Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 349
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 609(7928): 854-859, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35940204

RESUMO

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.


Assuntos
Imunoglobulinas Estimuladoras da Glândula Tireoide , Receptores da Tireotropina , Tireotropina , Doença de Graves/imunologia , Doença de Graves/metabolismo , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Microdomínios da Membrana , Receptores do LH , Receptores da Tireotropina/agonistas , Receptores da Tireotropina/química , Receptores da Tireotropina/imunologia , Receptores da Tireotropina/metabolismo , Tireotropina/metabolismo
2.
Nature ; 609(7928): 846-853, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35940205

RESUMO

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.


Assuntos
Microscopia Crioeletrônica , Imunoglobulinas Estimuladoras da Glândula Tireoide , Receptores da Tireotropina , Tireotropina , Membrana Celular/metabolismo , Doença de Graves/imunologia , Doença de Graves/metabolismo , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/química , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Imunoglobulinas Estimuladoras da Glândula Tireoide/farmacologia , Imunoglobulinas Estimuladoras da Glândula Tireoide/ultraestrutura , Fosfolipídeos/metabolismo , Domínios Proteicos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/ultraestrutura , Receptores da Tireotropina/agonistas , Receptores da Tireotropina/química , Receptores da Tireotropina/imunologia , Receptores da Tireotropina/ultraestrutura , Rotação , Tireotropina/química , Tireotropina/metabolismo , Tireotropina/farmacologia
3.
J Immunol ; 202(9): 2570-2577, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30944161

RESUMO

Transgenic NOD.H2h4 mice that express the human (h) TSHR A-subunit in the thyroid gland spontaneously develop pathogenic TSHR autoantibodies resembling those in patients with Graves disease. Nanoparticles coupled to recombinant hTSHR A-subunit protein and a tolerogenic molecule (ligand for the endogenous aryl-hydrocarbon receptor; ITE) were injected i.p. four times at weekly intervals into hTSHR/NOD.H2h4 mice with the goal of blocking TSHR Ab development. Unexpectedly, in transgenic mice, injecting TSHR A-subunit-ITE nanoparticles (not ITE-nanoparticles or buffer) accelerated and enhanced the development of pathogenic TSHR Abs measured by inhibition of TSH binding to the TSHR. Nonpathogenic TSHR Abs (ELISA) were enhanced in transgenics and induced in wild-type littermates. Serendipitously, these findings have important implications for disease pathogenesis: development of Graves TSHR Abs is limited by the availability of A-subunit protein, which is shed from membrane bound TSHR, expressed at low levels in the thyroid. The enhanced TSHR Ab response following injected TSHR A-subunit protein-nanoparticles is reminiscent of the transient increase in pathogenic TSHR Abs following the release of thyroid autoantigens after radio-iodine therapy in Graves patients. However, in the hTSHR/NOD.H2h4 model, enhancement is specific for TSHR Abs, with Abs to thyroglobulin and thyroid peroxidase remaining unchanged. In conclusion, despite the inclusion of a tolerogenic molecule, injected nanoparticles coated with TSHR A-subunit protein enhanced and accelerated development of pathogenic TSHR Abs in hTSHR/NOD. NOD.H2h4 These findings emphasize the need for sufficient TSHR A-subunit protein to activate the immune system and the generation of stimulatory TSHR Abs in genetically predisposed individuals.


Assuntos
Autoanticorpos/imunologia , Doença de Graves/imunologia , Tolerância Imunológica/efeitos dos fármacos , Nanopartículas/química , Receptores da Tireotropina/imunologia , Animais , Doença de Graves/patologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Compostos Orgânicos/química , Compostos Orgânicos/imunologia , Compostos Orgânicos/farmacologia , Receptores da Tireotropina/química
4.
Mol Biol Rep ; 48(11): 7443-7456, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34716862

RESUMO

BACKGROUND: Survival rate of patients affected with anaplastic thyroid carcinoma (ATC) is less than 5% with current treatment. In ATC, BRAFV600E mutation is the major mutation that results in the transformation of normal cells in to an undifferentiated cancer cells via aberrant molecular signaling mechanisms. Although vemurufenib is a selective oral drug for the BRAFV600E mutant kinase with a response rate of nearly 50% in metastatic melanoma, our study has showed resistance to this drug in ATC. Hence the rationale of the study is to explore combinational therapeutic effect to improve the efficacy of vemurafenib along with metformin. Metformin, a diabetic drug is an AMPK activator and has recently proved to be involved in preventing or treating several types of cancer. METHODS AND RESULTS: Using iGEMDock software, a protein-ligand interaction was successful between Metformin and TSHR (receptor present in the thyroid follicular cells). Our study demonstrates that combination of vemurufenib with metformin has synergistic anti-cancer effects which was evaluated through MTT assay (cytotoxicity), colony formation assay (antiproliferation evaluation) and suppressed the progression of ATC cells growth by inducing significant apoptosis, proven by Annexin V-FITC assay (Early Apoptosis Detection). Downregulation of ERK signaling, upregulation of AMPK pathway and precision in epithelial-mesenchymal transition (EMT) pathway which were assessed by RT-PCR and Western blot provide the evidence that the combination of drugs involved in the precision of altered molecular signaling Further our results suggest that Metformin act as a demethylating agent in anaplastic thyroid cancer cells by inducing the expression of NIS and TSHR. Our study for the first time explored cAMP signaling in ATC wherein cAMP signaling is downregulated due to decrease in intracellular cAMP level upon metformin treatment. CONCLUSION: To conclude, our findings demonstrate novel therapeutic targets and treatment strategies for undifferentiated ATC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Proteínas de Neoplasias , Receptores da Tireotropina , Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Metformina/química , Metformina/farmacologia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Receptores da Tireotropina/química , Receptores da Tireotropina/metabolismo , Carcinoma Anaplásico da Tireoide/química , Carcinoma Anaplásico da Tireoide/tratamento farmacológico , Carcinoma Anaplásico da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/química , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/metabolismo , Vemurafenib/química , Vemurafenib/farmacologia
5.
J Autoimmun ; 108: 102402, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31980336

RESUMO

We have previously identified a signature HLA-DR3 pocket variant, designated HLA-DRß1-Arg74 that confers a high risk for Graves' Disease (GD). In view of the key role of HLA-DRß1-Arg74 in triggering GD we hypothesized that thyroid-stimulating hormone receptor (TSHR) peptides that bind to the HLA-DRß1-Arg74 pocket with high affinity represent key pathogenic TSHR peptides triggering GD, and that blocking their presentation to CD4+ T-cells can be used as a novel therapeutic approach in GD. There were several previous attempts to identify the major pathogenic TSHR peptide utilizing different methodologies, however the results were inconsistent and inconclusive. Therefore, the aim of our study was to use TSHR peptide binding affinity to HLA-DRß1-Arg74 as a method to identify the key pathogenic TSHR peptides that trigger GD. Using virtual screening and ELISA and cellular binding assays we identified 2 TSHR peptides that bound with high affinity to HLA-DRß1-Arg74 - TSHR.132 and TSHR.197. Peptide immunization studies in humanized DR3 mice showed that only TSHR.132, but not TSHR.197, induced autoreactive T-cell proliferation and cytokine responses. Next, we induced experimental autoimmune Graves' disease (EAGD) in a novel BALB/c-DR3 humanized mouse model we created and confirmed TSHR.132 as a major DRß1-Arg74 binding peptide triggering GD in our mouse model. Furthermore, we demonstrated that Cepharanthine, a compound we have previously identified as DRß1-Arg74 blocker, could block the presentation and T-cell responses to TSHR.132 in the EAGD model.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Benzilisoquinolinas/farmacologia , Antígeno HLA-DR3/imunologia , Peptídeos/antagonistas & inibidores , Peptídeos/imunologia , Receptores da Tireotropina/imunologia , Sequência de Aminoácidos , Animais , Benzilisoquinolinas/química , Mapeamento de Epitopos/métodos , Epitopos de Linfócito T/imunologia , Citometria de Fluxo , Doença de Graves/diagnóstico , Doença de Graves/tratamento farmacológico , Doença de Graves/imunologia , Antígeno HLA-DR3/genética , Humanos , Imuno-Histoquímica , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Moleculares , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/imunologia , Peptídeos/química , Ligação Proteica , Receptores da Tireotropina/química , Relação Estrutura-Atividade , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
Mol Pharmacol ; 96(4): 452-462, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31399504

RESUMO

The large TSH-bound ectodomain of the thyrotropin receptor (TSHR) activates the transmembrane domain (TMD) indirectly via an internal agonist (IA). The ectodomain/TMD interface consists of a converging helix, a Cys-Cys-bridge-linked IA, and extracellular loops (ECL). To investigate the intramolecular course of molecular activation, especially details of the indirect activation, we narrowed down allosteric inhibition sites of negative allosteric modulator (NAM) by mutagenesis, homology modeling, and competition studies with positive allosteric modulator (PAM). From the inhibitory effects of NAM S37a on: 1) chimeras with swapped ectodomain, 2) stepwise N-terminal truncations, 3) distinct constitutively active mutations distributed across the hinge region and ECL, but not across the TMD, we conclude that S37a binds at the ectodomain/TMD interface, between the converging helix, ECL1, and the IA. This is also supported by the noncompetitive inhibition of PAM-C2-activation by S37a in the TSHR-TMD construct lacking the ectodomain. Mutagenesis studies on the IA and ECL were guided by our refined model of the ectodomain/TMD interface and indicate an interaction with the TSHR-specific residues E404 (preceding IA) and H478 (ECL1). At this new allosteric interaction site, NAM S37a blocks both TSH- and PAM-induced activation of the TSHR. Our refined models, mutations, and new allosteric binding pocket helped us to gain more detailed insights into the intramolecular course of TSHR activation at the ectodomain/TMD interface, including the delocalization of the converging helix and rearrangement of the conformation of IA. These changes are embedded between the ECL and cooperatively trigger active conformations of TMD. SIGNIFICANCE STATEMENT: The intramolecular activation mechanisms of the TSHR appear to be distinct from those of other G protein-coupled receptors, as the TSHR has a uniquely large N-terminal ectodomain that includes the hormone binding site and an internal agonist sequence. We present new molecular and structural insights into the interface between ectodomain and transmembrane domain in the TSHR, as well as the transfer of activation to the transmembrane domain. This knowledge is critical for understanding activation or inhibition of the receptor by allosteric ligands. We have identified a new allosteric antagonist binding pocket that is located exactly at this interface and possesses specific features that may allow the generation of potent highly TSHR-selective drugs, of potential value for the treatment of Graves' orbitopathy.


Assuntos
Receptores da Tireotropina/química , Receptores da Tireotropina/metabolismo , Tireotropina/metabolismo , Regulação Alostérica , Regulação da Expressão Gênica , Células HEK293 , Humanos , Modelos Moleculares , Mutação , Domínios Proteicos , Receptores da Tireotropina/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais
7.
Int J Mol Sci ; 20(22)2019 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-31703413

RESUMO

1) Background: Central congenital hypothyroidism (CCH) is a rare endocrine disorder that can be caused by mutations in the ß-subunit of thyrotropin (TSHB). The TSHB mutation C105Vfs114X leads to isolated thyroid-stimulating-hormone-(TSH)-deficiency and results in a severe phenotype. The aim of this study was to gain more insight into the underlying molecular mechanism and the functional effects of this mutation based on two assumptions: a) the three-dimensional (3D) structure of TSH should be modified with the C105V substitution, and/or b) whether the C-terminal modifications lead to signaling differences. 2) Methods: wild-type (WT) and different mutants of hTSH were generated in human embryonic kidney 293 cells (HEK293 cells) and TSH preparations were used to stimulate thyrotropin receptor (TSHR) stably transfected into follicular thyroid cancer cells (FTC133-TSHR cells) and transiently transfected into HEK293 cells. Functional characterization was performed by determination of Gs, mitogen activated protein kinase (MAPK) and Gq/11 activation. 3) Results: The patient mutation C105Vfs114X and further designed TSH mutants diminished cyclic adenosine monophosphate (cAMP) signaling activity. Surprisingly, MAPK signaling for all mutants was comparable to WT, while none of the mutants induced PLC activation. 4) Conclusion: We characterized the patient mutation C105Vfs114X concerning different signaling pathways. We identified a strong decrease of cAMP signaling induction and speculate that this could, in combination with diverse signaling regarding the other pathways, accounting for the patient's severe phenotype.


Assuntos
Hipotireoidismo Congênito , Sistema de Sinalização das MAP Quinases , Mutação , Receptores da Tireotropina , Sistemas do Segundo Mensageiro , Tireotropina Subunidade beta , Linhagem Celular Tumoral , Hipotireoidismo Congênito/genética , Hipotireoidismo Congênito/metabolismo , AMP Cíclico/genética , AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Domínios Proteicos , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Receptores da Tireotropina/metabolismo , Tireotropina Subunidade beta/química , Tireotropina Subunidade beta/genética , Tireotropina Subunidade beta/metabolismo
8.
Horm Metab Res ; 50(12): 894-907, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30366347

RESUMO

The thyrotropin receptor (TSHR), the lutropin- (LHR), and the follicotropin receptor (FSHR) belong to glycoprotein hormone receptors (GPHR), a subgroup of the class A G-protein coupled receptors. In this review, the unique features of GPHR have been taken into account for their pharmacological interventions: i) The respective hormone and stimulating or blocking antibodies are binding on the large ectodomain that is ii) via a hinge region, containing iii) an internal tethered agonist linked to the transmembrane domain. iv) Multimerization and mechanisms for negative or positive cooperativity of GPHR upon ligand binding and v) dimer- and oligomeric arrangements enabling trans-activation on GPHR signaling are considered. Available knowledge concerning the modulation of the GPHR (mal)-function and associated structural aspects by diverse entities such as antibodies, chaperones, peptides, small molecule agonists, inverse agonists, and antagonists is summarized. The TSHR is important with respect to autoimmune [Graves' disease (GD), Graves' orbitopathy (GO)] or non-autoimmune thyroid dysfunctions and cancer-development. To date there is neither an agonist nor antagonist modulator of pathogenic such as TSHR signaling in the clinics. However, several different ligands monoclonal stimulating and inhibiting antibodies and small molecule drug-like ligands have been reported in the last decade. In special focus are the most recent findings regarding the development and use of small molecule TSHR ligands. Finally, limitations of current knowledge and lack of information are discussed highlighting the need for intensified efforts towards understanding the interplay of TSHR multimers, especially their interaction with drug-like ligands. Important in this context is the biased ligand development.


Assuntos
Receptores da Tireotropina/metabolismo , Regulação Alostérica , Animais , Humanos , Ligantes , Mutação/genética , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
9.
Int J Mol Sci ; 19(9)2018 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-30227620

RESUMO

The key proteins responsible for hormone synthesis in the thyroid are glycosylated. Oligosaccharides strongly affect the function of glycosylated proteins. Both thyroid-stimulating hormone (TSH) secreted by the pituitary gland and TSH receptors on the surface of thyrocytes contain N-glycans, which are crucial to their proper activity. Thyroglobulin (Tg), the protein backbone for synthesis of thyroid hormones, is a heavily N-glycosylated protein, containing 20 putative N-glycosylated sites. N-oligosaccharides play a role in Tg transport into the follicular lumen, where thyroid hormones are produced, and into thyrocytes, where hyposialylated Tg is degraded. N-glycans of the cell membrane transporters sodium/iodide symporter and pendrin are necessary for iodide transport. Some changes in glycosylation result in abnormal activity of the thyroid and alteration of the metabolic clearance rate of hormones. Alteration of glycan structures is a pathological process related to the progression of chronic diseases such as thyroid cancers and autoimmunity. Thyroid carcinogenesis is accompanied by changes in sialylation and fucosylation, ß1,6-branching of glycans, the content and structure of poly-LacNAc chains, as well as O-GlcNAcylation, while in thyroid autoimmunity the main processes affected are sialylation and fucosylation. The glycobiology of the thyroid gland is an intensively studied field of research, providing new data helpful in understanding the role of the sugar component in thyroid protein biology and disorders.


Assuntos
Doenças da Glândula Tireoide/metabolismo , Doenças da Glândula Tireoide/patologia , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Animais , Glicosilação , Humanos , Polissacarídeos/análise , Polissacarídeos/metabolismo , Receptores da Tireotropina/química , Receptores da Tireotropina/metabolismo , Transportadores de Sulfato/química , Transportadores de Sulfato/metabolismo , Simportadores/química , Simportadores/metabolismo , Tireoglobulina/química , Tireoglobulina/metabolismo , Glândula Tireoide/citologia , Tireotropina/química , Tireotropina/metabolismo
10.
J Biol Chem ; 291(27): 14095-14108, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27129207

RESUMO

The thyroid stimulating hormone receptor (TSHR) is a G protein-coupled receptor (GPCR) with a characteristic large extracellular domain (ECD). TSHR activation is initiated by binding of the hormone ligand TSH to the ECD. How the extracellular binding event triggers the conformational changes in the transmembrane domain (TMD) necessary for intracellular G protein activation is poorly understood. To gain insight in this process, the knowledge on the relative positioning of ECD and TMD and the conformation of the linker region at the interface of ECD and TMD are of particular importance. To generate a structural model for the TSHR we applied an integrated structural biology approach combining computational techniques with experimental data. Chemical cross-linking followed by mass spectrometry yielded 17 unique distance restraints within the ECD of the TSHR, its ligand TSH, and the hormone-receptor complex. These structural restraints generally confirm the expected binding mode of TSH to the ECD as well as the general fold of the domains and were used to guide homology modeling of the ECD. Functional characterization of TSHR mutants confirms the previously suggested close proximity of Ser-281 and Ile-486 within the TSHR. Rigidifying this contact permanently with a disulfide bridge disrupts ligand-induced receptor activation and indicates that rearrangement of the ECD/extracellular loop 1 (ECL1) interface is a critical step in receptor activation. The experimentally verified contact of Ser-281 (ECD) and Ile-486 (TMD) was subsequently utilized in docking homology models of the ECD and the TMD to create a full-length model of a glycoprotein hormone receptor.


Assuntos
Receptores da Tireotropina/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Glicosilação , Humanos , Espectrometria de Massas , Modelos Moleculares , Mutação , Proteólise , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Ressonância de Plasmônio de Superfície
11.
J Endocrinol Invest ; 40(1): 41-48, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27531173

RESUMO

OBJECTIVE: Thyroid-stimulating hormone (TSH) binds TSH receptor (TSHR) on thyroid cell membranes, which will lead activation of cyclic adenosine 3',5'-monophosphate/protein kinase A signaling pathway. Through this pathway, TSHR regulates the expression of sodium-iodide symporter (NIS) to complete iodine intake. In recent studies, it is found that TSHR is widely expressed in a variety of extra-thyroidal tissues. TSHR expressions as well as distribution in normal mammary gland tissues have not been reported. The physiological mechanism of the TSHR in the extra-thyroidal tissues has also been controversial. METHODS: In this study, immunohistochemistry and immunofluorescence were used to characterize the expression distribution of TSHR protein in lactating breast. DNA sequence of TSHR cDNA from mice lactating breast was determined and then compared with TSHR cDNA from mice thyroidal tissue. RESULTS: A 173 amino acid (AA) fragment deletion was found in the extra-cellular domain of lactating breast TSHR. The expression levels of NIS mRNA were compared between two tissues, and the level of NIS mRNA in lactating breasts was lower than the one in thyroidal tissues. CONCLUSION: The lower expression of NIS in lactating breast may be due to the 173 AA deletion in the TSHR resulting the lower binding of TSH to the TSHR. For the first time, this finding may explain the reason of the lower NIS expression in lactating breast.


Assuntos
Biomarcadores/metabolismo , Mama/metabolismo , Regulação da Expressão Gênica , Lactação/fisiologia , Receptores da Tireotropina/química , Simportadores/metabolismo , Glândula Tireoide/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Células Cultivadas , Feminino , Imunofluorescência , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Receptores da Tireotropina/genética , Receptores da Tireotropina/metabolismo , Transdução de Sinais , Simportadores/genética
12.
Biophys J ; 111(12): 2620-2628, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-28002738

RESUMO

The signaling of a G protein-coupled receptor (GPCR) is dictated by the complementary responsiveness of interacting intracellular effectors such as G proteins. Many GPCRs are known to couple to more than one G protein subtype and induce a multitude of signaling pathways, although the in vivo relevance of particular pathways is mostly unrecognized. Dissecting GPCR signaling in terms of the pathways that are activated will boost our understanding of the molecular fundamentals of hormone action. The structural determinants governing the selectivity of GPCR/G protein coupling, however, remain obscure. Here, we describe the design of soluble GPCR mimetics to study the details of the interplay between G-proteins and activators. We constructed functional mimetics of the intracellular domain of a model GPCR, the thyrotropin receptor. We based the construction on a unique scaffold, 6-Helix, an artificial protein that was derived from the elements of the trimer-of-hairpins structure of HIV gp41 and represents a bundle of six α-helices. The 6-Helix scaffold, which endowed the substituted thyrotropin receptor intracellular domain elements with spatial constraints analogous to those found in native receptors, enabled the reconstitution of a microdomain that consists of intracellular loops 2 and 3, and is capable of binding and activating Gα-(s). The 6-Helix-based mimetics could be used as a platform to study the molecular basis of GPCR/G protein recognition. Such knowledge could help investigators develop novel therapeutic strategies for GPCR-related disorders by targeting the GPCR/G protein interfaces and counteracting cellular dysfunctions via focused tuning of GPCR signaling.


Assuntos
Materiais Biomiméticos/química , Espaço Intracelular/metabolismo , Receptores da Tireotropina/química , Materiais Biomiméticos/metabolismo , Modelos Moleculares , Conformação Proteica em alfa-Hélice , Domínios Proteicos
13.
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
14.
Endocr J ; 62(5): 393-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25797365

RESUMO

Genetic defects of hormone receptors are the most common form of end-organ hormone resistance. One example of such defects is TSH resistance, which is caused by biallelic inactivating mutations in the TSH receptor gene (TSHR). TSH, a master regulator of thyroid functions, affects virtually all cellular processes involving thyroid hormone production, including thyroidal iodine uptake, thyroglobulin iodination, reuptake of iodinated thyroglobulin and thyroid cell growth. Resistance to TSH results in defective thyroid hormone production from the neonatal period, namely congenital hypothyroidism. Classically, clinical phenotypes of TSH resistance due to inactivating TSHR mutations were thought to vary depending on the residual mutant receptor activity. Nonfunctional mutations in the two alleles produce severe thyroid hypoplasia with overt hypothyroidism (uncompensated TSH resistance), while hypomorphic mutations in at least one allele produce normal-sized thyroid gland with preserved hormone-producing capacity (compensated TSH resistance). More recently, a new subgroup of TSH resistance (nonclassic TSH resistance) that is characterized by paradoxically high thyroidal iodine uptake has been reported. In this article, the pathophysiology and clinical features of TSH resistance due to inactivating TSHR mutations are reviewed, with particular attention to the nonclassic form.


Assuntos
Síndrome da Resistência aos Hormônios Tireóideos , Tireotropina , Alelos , Hipotireoidismo Congênito/genética , Genótipo , Humanos , Iodo/metabolismo , Mutação , Fenótipo , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Receptores da Tireotropina/fisiologia , Disgenesia da Tireoide/genética , Glândula Tireoide/patologia , Glândula Tireoide/fisiopatologia , Síndrome da Resistência aos Hormônios Tireóideos/complicações , Síndrome da Resistência aos Hormônios Tireóideos/genética , Hormônios Tireóideos/biossíntese
15.
Tsitologiia ; 56(7): 526-35, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25696997

RESUMO

The regulation of the specific activity of the thyroid gland is carried by thyroid-stimulating hormone (TSH) through TSH receptor (TSHR). This receptor is coupled to different types of G-proteins, including the G(s)-proteins, through which TSH stimulates the enzyme adenylyl cyclase (AC). As the application of TSH in medicine is limited, the development of selective regulators of TSHR with agonistic and antagonistic activity is carried out. One of the approaches to their creation is to develop the peptides corresponding to functionally important regions of TSHR which are located in its intracellular loops (ICL) and are involved in the binding and activation of G-proteins. We have synthesized peptide corresponding to the C-terminal region 612-627 of the third ICL of TSHR and its derivatives modified by palmitic acid residue (at the N- or the C-terminus) or by polylysine dendrimer (at the N-terminus), and studied their effect on the basal and TSH-stimulated AC activity in the membrane fraction isolated from the rat thyroid. The most active was peptide 612-627-K(Pal)A modified by palmitate at the C-terminus, where in TSHR the hydrophobic transmembrane region is located. At the micromolar concentrations the peptide increased AC activity and reduced the AC stimulating effect of TSH. The action of the 612-627-K(Pal)A has been directed onto TSHR homologous to it, as indicated by the following facts: 1) the inhibition of G(s)-protein, the downstream component of AC system, by treating the membranes with cholera toxin led to the blocking of peptide AC effect, 2) this effect was not detected in the tissues where no TSHR, 3) the peptide did not significantly affect the AC stimulating effects of hormones acting via other receptors. The unmodified peptide and the peptide with N-terminal dendrimer are far behind the 612-627-K(Pal)A in their ability to activate AC in the thyroid, while the peptide modified by palmitate at the N-terminus was inactive. At the same time, the peptide modified by dendrimer was comparable to the 612-627-K(Pal)A in the ability to inhibit the AC effect of TSH, but, although to a lesser extent that it decreased the AC effects of other hormones, demonstrating the low receptor specificity. Thus, these data point to the high efficiency of peptide 612-627-K(Pal)A, as a regulator of TSHR, and the prospects of creating the drugs based on it to control the thyroid functions in pathology.


Assuntos
Adenilil Ciclases/metabolismo , Membrana Celular/efeitos dos fármacos , Peptídeos/farmacologia , Receptores da Tireotropina/metabolismo , Glândula Tireoide/efeitos dos fármacos , Adenilil Ciclases/genética , Sequência de Aminoácidos , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Membrana Celular/metabolismo , Regulação da Expressão Gênica , Masculino , Dados de Sequência Molecular , Miocárdio/citologia , Miocárdio/metabolismo , Ácido Palmítico/química , Peptídeos/síntese química , Peptídeos/química , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Polilisina/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores da Tireotropina/agonistas , Receptores da Tireotropina/antagonistas & inibidores , Receptores da Tireotropina/química , Transdução de Sinais , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Tireotropina/metabolismo , Tireotropina/farmacologia
16.
Endocrinology ; 165(10)2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39116382

RESUMO

The TSH receptor (TSHR) and its many forms are the primary antigens of Graves' disease as evidenced by the presence of TSHR antibodies of differing biological activity. The TSH holoreceptor undergoes complex posttranslational changes including cleavage of its ectodomain and oligomer formation. We have previously shown that the TSHR exists in both monomeric and dimeric structures in the thyroid cell membrane and have demonstrated, by modeling, that the transmembrane domains (TMD) can form stable dimeric structures. Based on these earlier simulations of the TSHR-TMD structure and our most recent model of the full-length TSHR, we have now built models of full-length TSHR multimers with and without TSH ligand in addition to multimers of the extracellular leucine-rich domain, the site of TSH and autoantibody binding. Starting from these models we ran molecular dynamics simulations of the receptor oligomers solvated with water and counterions; the full-length oligomers also were embedded in a dipalmitoylphosphatidylcholine bilayer. The full-length TSHR dimer and trimer models stayed in the same relative orientation and distance during 2000 ns (or longer) molecular dynamics simulation in keeping with our earlier report of TMD dimerization. Simulations were also performed to model oligomers of the leucine-rich domain alone; we found a trimeric complex to be even more stable than the dimers. These data provide further evidence that different forms of the TSHR add to the complexity of the immune response to this antigen that, in patients with autoimmune thyroid disease, generate an autoantibody reactome with multiple types of autoantibody to the TSHR.


Assuntos
Autoanticorpos , Simulação de Dinâmica Molecular , Multimerização Proteica , Receptores da Tireotropina , Receptores da Tireotropina/imunologia , Receptores da Tireotropina/química , Humanos , Autoanticorpos/imunologia , Doença de Graves/imunologia , Domínios Proteicos
17.
J Mol Graph Model ; 129: 108725, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38373379

RESUMO

The receptor for thyroid stimulating hormone (TSHR), a GPCR, is the primary antigen in autoimmune hyperthyroidism (Graves' disease) caused by stimulating TSHR antibodies. While we have previously published a full length model of the TSHR, including its leucine rich domain (LRD), linker region (LR) and transmembrane domain (TMD), to date, only a partial LRD (aa 21-261) stabilized with TSHR autoantibodies has been crystallized. Recently, however, cryo-EM structures of the full-length TSHR have been published but they include only an incomplete LR. We have now utilized the cryo-EM models, added disulfide bonds to the LR and performed longer (3000 ns) molecular dynamic (MD) simulations to update our previous model of the entire full-length TSHR, with and without the presence of TSH ligand. As in our earlier work, the new model was embedded in a lipid membrane and was solvated with water and counterions. We found that the 3000 ns Molecular Dynamic simulations showed that the structure of the LRD and TMD were remarkably constant while the LR, known more commonly as the "hinge region", again showed significant flexibility, forming several transient secondary structural elements. Analysis of the new simulations permitted a detailed examination of the effect of TSH binding on the structure of the TSHR. We found a structure-stabilizing effect of TSH, including increased stability of the LR, which was clearly demonstrated by analyzing several intrinsic receptor properties including hydrogen bonding, fluctuation of the LRD orientation, and radius of gyration. In conclusion, we were able to quantify the flexibility of the TSHR and show its increased stability after TSH binding. These data indicated the important role of ligands in directing the signaling structure of a receptor.


Assuntos
Receptores da Tireotropina , Tireotropina , Receptores da Tireotropina/química , Receptores da Tireotropina/metabolismo , Ligantes , Tireotropina/química , Tireotropina/metabolismo , Leucina
18.
Biochem Soc Trans ; 41(1): 213-7, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23356285

RESUMO

The TSHR (thyrotropin receptor) is activated endogenously by the large hormone thyrotropin and activated pathologically by auto-antibodies. Both activate and bind at the extracellular domain. Recently, SMLs (small-molecule ligands) have been identified, which bind in an allosteric binding pocket within the transmembrane domain. Modelling driven site-directed mutagenesis of amino acids lining this pocket led to the delineation of activation and inactivation sensitive residues. Modified residues showing CAMs (constitutively activating mutations) indicate signalling-sensitive positions and mark potential trigger points for agonists. Silencing mutations lead to an impairment of basal activity and mark contact points for antagonists. Mapping these residues on to a structural model of TSHR indicates locations where an SML may switch the receptor to an inactive or active conformation. In the present article, we report the effects of SMLs on these signalling-sensitive amino acids at the TSHR. Surprisingly, the antagonistic effect of SML compound 52 was reversed to an agonistic effect, when tested at the CAM Y667A. Switching agonism to antagonism and the reverse by changing either SMLs or residues covering the binding pocket provides detailed knowledge about discriminative pharmacophores. It prepares the basis for rational optimization of new high-affinity antagonists to interfere with the pathogenic activation of the TSHR.


Assuntos
Sítio Alostérico , Receptores da Tireotropina/metabolismo , Sítios de Ligação , Modelos Moleculares , Receptores da Tireotropina/química
19.
Nat Genet ; 7(3): 396-401, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7920658

RESUMO

The thyrotropin receptor (TSHR), a member of the large family of G protein-coupled receptors, controls both the function and growth of thyroid cells via stimulation of adenylyl cyclase. We report two different mutations in the TSHR gene of affected members of two large pedigrees with non-autoimmune autosomal dominant hyperthyroidism (toxic thyroid hyperplasia), that involve residues in the third (Val509Ala) and seventh (Cys672Tyr) transmembrane segments. When expressed by transfection in COS-7 cells, the mutated receptors display a higher constitutive activation of adenylyl cyclase than wild type. This new disease entity is the germline counterpart of hyperfunctioning thyroid adenomas, in which different somatic mutations with similar functional characteristics have been demonstrated.


Assuntos
Genes Dominantes , Hipertireoidismo/genética , Mutação Puntual , Receptores da Tireotropina/genética , Adenoma/genética , Adenilil Ciclases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , AMP Cíclico/fisiologia , Análise Mutacional de DNA , Ativação Enzimática , Feminino , França/epidemiologia , Humanos , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Linhagem , Conformação Proteica , Receptores da Tireotropina/química , Receptores da Tireotropina/fisiologia , Sistemas do Segundo Mensageiro , Neoplasias da Glândula Tireoide/genética , Transfecção
20.
J Biol Chem ; 286(8): 6219-24, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21190937

RESUMO

Thyroid-stimulating hormone (TSH)-induced reduction in ligand binding affinity (negative cooperativity) requires TSH receptor (TSHR) homodimerization, the latter involving primarily the transmembrane domain (TMD) but with the extracellular domain (ECD) also contributing to this association. To test the role of the TMD in negative cooperativity, we studied the TSHR ECD tethered to the cell surface by a glycosylphosphatidylinositol (GPI) anchor that multimerizes despite the absence of the TMD. Using the infinite ligand dilution approach, we confirmed that TSH increased the rate of dissociation (k(off)) of prebound (125)I-TSH from CHO cells expressing the TSH holoreceptor. Such negative cooperativity did not occur with TSHR ECD-GPI-expressing cells. However, even in the absence of added TSH, (125)I-TSH dissociated much more rapidly from the TSHR ECD-GPI than from the TSH holoreceptor. This phenomenon, suggesting a lower TSH affinity for the former, was surprising because both the TSHR ECD and TSH holoreceptor contain the entire TSH-binding site, and the TSH binding affinities for both receptor forms should, theoretically, be identical. In ligand competition studies, we observed that the TSH binding affinity for the TSHR ECD-GPI was significantly lower than that for the TSH holoreceptor. Further evidence for a difference in ligand binding kinetics for the TSH holoreceptor and TSHR ECD-GPI was obtained upon comparison of the TSH K(d) values for these two receptor forms at 4 °C versus room temperature. Our data provide the first evidence that the wild-type TSHR TMD influences ligand binding affinity for the ECD, possibly by altering the conformation of the closely associated hinge region that contributes to the TSH-binding site.


Assuntos
Receptores da Tireotropina/metabolismo , Tireotropina/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Humanos , Cinética , Estrutura Terciária de Proteína , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Tireotropina/química , Tireotropina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA