinase I/genética , Caseína Quinase I/metabolismo , Linhagem Celular , Células Cultivadas , Proteínas da Matriz Extracelular/agonistas , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Doença de Graves/sangue , Doença de Graves/metabolismo , Doença de Graves/patologia , Halogenação , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Receptores da Tireotropina/genética , Receptores da Tireotropina/metabolismo , Células Epiteliais da Tireoide/citologia , Células Epiteliais da Tireoide/patologia , Tirosina/metabolismo , Regulação para Cima
5.
Magn Reson Med ; 80(2): 711-718, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29377287

RESUMO

PURPOSE: To evaluate key molecular and cellular features of Graves orbitopathy (GO) by simultaneous monitoring of alterations in morphology, inflammatory patterns, and tissue remodeling. METHODS: To this end, we utilized a murine model of GO induced by immunization with a human thyroid-stimulating hormone receptor A-subunit plasmid. Altogether, 52 mice were used: 27 GOs and 25 controls (Ctrl) immunized with ß-galactasidose plasmid. From these, 17 GO and 12 Ctrl mice were subjected to multimodal MRI at 9.4T, whereas 23 mice only underwent histology. Beyond anatomical hydrogen-1 (1 H) MRI, we employed transverse relaxation time (T2 ) mapping for visualization of edema, chemical exchange saturation transfer (CEST) for detection of hyaluronan, and fluorine-19 (19 F) MRI for tracking of in situ-labeled immune cells after intravenous injection of perfluorcarbons (PFCs). RESULTS: 1 H/19 F MRI demonstrated substantial infiltration of PFC-loaded immune cells in peri and retro-orbital regions of GO mice, whereas healthy Ctrls showed only minor 19 F signals. In parallel, T2 mapping indicated onset of edema in periorbital tissue and adjacent ocular glands (P = 0.038/0.017), which were associated with enhanced orbital CEST signals in GO mice (P = 0.031). Concomitantly, a moderate expansion of retrobulbar fat (P = 0.029) was apparent; however, no signs for extraocular myopathy were detectable. 19 F MRI-based visualization of orbital inflammation exhibited the highest significance level to discriminate between GO and Ctrl mice (P = 0.006) and showed the best correlation with the clinical score (P = 0.0007). CONCLUSION: The present approach permits the comprehensive characterization of orbital tissue and holds the potential for accurate GO diagnosis in the clinical setting. Magn Reson Med 80:711-718, 2018. © 2018 International Society for Magnetic Resonance in Medicine.


Assuntos
Olho , Oftalmopatia de Graves , Inflamação , Imageamento por Ressonância Magnética/métodos , Animais , Modelos Animais de Doenças , Edema/diagnóstico por imagem , Edema/imunologia , Olho/diagnóstico por imagem , Olho/imunologia , Oftalmopatia de Graves/diagnóstico por imagem , Oftalmopatia de Graves/imunologia , Processamento de Imagem Assistida por Computador , Inflamação/diagnóstico por imagem , Inflamação/imunologia , Camundongos , Receptores da Tireotropina/genética , Receptores da Tireotropina/imunologia
6.
Horm Metab Res ; 50(12): 932-941, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30107619

RESUMO

Experimental models of hyperthyroid Graves' disease (GD) and Graves' orbitopathy (GO) are efficiently developed by genetic immunisation by electroporation with human thyrotropin hormone receptor (hTSHR) A-subunit plasmid in female BALB/c (H-2d) mice. We investigated susceptibility in C57BL/6 J (H-2b) animals to allow studies on disease mechanisms in transgenic and immune response gene knock-out mice. Higher numbers of female C57BL/6 J were positive for pathogenic thyroid stimulating antibodies, but induced hyperthyroidism remained at a low frequency compared to BALB/c animals. Assessment of hTSHR specific T cells showed reduced proliferation in C57BL/6 J animals accompanied with anti-inflammatory IL-10, with less pro-inflammatory IFN-γ compared to BALB/c. Whilst the orbital tissue from immune BALB/c mice showed inflammation and adipogenesis, in contrast C57BL/6 J animals showed normal pathology. We characterised the gut microbiota using 16 S ribosomal RNA gene sequencing to explore its possible pathogenic role in the model. Despite being housed under identical conditions, we observed significantly different organisation of the microbiota (beta-diversity) in the two strains. Taxonomic differences were also noted, with C57BL/6 J showing an enrichment of Operational Taxonomic Units (OTUs) belonging to the Paludibacter and Allobaculum, followed by Limibacter, Anaerophaga and Ureaplasma genera. A higher number of genera significantly correlating with clinical features was observed in C57BL/6 J compared to BALB/c; for example, Limibacter OTUs correlated negatively with thyroid-stimulating antibodies in C57BL/6 J mice. Thus, our data suggest gut microbiota may play a pivotal immunomodulatory role that differentiates the thyroid function and orbital pathology outcome in these two inbred strains undergoing experimental GO.


Assuntos
Autoimunidade , Microbioma Gastrointestinal , Glândula Tireoide/imunologia , Glândula Tireoide/fisiopatologia , Animais , Proliferação de Células , Citocinas/metabolismo , Feminino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Órbita/patologia , Receptores da Tireotropina/metabolismo , Linfócitos T/metabolismo
7.
J Immunol ; 190(11): 5373-81, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23630351

RESUMO

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.


Assuntos
Mutação em Linhagem Germinativa , Doença de Graves/etiologia , Imunoglobulinas Estimuladoras da Glândula Tireoide/genética , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Yersinia enterocolitica/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Expressão Gênica , Doença de Graves/genética , Doença de Graves/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/metabolismo , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Região Variável de Imunoglobulina/genética , Imunoglobulinas Estimuladoras da Glândula Tireoide/metabolismo , Ligação Proteica/imunologia , Subunidades Proteicas/imunologia , Subunidades Proteicas/metabolismo , Receptores da Tireotropina/química , Receptores da Tireotropina/imunologia , Receptores da Tireotropina/metabolismo , Proteínas Recombinantes
9.
Front Endocrinol (Lausanne) ; 14: 1211473, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37435490

RESUMO

Introduction: Graves' disease (GD) is an autoimmune disorder caused by autoantibodies against the thyroid stimulating hormone receptor (TSHR) leading to overstimulation of the thyroid gland. Thyroid eye disease (TED) is the most common extra thyroidal manifestation of GD. Therapeutic options to treat TED are very limited and novel treatments need to be developed. In the present study we investigated the effect of linsitinib, a dual small-molecule kinase inhibitor of the insulin-like growth factor 1 receptor (IGF-1R) and the Insulin receptor (IR) on the disease outcome of GD and TED. Methods: Linsitinib was administered orally for four weeks with therapy initiating in either the early ("active") or the late ("chronic") phases of the disease. In the thyroid and the orbit, autoimmune hyperthyroidism and orbitopathy were analyzed serologically (total anti-TSHR binding antibodies, stimulating anti TSHR antibodies, total T4 levels), immunohistochemically (H&E-, CD3-, TNFa- and Sirius red staining) and with immunofluorescence (F4/80 staining). An MRI was performed to quantify in vivo tissue remodeling inside the orbit. Results: Linsitinib prevented autoimmune hyperthyroidism in the early state of the disease, by reducing morphological changes indicative for hyperthyroidism and blocking T-cell infiltration, visualized by CD3 staining. In the late state of the disease linsitinib had its main effect in the orbit. Linsitinib reduced immune infiltration of T-cells (CD3 staining) and macrophages (F4/80 and TNFa staining) in the orbita in experimental GD suggesting an additional, direct effect of linsitinib on the autoimmune response. In addition, treatment with linsitinib normalized the amount of brown adipose tissue in both the early and late group. An in vivo MRI of the late group was performed and revealed a marked decrease of inflammation, visualized by 19F MR imaging, significant reduction of existing muscle edema and formation of brown adipose tissue. Conclusion: Here, we demonstrate that linsitinib effectively prevents development and progression of thyroid eye disease in an experimental murine model for Graves' disease. Linsitinib improved the total disease outcome, indicating the clinical significance of the findings and providing a path to therapeutic intervention of Graves' Disease. Our data support the use of linsitinib as a novel treatment for thyroid eye disease.


Assuntos
Doença de Graves , Oftalmopatia de Graves , Inibidores de Proteínas Quinases , Receptor IGF Tipo 1 , Animais , Camundongos , Doença de Graves/tratamento farmacológico , Oftalmopatia de Graves/tratamento farmacológico , Hipertireoidismo , Imidazóis , Inibidores de Proteínas Quinases/uso terapêutico , Receptor IGF Tipo 1/antagonistas & inibidores
10.
Nat Struct Mol Biol ; 14(4): 280-6, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17384644

RESUMO

Gamma-aminobutyric acid (GABA) is synthesized by two isoforms of the pyridoxal 5'-phosphate-dependent enzyme glutamic acid decarboxylase (GAD65 and GAD67). GAD67 is constitutively active and is responsible for basal GABA production. In contrast, GAD65, an autoantigen in type I diabetes, is transiently activated in response to the demand for extra GABA in neurotransmission, and cycles between an active holo form and an inactive apo form. We have determined the crystal structures of N-terminal truncations of both GAD isoforms. The structure of GAD67 shows a tethered loop covering the active site, providing a catalytic environment that sustains GABA production. In contrast, the same catalytic loop is inherently mobile in GAD65. Kinetic studies suggest that mobility in the catalytic loop promotes a side reaction that results in cofactor release and GAD65 autoinactivation. These data reveal the molecular basis for regulation of GABA homeostasis.


Assuntos
Glutamato Descarboxilase/metabolismo , Isoenzimas/metabolismo , Ácido gama-Aminobutírico/biossíntese , Sequência de Aminoácidos , Autoantígenos/imunologia , Sítios de Ligação/efeitos dos fármacos , Catálise/efeitos dos fármacos , Cristalografia por Raios X , Dimerização , Ativação Enzimática/efeitos dos fármacos , Glutamato Descarboxilase/química , Glutamato Descarboxilase/imunologia , Ácido Glutâmico/farmacologia , Humanos , Isoenzimas/química , Isoenzimas/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína/efeitos dos fármacos
11.
Proc Natl Acad Sci U S A ; 105(14): 5471-6, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18367670

RESUMO

Autoantibodies to glutamate decarboxylase 65 (GAD65Ab) are commonly believed to be a major characteristic for type 1 diabetes (T1D). We investigated the presence of GAD65Ab in healthy individuals (n = 238) and first-degree relatives (FDRs) of T1D patients (n = 27) who tested negative for GAD65Ab in conventional RIAs. Sera were applied to affinity columns coated with GAD65-specific mAbs to absorb anti-idiotypic antibodies (anti-Ids). The absorbed sera were analyzed for binding to GAD65 by RIAs. Both healthy individuals and FDRs present GAD65Ab that are inhibited by anti-Id, masking them in conventional detection methods. The presence of GAD65Ab-specific anti-Ids was confirmed by competitive ELISA. Remarkably, T1D patients (n = 54) and Stiff Person Syndrome patients (n = 8) show a specific lack of anti-Ids to disease-associated GAD65Ab epitopes. Purified anti-Ids from healthy individuals and FDRs inhibited the binding of GAD65Ab from T1D patients to GAD65. We conclude that masked GAD65Ab are present in the healthy population and that a lack of particular anti-Ids, rather than GAD65Ab per se, is a characteristic of T1D. The lack of these inhibitory antibodies may contribute to T cell activation by GAD65Ab.


Assuntos
Anticorpos Anti-Idiotípicos/sangue , Autoanticorpos/sangue , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/imunologia , Glutamato Descarboxilase/imunologia , Estudos de Casos e Controles , Saúde da Família , Humanos , Radioimunoensaio , Rigidez Muscular Espasmódica
12.
Microbiome ; 9(1): 45, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33593429

RESUMO

BACKGROUND: Graves' disease (GD) is an autoimmune condition in which autoantibodies to the thyrotropin receptor (TSHR) cause hyperthyroidism. About 50% of GD patients also have Graves' orbitopathy (GO), an intractable disease in which expansion of the orbital contents causes diplopia, proptosis and even blindness. Murine models of GD/GO, developed in different centres, demonstrated significant variation in gut microbiota composition which correlated with TSHR-induced disease heterogeneity. To investigate whether correlation indicates causation, we modified the gut microbiota to determine whether it has a role in thyroid autoimmunity. Female BALB/c mice were treated with either vancomycin, probiotic bacteria, human fecal material transfer (hFMT) from patients with severe GO or ddH2O from birth to immunization with TSHR-A subunit or beta-galactosidase (ßgal; age ~ 6 weeks). Incidence and severity of GD (TSHR autoantibodies, thyroid histology, thyroxine level) and GO (orbital fat and muscle histology), lymphocyte phenotype, cytokine profile and gut microbiota were analysed at sacrifice (~ 22 weeks). RESULTS: In ddH2O-TSHR mice, 84% had pathological autoantibodies, 67% elevated thyroxine, 77% hyperplastic thyroids and 70% orbital pathology. Firmicutes were increased, and Bacteroidetes reduced relative to ddH2O-ßgal; CCL5 was increased. The random forest algorithm at the genus level predicted vancomycin treatment with 100% accuracy but 74% and 70% for hFMT and probiotic, respectively. Vancomycin significantly reduced gut microbiota richness and diversity compared with all other groups; the incidence and severity of both GD and GO also decreased; reduced orbital pathology correlated positively with Akkermansia spp. whilst IL-4 levels increased. Mice receiving hFMT initially inherited their GO donors' microbiota, and the severity of induced GD increased, as did the orbital brown adipose tissue volume in TSHR mice. Furthermore, genus Bacteroides, which is reduced in GD patients, was significantly increased by vancomycin but reduced in hFMT-treated mice. Probiotic treatment significantly increased CD25+ Treg cells in orbital draining lymph nodes but exacerbated induced autoimmune hyperthyroidism and GO. CONCLUSIONS: These results strongly support a role for the gut microbiota in TSHR-induced disease. Whilst changes to the gut microbiota have a profound effect on quantifiable GD endocrine and immune factors, the impact on GO cellular changes is more nuanced. The findings have translational potential for novel, improved treatments. Video abstract.


Assuntos
Microbioma Gastrointestinal , Oftalmopatia de Graves/microbiologia , Animais , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Feminino , Oftalmopatia de Graves/imunologia , Oftalmopatia de Graves/metabolismo , Oftalmopatia de Graves/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C
13.
Immunology ; 129(2): 300-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19845794

RESUMO

Graves' disease is characterized by production of agonist antibodies to the thyroid-stimulating hormone receptor (TSHR), but knowledge of the genetic and somatic events leading to their aberrant production is limited. We describe the genetic analysis of two monoclonal antibodies (mAbs) with thyroid-stimulating activity (TSAb) obtained from a single mouse with experimental Graves' disease. The mAbs were class switched, but used the same rearrangement of immunoglobulin heavy chain, variable region (IGHV) and immunoglobulin light chain, variable region (IGLV) germline genes, implying a clonal relationship and derivation from a single precursor B-cell clone. The IGHV-region genes of the two mAbs underwent high degrees of somatic hypermutation by sharing numerous mutations before diverging, while the IGLV genes evolved separately. Interestingly, the mutations were present in both the complementarity-determining regions (CDRs) and the framework regions. The cloned IGHV and IGLV genes were confirmed to have TSAb properties in experiments in which they were expressed as recombinant Fabs (rFabs). In other experiments, we swapped the IGLV genes with IGHV genes by constructing chimeric rFabs and showed that the chimeras retained TSAb activities, confirming the close functional relatedness of the V-region genes. Importantly, the IGLV genes in chimeric rFabs had a dominant stimulatory effect at low concentrations, while the IGHV genes had a dominant effect at higher concentrations. Our findings demonstrate that, in experimentally immunized mice, multiple pathogenic antibodies to TSHR can arise from a single clone by a series of somatic mutations in the V-region genes and may give an insight into how such antibodies develop spontaneously in autoimmune Graves' disease.


Assuntos
Anticorpos Monoclonais , Doença de Graves/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias lambda de Imunoglobulina/genética , Imunoglobulinas Estimuladoras da Glândula Tireoide/genética , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Animais , Diversidade de Anticorpos/genética , Autoimunidade , Regiões Determinantes de Complementaridade/genética , Análise Mutacional de DNA , Embaralhamento de DNA , Engenharia Genética , Doença de Graves/genética , Hibridomas , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/metabolismo , Cadeias lambda de Imunoglobulina/imunologia , Cadeias lambda de Imunoglobulina/metabolismo , Imunoglobulinas Estimuladoras da Glândula Tireoide/metabolismo , Camundongos , Receptores da Tireotropina/imunologia , Proteínas Recombinantes de Fusão/genética , Hipermutação Somática de Imunoglobulina
14.
Thyroid ; 30(2): 270-276, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31805824

RESUMO

Background: Thyrotropin (TSH) and thyroid-stimulating antibodies (TSAbs) activate TSH receptor (TSHR) signaling by binding to its extracellular domain. TSHR signaling has been studied extensively in animal thyrocytes and in engineered cell lines, and differences in signaling have been observed in different cell systems. We, therefore, decided to characterize and compare TSHR signaling mediated by TSH and monoclonal TSAbs in human thyrocytes in primary culture. Methods: We used quantitative reverse transcription-polymerase chain reaction to measure mRNA levels of thyroid-specific genes thyroglobulin (TG), thyroperoxidase (TPO), iodothyronine deiodinase type 2 (DIO2), sodium-iodide symporter (NIS), and TSHR after stimulation by TSH or two monoclonal TSAbs, KSAb1 and M22. We also compared secreted TG protein after TSHR activation by TSH and TSAbs using an enzyme-linked immunosorbent assay. TSHR cell surface expression was determined using fluorescence activated cell sorting (FACS). Results: We found that TSH at low doses increases and at high doses (>1 mU/mL) decreases levels of gene expression for TSHR, TG, TPO, NIS, and DIO2. The biphasic effect of TSH on signaling was not caused by downregulation of cell surface TSHRs. This bell-shaped biphasic dose-response curve has been termed an inverted U-shaped dose-response curve (IUDRC). An IUDRC was also found for TSH-induced regulation of TG secretion. In contrast, KSAb1- and M22-induced regulation of TSHR, TG, TPO, NIS, and DIO2 gene expression, and secreted TG followed a monotonic dose-response curve that plateaus at high doses of activating antibody. Conclusions: Our data demonstrate that the physiological activation of TSHRs by TSH in primary cultures of human thyrocytes is characterized by a regulatory mechanism that may inhibit thyrocyte overstimulation. In contrast, TSAbs do not exhibit biphasic regulation. Although KSAb1 and M22 may not be representative of all TSAbs found in patients with Graves' disease, we suggest that persistent robust stimulation of TSHRs by TSAbs, unrelieved by a decrease at high TSAb levels, fosters chronic stimulation of thyrocytes in Graves' hyperthyroidism.


Assuntos
Expressão Gênica/efeitos dos fármacos , Imunoglobulinas Estimuladoras da Glândula Tireoide/farmacologia , Células Epiteliais da Tireoide/efeitos dos fármacos , Tireotropina/farmacologia , Autoantígenos/genética , Autoantígenos/metabolismo , Células Cultivadas , Humanos , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Receptores da Tireotropina/genética , Receptores da Tireotropina/metabolismo , Simportadores/genética , Simportadores/metabolismo , Tireoglobulina/genética , Tireoglobulina/metabolismo , Células Epiteliais da Tireoide/metabolismo , Iodotironina Desiodinase Tipo II
15.
Endocrinology ; 161(2)2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32022847

RESUMO

Thyroid peroxidase (TPO) is a critical membrane-bound enzyme involved in the biosynthesis of multiple thyroid hormones, and is a major autoantigen in autoimmune thyroid diseases such as destructive (Hashimoto) thyroiditis. Here we report the biophysical and structural characterization of a novel TPO construct containing only the ectodomain of TPO and lacking the propeptide. The construct was enzymatically active and able to bind the patient-derived TR1.9 autoantibody. Analytical ultracentrifugation data suggest that TPO can exist as both a monomer and a dimer. Combined with negative stain electron microscopy and molecular dynamics simulations, these data show that the TR1.9 autoantibody preferentially binds the TPO monomer, revealing conformational changes that bring together previously disparate residues into a continuous epitope. In addition to providing plausible structural models of a TPO-autoantibody complex, this study provides validated TPO constructs that will facilitate further characterization, and advances our understanding of the structural, functional, and antigenic characteristics of TPO, an autoantigen implicated in some of the most common autoimmune diseases.


Assuntos
Autoanticorpos/metabolismo , Iodeto Peroxidase/metabolismo , Tireoidite Autoimune/enzimologia , Dimerização , Células HEK293 , Humanos , Iodeto Peroxidase/química , Iodeto Peroxidase/isolamento & purificação , Iodeto Peroxidase/ultraestrutura , Multimerização Proteica , Estrutura Quaternária de Proteína
16.
Clin Immunol ; 130(3): 252-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18964302

RESUMO

Treatment of Graves' disease (GD) with the B-lymphocyte depleting agent rituximab in addition to standard methimazole-therapy prolongs remission. Paradoxically, it does not mediate a reduction in thyrotropin receptor antibody (TRAb) levels over that of methimazole monotherapy. Using a bioassay involving Chinese hamster ovary cells transfected with the human thyrotropin receptor, we found that the stimulatory capacity of TRAbs was reduced markedly, by 66+/-22%, upon treatment with rituximab and methimazole for 21 days (p<0.0001), compared to an increase by 33% on average (NS) in patients receiving methimazole alone (p=0.04 between groups). The overall levels of TRAbs decreased by around 15% in both groups. Within one year of follow-up, rituximab therapy mediated specific decreases in thyroid-peroxidase antibody- and IgM levels, whereas IgG levels were unaffected. The data indicate that rituximab therapy has differential effects on pathogenic and non-pathogenic autoantibodies, even when directed against the same antigen. The possible mechanisms underlying this hitherto unappreciated phenomenon are discussed.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Formação de Anticorpos/efeitos dos fármacos , Doença de Graves/tratamento farmacológico , Imunoglobulinas Estimuladoras da Glândula Tireoide/metabolismo , Fatores Imunológicos/uso terapêutico , Animais , Anticorpos Monoclonais Murinos , Autoanticorpos/sangue , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Doença de Graves/imunologia , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/efeitos dos fármacos , Rituximab
17.
Stem Cell Res Ther ; 10(1): 246, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31399042

RESUMO

BACKGROUND: Graves' ophthalmopathy (GO) is a complication of Graves' disease (GD), in which orbital connective tissues become inflamed and increase in volume and orbital fibroblasts within the orbital fat and extraocular muscles differentiate into adipocytes in vitro when stimulated by hormones, several cytokines, and growth factors including TSH, IGF-1, IL-1, interferon γ, and platelet-derived growth factor. Human placental mesenchymal stem cells (hPMSCs) have immunomodulatory effects in disease pathogenesis. Although a number of studies have reported that hPMSCs can elicit therapeutic effects, these are not sufficient. Therefore, we constructed a GO animal model in order to find out the hPMSCs recovery effect. METHODS: We investigated their anti-adipogenic effects in in vitro cultures of orbital fibroblasts established from GO patients. Primary orbital fibroblasts were exposed to differentiation medium for 10 days. After being co-cultured with hPMSCs, the characteristics of orbital fibroblast were determined by Oil Red O stain and real-time PCR. Then, we explored the in vivo regulatory effects of hPMSCs in an experimental mouse model of GO. We developed the GO mouse model using immunization by leg muscle electroporation of pTriEx1.1Neo-hTSHR A-subunit plasmid. Human PMSC injection was performed into the left orbit. We also analyzed the effects of hPMSCs in the GO animal model. RESULT: We found that hPMSCs inhibited a lipid accumulation and activated factors, such as ADIPONECTIN, PPARγ, C/EBPα, and TGFß2 genes in adipogenesis-induced primary orbital fibroblasts from GO patients. Moreover, hPMSCs were highly effective at ameliorating adipogenesis in the orbital tissue of the model. CONCLUSION: These data indicate that hPMSCs recover pathogenic activation of orbital fibroblasts in animals undergoing experimental GO and confirm the feasibility of applying hPMSCs as a novel treatment for GO patients.


Assuntos
Adipogenia , Diferenciação Celular , Oftalmopatia de Graves/patologia , Células-Tronco Mesenquimais/metabolismo , Placenta/citologia , Adiponectina/metabolismo , Animais , Células Cultivadas , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Órbita/citologia , Órbita/patologia , PPAR gama/metabolismo , Gravidez , Glândula Tireoide/patologia
18.
Endocrinology ; 160(6): 1468-1479, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31127272

RESUMO

Endogenously expressed TSH receptors (TSHRs) on orbital fibroblasts of patients with Graves ophthalmopathy (GO) use crosstalk with IGF1 receptors (IGF1R) to synergistically stimulate secretion of hyaluronan (HA), a major component of GO pathology. We previously showed crosstalk occurred upstream of mitogen-activated protein kinase (ERK) phosphorylation. Because other G protein-coupled receptors engage arrestin-ß-1 (ARRB1) and ERK, we tested whether ARRB1 was a necessary component of TSHR/IGF1R crosstalk. HA secretion was stimulated by the TSHR-stimulating monoclonal antibodies M22 and KSAb1, or immunoglobulins from patients with GO (GO-Igs). Treatment with M22, as previously shown, resulted in biphasic dose-response stimulation of HA secretion. The high-potency phase was IGF1R dependent, and the low-potency phase was partly IGF1R independent. KSAb1 produced a monophasic dose-response stimulation of HA secretion, whose potency was lowered >20-fold after IGF1R knockdown. ARRB1 knockdown abolished M22's high-potency phase and lowered KSAb1's potency and efficacy. ARRB1 knockdown inhibited GO-Ig stimulation of HA secretion and of ERK phosphorylation. Last, ARRB1 was shown to be necessary for TSHR/IGF1R proximity. In contrast, ARRB2 knockdowns did not show these effects. Thus, TSHR must neighbor IGF1R for crosstalk in GO fibroblasts to occur, and this depends on ARRB1 acting as a scaffold. Similar scaffolding of TSHR and IGF1R by ARRB1 was found in human osteoblast-like cells and human thyrocytes. These findings support a model of TSHR/IGF1R crosstalk that may be a general mechanism for G-protein-coupled receptor/receptor tyrosine kinase crosstalk dependent on ARRB1.


Assuntos
Receptor IGF Tipo 1/metabolismo , Receptores da Tireotropina/metabolismo , Células Epiteliais da Tireoide/metabolismo , beta-Arrestina 1/metabolismo , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Oftalmopatia de Graves/metabolismo , Humanos , Camundongos , Fosforilação , Receptor IGF Tipo 1/genética , Receptores da Tireotropina/genética , Transdução de Sinais/fisiologia , beta-Arrestina 1/genética
19.
Immunology ; 123(4): 547-54, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18005036

RESUMO

Type 1 diabetes is caused by the autoimmune destruction of pancreatic beta cells. Here we show that administration of a human monoclonal antibody (b96.11) specific to the 65-kDa isoform of glutamate decarboxylase (GAD65) to prediabetic non-obese diabetic (NOD) mice significantly delays the onset of autoimmune diabetes. We found this effect to be epitope-specific, as only b96.11 showed this therapeutic property, while a GAD65-specific human monoclonal control antibody (b78) derived from the same patient, but specific to a different determinant of GAD65, had no significant effect on the progression of disease. Administration of b96.11 or b78 to NOD mice was accompanied by the generation of anti-idiotypic antibodies. Importantly, the induced anti-idiotypic antibodies were specific for the immunizing antibody and blocked the binding of GAD65 by the respective antibody. These findings suggest a potential role for the internal image of the GAD65 determinant recognized by b96.11 in the anti-idiotypic antibody, supporting an immunomodulatory role for GAD65-specific autoantibodies, as originally postulated by Jerne.


Assuntos
Anticorpos Anti-Idiotípicos/sangue , Anticorpos Monoclonais/uso terapêutico , Diabetes Mellitus Tipo 1/prevenção & controle , Glutamato Descarboxilase/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Ligação Competitiva , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Epitopos/imunologia , Feminino , Imunoglobulina G/sangue , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Dados de Sequência Molecular , Alinhamento de Sequência , Índice de Gravidade de Doença , Redução de Peso
20.
Clin Endocrinol (Oxf) ; 69(4): 664-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18363888

RESUMO

OBJECTIVE: Thyroid peroxidase antibodies (TPOAb) are markers of autoimmune thyroid disease (AITD), including Hashimoto's thyroiditis (HT), but naturally occurring TPOAb are also detectable in healthy, euthyroid individuals. In AITD, circulating TPOAb react mainly with two immunodominant regions (IDR), IDR-A and IDR-B. The present study was undertaken in order to compare the epitope recognition pattern of TPOAb in HT patients and healthy subjects. DESIGN: Sera from 21 out of 98 healthy controls were selected on the basis of high TPOAb values, required for determination of TPOAb recognition pattern; as were sera from 92 HT patients. MEASUREMENTS: Measurement of IDR-reactivity was possible in 90 patients and 12 controls. IDR-A-, IDR-B- and non-IDR-A/non-IDR-B-Ab constituted 24 +/- 11%, 50 +/- 15% and 26 +/- 12%, respectively, in the patients. The distribution in the controls was distinctly different, only 12 +/- 13% being directed against IDR-A (P < 0.002) and 66 +/- 22% against IDR-B (P < 0.002). Half of the healthy individuals, vs. none of the HT patients, lacked IDR-A reactivity completely (P < 0.0001). In HT patients, IDR-B-Ab proportions increased slightly with increasing TPOAb levels (P < 0.05), while IDR-B-Ab of the controls showed a strong opposite trend (P < 0.0001). Accordingly, the proportion of non-A/non-B-Ab correlated with TPOAb levels in the healthy controls (P < 0.008), and an inverse correlation was seen in HT patients (P < 0.02). CONCLUSION: The data suggest that TPOAb do not differ only in quantity between HT patients and healthy individuals, but may also follow distinct qualitative patterns. Larger studies are required to confirm this, and to determine whether the propensity to produce antibodies to certain TPO epitopes, for example, IDR-A, is of pathogenic relevance.


Assuntos
Reações Antígeno-Anticorpo/imunologia , Autoanticorpos/imunologia , Doença de Hashimoto/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Especificidade de Anticorpos , Autoanticorpos/sangue , Estudos de Casos e Controles , Mapeamento de Epitopos , Feminino , Doença de Hashimoto/sangue , Saúde , Humanos , Iodeto Peroxidase/química , Iodeto Peroxidase/imunologia , Pessoa de Meia-Idade , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA