Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 627(8003): 407-415, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38383779

RESUMO

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Assuntos
Aquaporina 4 , Autoanticorpos , Autoantígenos , Linfócitos B , Tolerância Imunológica , Neuromielite Óptica , Animais , Humanos , Camundongos , 60533 , Aquaporina 4/deficiência , Aquaporina 4/genética , Aquaporina 4/imunologia , Aquaporina 4/metabolismo , Autoanticorpos/imunologia , Autoantígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD40/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Neuromielite Óptica/imunologia , Neuromielite Óptica/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo , Transcriptoma
2.
Thyroid ; 32(1): 90-96, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34714162

RESUMO

Background: The pathogenesis of Graves' hyperthyroidism (GH) and associated Graves' orbitopathy (GO) appears to involve stimulatory autoantibodies (thyrotropin receptor [TSHR]-stimulating antibodies [TSAbs]) that bind to and activate TSHRs on thyrocytes and orbital fibroblasts. In general, measurement of circulating TSHR antibodies by clinical assays correlates with the status of GH and GO. However, most clinical measurements of TSHR antibodies use competitive binding assays that do not distinguish between TSAbs and antibodies that bind to but do not activate TSHRs. Moreover, clinical assays for TSAbs measure stimulation of only one signaling pathway, the cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) pathway, in engineered cells that are not thyrocytes or orbital fibroblasts. We determined whether measuring TSAbs by a cAMP-PKA readout in engineered cells accurately reveals the efficacies of stimulation by these antibodies on thyrocytes and orbital fibroblasts. Methods: We measured TSAb stimulation of normal human thyrocytes and orbital fibroblasts from patients with GO in primary cultures in vitro. In thyrocytes, we measured secretion of thyroglobulin (TG) and in orbital fibroblasts secretion of hyaluronan (hyaluronic acid [HA]). We also measured stimulation of cAMP production in engineered TSHR-expressing cells in an assay similar to clinical assays. Furthermore, we determined whether there were differences in stimulation of thyrocytes and orbital fibroblasts by TSAbs from patients with GH alone versus from patients with GO understanding that patients with GO have accompanying GH. Results: We found a positive correlation between TSAb stimulation of cAMP production in engineered cells and TG secretion by thyrocytes as well as HA secretion by orbital fibroblasts. However, TSAbs from GH patients stimulated thyrocytes more effectively than TSAbs from GO patients, whereas TSAbs from GO patients were more effective in activating orbital fibroblasts than TSAbs from GH patients. Conclusions: Clinical assays of stimulation by TSAbs measuring activation of the cAMP-PKA pathway do correlate with stimulation of thyrocytes and orbital fibroblasts; however, they do not distinguish between TSAbs from GH and GO patients. In vitro, TSAbs exhibit selectivity in activating TSHRs since TSAbs from GO patients were more effective in stimulating orbital fibroblasts and TSAbs from GH patients were more effective in stimulating thyrocytes.


Assuntos
Autoanticorpos/imunologia , Fibroblastos/imunologia , Oftalmopatia de Graves/complicações , Células Epiteliais da Tireoide/imunologia , Adulto , Autoanticorpos/análise , Feminino , Fibroblastos/metabolismo , Doença de Graves/sangue , Doença de Graves/imunologia , Oftalmopatia de Graves/sangue , Oftalmopatia de Graves/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Células Epiteliais da Tireoide/metabolismo , Tireotropina/metabolismo
3.
Endocrinology ; 162(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34496027

RESUMO

Hashimoto thyroiditis (HT) is a pathology that often causes a gradual thyroid insufficiency in affected patients due to the autoimmune destruction of this gland. The cellular immune response mediated by T helper lymphocytes TH1 and TH17 can induce the HT disease. In this pathologic condition, there is an imbalance between the TH17 and Treg lymphocytes as well as a gut microbiota dysfunction. The objective of this work was to describe the interactions of the cell subpopulations that participate in HT. To achieve this goal, we generated a mathematical model that allowed the simulation of different scenarios for the dynamic interaction between thyroid cells, the immune system, and the gut microbiota. We used a hypothetical-deductive design of mathematical modeling based on a system of ordinary differential equations, where the state variables are the TH1, TH17, and Treg lymphocytes, the thyrocytes, and the bacteria from gut microbiota. This work generated a compartmental model of the cellular immune response occurring in the thyroid gland. It was observed that TH1 and TH17 lymphocytes could increase the immune cells' activity, as well as activate effector cells directly and trigger the apoptosis and inflammation processes of healthy thyrocytes indirectly. Likewise, the model showed that a reduction in Treg lymphocytes could increase the activity of TH17 lymphocytes when an imbalance of the gut microbiota composition occurred. The numerical results highlight the TH1, TH17, and bacterial balance of the gut microbiota activities as important factors for the development of HT disease.


Assuntos
Doença de Hashimoto/imunologia , Doença de Hashimoto/patologia , Modelos Teóricos , Animais , Simulação por Computador , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Doença de Hashimoto/microbiologia , Humanos , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/patologia , Tireoidite Autoimune/imunologia , Tireoidite Autoimune/microbiologia , Tireoidite Autoimune/patologia
4.
Front Endocrinol (Lausanne) ; 11: 579648, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33304319

RESUMO

Prunella vulgaris (PV), a perennial herb, has been used to treat thyroid diseases in China for over 2,000 years. In particular, its therapeutic effect has been described for Hashimoto's thyroiditis, including reducing titers autoantibodies against thyroid peroxidase and thyroglobulin of and T helper 17 (Th17) cells. However, the underlying mechanism for how PV exerts such effects has not been investigated. We examined the effects of PV on innate immune activation, which is thought to be one of the triggers for the development of autoimmune diseases, including Hashimoto's thyroiditis. In cultured thyrocytes, PV reduced mRNA levels of inflammatory cytokines that were originally induced as a result of innate immune activation initiated by transfection of double-stranded DNA (dsDNA) or dsRNA. PV suppressed activation of nuclear factor κB (NF-κB) and interferon regulatory factor 3 (IRF3), and suppressed corresponding promoter activation, which were initially activated by dsDNA or dsRNA. PV also suppressed the mRNA levels of molecules responsible for antigen processing and presentation, and PV protected thyrocytes from apoptosis induced by dsDNA and dsRNA. Additionally, PV suppressed the expression of genes involved in iodide uptake and oxidation. Taken together, these results suggest that PV exerts its protective effect on thyrocytes by suppressing both innate and adaptive immune responses and cell death. PV may also protect cells from iodide-associated oxidative injury. This report is among the first to identify the mechanisms to explain PV's beneficial effects in Hashimoto's thyroiditis.


Assuntos
Citocinas/metabolismo , Doença de Hashimoto/tratamento farmacológico , Imunidade Inata/imunologia , Inflamação/tratamento farmacológico , Extratos Vegetais/farmacologia , Prunella/química , Células Epiteliais da Tireoide/efeitos dos fármacos , Animais , Células Cultivadas , Citocinas/genética , Doença de Hashimoto/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Ratos , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo
5.
Biomolecules ; 10(2)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31979029

RESUMO

Antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) are involved in destruction of thyroid tissue in Hashimoto's thyroiditis (HT). N-glycosylation of the Fc fragment affects the effector functions of IgG by enhancing or suppressing the cytotoxicity effect. The aim of the present study was to assess the impact of HT-specific IgG glycosylation in ADCC and CDC, using in vitro models. The normal thyroid Nthy-ori 3-1 cell line and thyroid carcinoma FTC-133 cells were used as the target cells. Peripheral blood mononuclear cells (PBMCs) from healthy donors and the HL-60 human promyelotic leukemia cell line served as the effector cells. IgG was isolated from sera of HT and healthy donors and then treated with α2-3,6,8-neuraminidase to cut off sialic acids (SA) from N-glycans. We observed more intensive cytotoxicity in the presence of IgG from HT patients than in the presence of IgG from healthy donors. Removal of SA from IgG N-glycans increased ADCC intensity and reduced CDC. We conclude that the enhanced thyrocyte lysis resulted from the higher anti-TPO content in the whole IgG pool of HT donors and from altered IgG glycosylation in HT autoimmunity.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Proteínas do Sistema Complemento/imunologia , Doença de Hashimoto/imunologia , Imunoglobulina G/química , Autoanticorpos/química , Autoimunidade , Linhagem Celular Tumoral , Glicosilação , Células HL-60 , Humanos , Lectinas/química , Leucócitos Mononucleares/citologia , Polissacarídeos , Ácidos Siálicos/química , Células Epiteliais da Tireoide/imunologia , Glândula Tireoide/imunologia , Glândula Tireoide/fisiopatologia
6.
Surgery ; 167(1): 66-72, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31439400

RESUMO

BACKGROUND: An altered immune microenvironment may contribute to papillary thyroid cancer development, as immune infiltrates are identified postoperatively in many papillary thyroid cancer cases with or without diagnosed thyroiditis. Oxygen radicals, endogenous or inflammation-induced, can generate DNA damage, which causes mutations when repaired incorrectly. We hypothesized that infiltrating immune cells might promote aberrant DNA repair, predisposing thyrocytes to papillary thyroid cancer. METHODS: Quantitative reverse-transcriptase polymerase chain reaction assays measured gene expression in fresh-frozen samples (n = 55). RNA-seq data was obtained for papillary thyroid cancer and normal thyroid samples from the Cancer Genome Atlas (n = 564), and Hashimoto's-affected and normal thyroids from the Genotype-Tissue Expression project (n = 279). Immune cell marker expression levels were compared to histological estimates and to selected DNA repair genes. Immunohistochemistry localized gene expression to specific cell types. RESULTS: DNA polymerase theta expression by quantitative reverse-transcriptase Polymerase chain reaction was higher in papillary thyroid cancer and papillary thyroid cancer-adjacent samples than in benign normal thyroid (P < .001). Immune markers including CD4 correlated with DNA polymerase theta expression (r = 0.50) but not other DNA repair genes examined. Benign tissue with Hashimoto's exhibited increased DNA polymerase theta (P < .0001) and CD3E (P < .0001) expression. DNA polymerase theta localized to thyrocytes, not lymphocytes. CONCLUSION: We identified a strong correlation between immune cell infiltrate and dysregulated thyrocyte DNA repair genes, likely reflecting a pathway to papillary thyroid cancer development.


Assuntos
Carcinogênese/genética , Reparo do DNA/imunologia , Linfócitos do Interstício Tumoral/imunologia , Câncer Papilífero da Tireoide/imunologia , Neoplasias da Glândula Tireoide/imunologia , Adulto , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Carcinogênese/imunologia , DNA Polimerase Dirigida por DNA/imunologia , DNA Polimerase Dirigida por DNA/metabolismo , Feminino , Humanos , Masculino , RNA-Seq , Espécies Reativas de Oxigênio/imunologia , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/cirurgia , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo , Células Epiteliais da Tireoide/patologia , Glândula Tireoide/citologia , Glândula Tireoide/imunologia , Glândula Tireoide/patologia , Glândula Tireoide/cirurgia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/cirurgia , Tireoidectomia , Microambiente Tumoral/imunologia
7.
J Clin Endocrinol Metab ; 105(4)2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31784757

RESUMO

CONTEXT: Hepatitis C virus (HCV) infection is a prevalent disease worldwide. Thyroid dysfunction is one of the most common extrahepatic manifestations of HCV infection. We hypothesized that HCV can directly infect human thyrocytes thereby causing thyroid dysfunction. SETTING: Human thyrocytes in primary cell culture, ML-1 human thyroid cell line, and Huh7.5 human hepatocyte cell line were infected with HCV using the Huh7.5JFH1 cell line that releases infectious HCV virions. After infection, the release of new virions, production of proinflammatory cytokines, and expression of miR-122 were evaluated. Ribonucleic acid (RNA) extracted from HCV-infected cells and mock-infected cells was subjected to RNA sequencing and transcriptomic analysis. Ingenuity pathway analysis was used to detect up- and down-regulated pathways. RESULTS: Human thyrocytes express major HCV entry factors including CD81, occludin, claudin-1, and scavenger receptor class B1. Viral infection of thyroid cells was confirmed by detection of HCV core protein in supernatants and negative-sense HCV RNA in cell lysates. HCV infection of thyrocytes induced the production of the chemokine CXCL-8 and the proinflammatory cytokines tumor necrosis factor alpha (TNF-α) and significantly increased the expression of miR-122. Moreover, HCV infection of thyrocytes decreased expression of the thyroid peroxidase and thyroglobulin genes and increased expression of the deiodinase 2 gene. The top upregulated pathways in HCV-infected thyrocytes were immune pathways and metabolic pathways, while infected hepatocytes upregulated lipid and glucose metabolism pathways as previously reported. CONCLUSIONS: HCV infection may induce thyroid dysfunction by different mechanisms including direct infection of thyrocytes leading to activation of inflammatory pathways and upregulation of miR-122. These findings support a general mechanism for viral induction of autoimmunity through direct infection of target tissues.


Assuntos
Biomarcadores/análise , Hepacivirus/fisiologia , Hepatite C/virologia , Hepatócitos/virologia , Inflamação/virologia , MicroRNAs/genética , Células Epiteliais da Tireoide/virologia , Células Cultivadas , Citocinas/metabolismo , Feminino , Perfilação da Expressão Gênica , Hepatite C/imunologia , Hepatite C/metabolismo , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo , Replicação Viral
8.
Sci Rep ; 9(1): 10090, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31300682

RESUMO

Circulating T follicular helper (cTfh) cells have been identified as counterparts of germinal center Tfh (GC Tfh) cells in humans and can support T-dependent B cell maturation and antibody production in vitro. However, the role of cTfh cells in neutralizing antibody (nAb) responses in HCV infection remains unclear. Here, we characterized the phenotype and function of cTfh cells and demonstrated the associations of cTfh cells and their subsets with nAb responses in HCV infection. A total of 38 HCV-infected individuals and 28 healthy controls were enrolled from a pool of injection drug users. The frequency and function of blood Tfh cells were analyzed by flow cytometry. The titers and breadths of serum nAbs were measured using HCV pseudo-particle neutralization assays. Herein, we report several key observations. First, HCV infection skewed cTfh toward CXCR3+ cTfh cell differentiation. Second, the frequency of CXCR3+ cTfh cells positively correlated with HCV nAb titers and breadths. Third, CXCR3+ cTfh cells showed higher expression of Tfh-associated molecules (PD-1, ICOS, IL-21, Bcl-6) compared with CXCR3- cTfh cells from individuals with HCV infection. Coculture of cTfh cells and autologous memory B cells in vitro indicated that CXCR3+ cTfh cells show a superior ability to support HCV E2-specific B cell expansion compared with CXCR3- cTfh cells from individuals with HCV infection. HCV infection skews cTfh cells toward CXCR3-biased Tfh cell differentiation, which positively correlates with the magnitude and breadth of the HCV nAb response. It is our hope that these findings will provide insights for the rational design of a nAb-based HCV vaccine.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Hepacivirus/imunologia , Receptores CXCR3/sangue , Linfócitos T Auxiliares-Indutores/imunologia , Células Epiteliais da Tireoide/imunologia , Adulto , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Feminino , Centro Germinativo/citologia , Centro Germinativo/imunologia , Células HEK293 , Hepatite C/imunologia , Humanos , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Linfócitos T Auxiliares-Indutores/citologia , Células Epiteliais da Tireoide/citologia
9.
Autoimmun Rev ; 18(7): 673-678, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31059842

RESUMO

BACKGROUND: Thyrocytes secrete CXC chemokines, particularly (C-X-C motif) ligand (CXCL)8 and CXCL10; its physiopathological significance remains unclear. This study investigates the modulation of the secretion of CXCL8 vs. CXCL10, in human primary cells cultures of thyroid follicular cells (TFC) in Graves' disease (GD), and fibroblasts (OF) or preadipocytes (OP) from Graves' ophthalmopathy (GO). METHODS: Cells were initially incubated with different concentrations of tumor necrosis factor (TNF)α (1, 5, 10 ng/mL). Then, CXCL8 and CXCL10 were measured in the supernatants of TFC, OF or OP cells basally and after 24 h of treatment with interferon (IFN)γ (1000 IU/mL) and/or TNFα (10 ng/mL), in presence/absence of the peroxisome proliferator activated receptor (PPAR)γ agonist pioglitazone (0, 0.1, 1, 5, 10, 20 µM), or the PPARα agonist fenofibrate (5, 10, 50, 100 µM). RESULTS: CXCL8, not CXCL10, was detected in basal conditions in TFC, OF and OP. CXCL8 secretion increased dose-dependently with increasing concentrations of TNFα. CXCL10 secretion was significantly stimulated by IFNγ (P < 0.01) and not by TNFα, whereas CXCL8 was induced by TNFα (P < 0.01), and inhibited by IFNγ (P < 0.01) in TFC, OF and OP. Combining TNFα and IFNγ, the IFNγ-induced CXCL10 secretion was synergistically increased (P < 0.01) while the TNFα-induced CXCL8 secretion (P < 0.01) was reversed in all cell types. Pioglitazone had no significant effect on the secretion of CXCL8 stimulated by TNFα, while inhibited CXCL10. Fenofibrate, in presence of IFNγ plus TNFα, dose-dependently inhibited both CXCL10 and CXCL8 release. CONCLUSION: We first show that TFC, OF, and OP secrete CXCL8 and CXCL10 differentially, sustained by specific proinflammatory cytokines or their combination. This could reflect a different role of the two chemokines in the course of the disease, as CXCL10 could be associated with the initial phase of the disease when IFNγ is preponderant, while CXCL8 could be associated with a later chronic phase of the disease, when TNFα prevails.


Assuntos
Adipócitos/imunologia , Citocinas/imunologia , Fibroblastos/imunologia , Doença de Graves/imunologia , Oftalmopatia de Graves/imunologia , PPAR gama/imunologia , Células Epiteliais da Tireoide/imunologia , Células Cultivadas , Humanos
10.
Cytokine ; 116: 128-133, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30711852

RESUMO

Autoimmune thyroid disease (ATD) is a chronic autoimmune thyroiditis with a complex pathogenesis including environmental factors, genetic background and immune system actions. Despite the large-scale research and discovery of new subpopulations of lymphocytes, cytokines, chemokines and their functions in the human body, the ethiology of ATD in many aspects remains a mystery. This article tries to summarize mostly the immunological aspects of this disease, including the roles of different cells types (dendritic cells, B cells, CD4+ and CD8+ T cells, NK cells and regulatory T cells) and of different cytokines (secreted by Th1/Th2/Th17/Th22 lymphocyte subpopulations and other, including the IL-23 and CXCL10). We describe the role of immunological abnormalities in the ATD pathogenesis and show that for some cells and cytokines their respective roles are not clear, and bi-directional action is possible. Finally, we propose a network of interactions between the immune cells and thyrocytes in the course of ATD.


Assuntos
Citocinas/imunologia , Células Dendríticas/imunologia , Doença de Hashimoto/imunologia , Linfócitos/imunologia , Células Epiteliais da Tireoide/imunologia , Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Citometria de Fluxo , Doença de Hashimoto/patologia , Humanos , Células Matadoras Naturais/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia
11.
Sci Signal ; 11(514)2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29363585

RESUMO

The thyroid-stimulating hormone receptor (TSHR) is a heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptor (GPCR). Autoimmune hyperthyroidism, commonly known as Graves' disease (GD), is caused by stimulating autoantibodies to the TSHR. We previously described TSHR-specific antibodies (TSHR-Abs) in GD that recognize linear epitopes in the cleavage region of the TSHR ectodomain (C-TSHR-Abs) and induce thyroid cell apoptosis instead of stimulating the TSHR. We found that C-TSHR-Abs entered the cell through clathrin-mediated endocytosis but did not trigger endosomal maturation and failed to undergo normal vesicular sorting and trafficking. We found that stimulating TSHR-Abs (S-TSHR-Abs) activated Gαs and, to a lesser extent, Gαq but that C-TSHR-Abs failed to activate any of the G proteins normally activated in response to TSH. Furthermore, specific inhibition of G proteins in the presence of S-TSHR-mAbs or TSH resulted in a similar failure of endosomal maturation as that caused by C-TSHR-mAbs. Hence, whereas S-TSHR-mAbs and TSH contributed to normal vesicular trafficking of TSHR through the activation of major G proteins, the C-TSHR-Abs resulted in GRK2- and ß-arrestin-1-dependent biased signaling, which is interpreted as a danger signal by the cell. Our observations suggest that the binding of antibodies to different TSHR epitopes may decrease cell survival. Antibody-induced cell injury and the response to cell death amplify the loss of self-tolerance, which most likely helps to perpetuate GPCR-mediated autoimmunity.


Assuntos
Anticorpos Monoclonais/farmacologia , Autoanticorpos/imunologia , Autoimunidade/imunologia , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Receptores da Tireotropina/imunologia , Células Epiteliais da Tireoide/citologia , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Endocitose , Proteínas de Ligação ao GTP/metabolismo , Ratos , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo , beta-Arrestinas/metabolismo
12.
Int Immunopharmacol ; 44: 53-60, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28081504

RESUMO

OBJECTIVE: Thyroglobulin (Tg) and thyroid peroxidase (TPO) antibodies (TgAb and TPOAb), which are primarily of the immunoglobulin G (IgG) class, can mediate antibody-dependent cell-mediated cytotoxicity in vitro. However, it is unclear whether any thyrocyte molecules can facilitate the transport and elimination of TgAb and TPOAb. The IgG transport receptor neonatal Fc receptor (FcRn) is a candidate mediator of these processes. In this study, we aimed to evaluate FcRn expression and function in normal and Hashimoto's thyroiditis (HT) thyrocytes. METHODS: FcRn expression in primary thyrocyte cultures (four normal and four HT groups) was examined by polymerase chain reaction (PCR) and Western blotting. Localization of FcRn was demonstrated by immunoelectron microscopy. A double immunofluorescence staining method was adopted to detect FcRn and internalized human TgAb IgG. Stimulation experiments were performed to assess the regulation of FcRn expression by T helper cell 1 (Th1) (IFN-γ and TNF-α) and Th2 cytokines (IL-10 and IL-4). RESULTS: FcRn expression was lower in HT thyrocytes than in normal thyrocytes. FcRn was located in the cytoplasm, membranes, mitochondria and transport vesicles of thyrocytes. Both human IgG and TgAb IgG were internalized by thyrocytes in a pH-dependent manner and co-localized with FcRn in thyrocytes. FcRn expression was downregulated by Th1 and Th2 cytokines in both normal and HT thyrocytes in a dose-dependent manner. CONCLUSIONS: Our results suggest that FcRn may be associated with the transport and metabolism of IgG in thyrocytes and that transport is independent of IgG type. FcRn may be involved in HT pathogenesis.


Assuntos
Doença de Graves/metabolismo , Doença de Hashimoto/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptores Fc/metabolismo , Células Epiteliais da Tireoide/metabolismo , Citotoxicidade Celular Dependente de Anticorpos , Células Cultivadas , Citocinas/metabolismo , Doença de Graves/imunologia , Doença de Hashimoto/imunologia , Células HeLa , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imunoglobulina G/sangue , Iodeto Peroxidase/imunologia , Receptores Fc/genética , Tireoglobulina/imunologia , Células Epiteliais da Tireoide/imunologia
13.
J Clin Endocrinol Metab ; 102(2): 689-697, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27860532

RESUMO

Context: Thyroiditis is one of the most common extrahepatic manifestations of hepatitis C virus (HCV) infection. By binding to surface cell receptor CD81, HCV envelope glycoprotein E2 mediates entry of HCV into cells. Studies have shown that different viral proteins may individually induce host responses to infection. We hypothesized that HCV E2 protein binding to CD81 expressed on thyroid cells activates a cascade of inflammatory responses that can trigger autoimmune thyroiditis in susceptible individuals. Setting: Human thyroid cell lines ML-1 and human thyrocytes in primary cell culture were treated with HCV recombinant E2 protein. The expression of major proinflammatory cytokines was measured at the messenger RNA and protein levels. Next-generation transcriptome analysis was used to identify early changes in gene expression in thyroid cells induced by E2. Results: HCV envelope protein E2 induced strong inflammatory responses in human thyrocytes, resulting in production of interleukin (IL)-8, IL-6, and tumor necrosis factor-α. Furthermore, the E2 protein induced production of several heat shock proteins including HSP60, HSP70p12A, and HSP10, in human primary thyrocytes. In thyroid cell line ML-1, RNA sequencing identified upregulation of molecules involved in innate immune pathways with high levels of proinflammatory cytokines and chemokines and increased expression of costimulatory molecules, specifically CD40, known to be a major thyroid autoimmunity gene. Conclusion: Our data support a key role for HCV envelope protein E2 in triggering thyroid autoimmunity through activation of cytokine pathways by bystander mechanisms.


Assuntos
Autoimunidade/imunologia , Perfilação da Expressão Gênica/métodos , Proteínas de Choque Térmico/metabolismo , Interleucina-8/metabolismo , Células Epiteliais da Tireoide/imunologia , Proteínas do Envelope Viral/imunologia , Linhagem Celular , Células Cultivadas , Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Expressão Gênica , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Interleucina-6/metabolismo , Células Epiteliais da Tireoide/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
14.
Biomed Res Int ; 2016: 6723807, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27274989

RESUMO

Background. mTOR signaling would be a promising target for thyroid cancer therapy. However, in clinical trials, objective response rate with mTOR inhibitor monotherapy in most cancer types was modest. A new focus on development of combinatorial strategies with rapalogs is increasing. Objective. Investigating the combinatorial antitumor effect of rapamycin and ß-elemene in follicular thyroid cancer cells. Methods. MTT assay was used to determine the FTC-133 cell proliferation after culturing with rapamycin and/or ß-elemene. To analyze their combinatorial effect, immunoblotting was performed to analyze the activation status of AKT. Moreover, ß-elemene attenuated rapamycin-induced immunosuppression was tested in mice. Results. Combination of rapamycin and ß-elemene exerted significant synergistic antiproliferative effects in FTC-133 cell lines in vitro, based on inhibiting the AKT feedback activation induced by rapamycin. In vivo, the ß-elemene could attenuate rapamycin-induced immunosuppression via reversing imbalance of Treg/Th17, with the underlying mechanism needed to be declared. Conclusions. We demonstrate that the novel combination of mTOR inhibitor with ß-elemene synergistically attenuates tumor cell growth in follicular thyroid cancer, which requires additional preclinical validation.


Assuntos
Sinergismo Farmacológico , Sesquiterpenos/administração & dosagem , Sirolimo/administração & dosagem , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Terapia de Imunossupressão , Camundongos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Células Epiteliais da Tireoide/efeitos dos fármacos , Células Epiteliais da Tireoide/imunologia , Neoplasias da Glândula Tireoide/imunologia , Neoplasias da Glândula Tireoide/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...