Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Autoimmun ; 67: 57-64, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26489998

RESUMO

Adoptive transfer of regulatory T (Treg) cells have been employed effectively for suppression of several animal models for autoimmune diseases. In order to employ Treg cell therapy in patients, it is necessary to generate Treg cells from the patient's own cells (autologous) that would be able to suppress effectively the disease in vivo, upon their reintroduction to the patient. Towards this objective, we report in the present study on a protocol for a successful immune-regulation of experimental autoimmune myasthenia gravis (EAMG) by ex vivo--generated autologous Treg cells. For this protocol bone marrow (BM) cells, are first cultured in the presence of GM-CSF, giving rise to a population of CD11c(+)MHCII(+)CD45RA(+)CD8(-) DCs (BMDCs). Splenic CD4(+) T cells are then co-cultured with the differentiated BM cells and expand to 90% of Foxp3(+) Treg cells. In vitro assay exhibits a similar dose dependent manner in the suppression of T effector cells proliferation between Treg cells obtained from either healthy or sick donors. In addition, both Treg cells inhibit similarly the secretion of IFN-γ from activated splenocytes. Administration of 1 × 10(6) ex-vivo generated Treg cells, I.V, to EAMG rats, modulates the disease following a single treatment, given 3 days or 3 weeks after disease induction. Similar disease inhibition was achieved when CD4 cells were taken from either healthy or sick donors. The disease suppression was accompanied by reduced levels of total AChR specific antibodies in the serum. Moreover, due to the polyclonality of the described Treg cell, we have examined whether this treatment approach could be also employed for the treatment of other autoimmune diseases involving Treg cells. Indeed, we demonstrated that the ex-vivo generated autologous Treg cells suppress Adjuvant Arthritis (AA) in rats. This study opens the way for the application of induced autologous Treg cell therapy for myasthenia gravis, as well as for other human autoimmune diseases involving Treg cells.


Assuntos
Terapia de Imunossupressão , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transferência Adotiva , Animais , Comunicação Celular , Movimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Imunofenotipagem , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Fenótipo , Ratos , Receptores Nicotínicos/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
2.
J Autoimmun ; 54: 51-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24970384

RESUMO

Myasthenia Gravis (MG) is an organ-specific autoimmune disease. In high percentage of patients there are autoantibodies to the nicotinic acetylcholine receptor (AChR) that attack AChR on muscle cells at the neuromuscular junction, resulting in muscle weakness. Experimental Autoimmune Myasthenia Gravis (EAMG) is an experimental model disease for MG. EAMG is induced in several animal species by immunization with acetylcholine receptor (AChR), usually isolated from the electric organ of electric fish, which is a rich source for this antigen. Our lab has been involved for several decades in research of AChR and of EAMG. The availability of an experimental autoimmune disease that mimics in many aspects the human disease, provides an excellent model system for elucidating the immunological nature and origin of MG, for studying various existing treatment modalities and for attempting the development of novel treatment approaches. In this review in honor of Michael Sela and Ruth Arnon, we report first on our early pioneering contributions to research on EAMG. These include the induction of EAMG in several animal species, early attempts for antigen-specific treatment for EAMG, elicitation and characterization of monoclonal antibodies and anti-idiotypic antibodies, measuring humoral and cellular AChR-specific immune responses in MG patient and more. In the second part of the review we discuss more recent studies from our lab towards developing and testing novel treatment approaches for myasthenia. These include antigen-dependent treatments aimed at specifically abrogating the humoral and cellular anti-AChR responses, as well as immunomodulatory approaches that could be used either alone, or in conjunction with antigen-specific treatments, or alternatively, serve as steroid-sparing agents.


Assuntos
Anticorpos Anti-Idiotípicos , Anticorpos Monoclonais , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Miastenia Gravis Autoimune Experimental , Animais , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Anti-Idiotípicos/uso terapêutico , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Autoanticorpos/imunologia , Humanos , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Receptores Nicotínicos/imunologia
3.
J Autoimmun ; 36(2): 135-41, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21193288

RESUMO

Suppressive regulatory T cells (Treg) and pathogenic T helper 17 (Th17) cells are two lymphocyte subsets with opposing activities in autoimmune diseases. The proinflammatory cytokine IL-6 is a potent factor in switching immune responses in vivo from the induction of Treg to pathogenic Th17 cells. We studied the Treg and Th17 cell compartments in experimental autoimmune myasthenia gravis (EAMG) and healthy control rats in order to assess whether the equilibrium between Treg and Th17 cells is perturbed in the disease. We found that Th17 cell-related genes are upregulated and Treg-related genes are downregulated in EAMG. The shift in favor of Th17 cells in EAMG could be reversed by antibodies to IL-6. Administration of anti-IL-6 antibodies to myasthenic rats suppressed EAMG when treatment started at the acute or at the chronic phase of disease. Suppression of EAMG by anti-IL-6 antibodies was accompanied by a decrease in the overall rat anti-AChR antibody titer and by a reduced number of B cells as compared with control treatment. Administration of anti-IL-6 antibodies led to down-regulation of several Th17 related genes including IL-17, IL-17R, IL-23R and IL-21 but did not affect the number of Treg cells in the lymph nodes. These data identify IL-6 as an important target for modulation of autoimmune responses.


Assuntos
Interleucina-6/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Miastenia Gravis Autoimune Experimental/genética , Miastenia Gravis Autoimune Experimental/prevenção & controle , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Células Th17/efeitos dos fármacos , Células Th17/metabolismo
4.
J Autoimmun ; 36(1): 16-24, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21035305

RESUMO

The autoimmune regulator (Aire) is involved in the prevention of autoimmunity by promoting thymic expression of tissue restricted antigens which leads to elimination of self-reactive T cells. We found that Aire knockout (KO) mice as well as mouse strains that are susceptible to experimental autoimmune myasthenia gravis (EAMG) have lower thymic expression of acetylcholine receptor (AChR- the main autoantigen in MG), compared to wild type (WT) mice and EAMG-resistant mouse strains, respectively. We demonstrated that Aire KO mice have a significant and reproducible lower frequency of CD4+Foxp3+ cells and a higher expression of Th17 markers in their thymus, compared to wild type (WT) mice. These findings led us to expect that Aire KO mice would display increased susceptibility to EAMG. Surprisingly, when EAMG was induced in young (2 month-old) mice, EAMG was milder in Aire KO than in WT mice for several weeks until the age of about 5 months. However, when EAMG was induced in relatively aged (6 month-old) mice, Aire KO mice presented higher disease severity than WT controls. This age-related change in susceptibility to EAMG correlated with an elevated proportion of Treg cells in the spleens of young but not old KO, compared to WT mice, suggesting a role for peripheral Treg cells in the course of disease. Our observations point to a possible link between Aire and Treg cells and suggest an involvement for both in the pathogenesis of myasthenia.


Assuntos
Miastenia Gravis Autoimune Experimental/imunologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Fatores de Transcrição/imunologia , Fatores Etários , Animais , Separação Celular , Suscetibilidade a Doenças/imunologia , Citometria de Fluxo , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Colinérgicos/imunologia , Receptores Colinérgicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Timo/citologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Proteína AIRE
5.
Front Immunol ; 11: 403, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32256489

RESUMO

Myasthenia gravis (MG) with antibodies to the muscle-specific receptor tyrosine kinase (MuSK) is a distinct sub-group of MG, affecting 5-8% of all MG patients. MuSK, a receptor tyrosine kinase, is expressed at the neuromuscular junctions (NMJs) from the earliest stages of synaptogenesis and plays a crucial role in the development and maintenance of the NMJ. MuSK-MG patients are more severely affected and more refractory to treatments currently used for MG. Most patients require long-term immunosuppression, stressing the need for improved treatments. Ideally, preferred treatments should specifically delete the antigen-specific autoimmune response, without affecting the entire immune system. Mucosal tolerance, induced by oral or nasal administration of an auto-antigen through the mucosal system, resulting in an antigen-specific immunological systemic hyporesponsiveness, might be considered as a treatment of choice for MuSK-MG. In the present study we have characterized several immunological parameters of murine MuSK-EAMG and have employed induction of oral tolerance in mouse MuSK-EAMG, by feeding with a recombinant MuSK protein one week before disease induction. Such a treatment has been shown to attenuate MuSK-EAMG. Both induction and progression of disease were ameliorated following oral treatment with the recombinant MuSK fragment, as indicated by lower clinical scores and lower anti-MuSK antibody titers.


Assuntos
Tolerância Imunológica/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Administração Oral , Animais , Feminino , Camundongos , Receptores Proteína Tirosina Quinases/administração & dosagem
6.
Mol Immunol ; 46(1): 192-201, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18799218

RESUMO

Myasthenia gravis (MG) and experimental autoimmune MG (EAMG) are antibody-mediated autoimmune diseases in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. Previously we have revealed that oral treatment with the less native recombinant fragment of the extracellular domain of the human AChR (Halpha1-205) suppressed ongoing EAMG, whereas the more native recombinant Trx-Halpha1-210 exacerbated EAMG. In this study, we speculated on the role of B-cell epitopes in oral tolerogens for the induction of oral tolerance in EAMG. We developed a B-cell epitope-free AChR fragment (BF-AChR) by removing two major B-cell epitopes (67-76 and 129-145) from Trx-Halpha1-210. BF-AChR exhibited a poor response to EAMG sera and to AChR-specific B- and T-cells while its parent fragment, Trx-Halpha1-210, showed much higher reactivity. Oral administration of BF-AChR ameliorated the symptoms in ongoing myasthenic rats accompanied by a significant decrease in AChR-specific humoral and Th1 cellular responses. The underlying mechanism for BF-AChR-induced oral tolerance was mediated by a shift from Th1 to regulatory T-cell (IL-10(+), CD4(+) TGF-beta(+) or Foxp3(+)) responses. This shift was assessed by changes in the cytokine profile and a deviation in the anti-AChR IgG isotypes from IgG2a/IgG2b to IgG1. Our results suggest that the removal of pathogenic B-cell epitopes from AChR fragments increases tolerogenicity by reducing the activation and proliferation of autoreactive B- and T-cells. Collectively, careful consideration of the immunogenicity of a tolerogen is necessary to induce successful oral tolerance in autoimmune disorders.


Assuntos
Epitopos de Linfócito B/imunologia , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Colinérgicos/imunologia , Proteínas Recombinantes/uso terapêutico , Administração Oral , Animais , Autoanticorpos/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Citocinas/imunologia , Regulação para Baixo/efeitos dos fármacos , Feminino , Imunoglobulina G/imunologia , Imuno-Histoquímica , Mediadores da Inflamação/imunologia , Miastenia Gravis Autoimune Experimental/sangue , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/administração & dosagem , Proteínas Recombinantes/farmacologia , Deleção de Sequência , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia
7.
J Neuroimmunol ; 201-202: 128-35, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18632163

RESUMO

Myasthenia gravis (MG) is frequently treated by corticosteroids such as methylprednisolone. However, continuous treatment with steroids often results in adverse effects. In the present study we evaluated the therapeutic potential of a combination of suboptimal doses of methylprednisolone (Solumedrol) and Pentoxifylline (PTX), a general phosphodiesterase (PDE) inhibitor, in rat experimental autoimmune MG (EAMG). This combined treatment resulted in a pronounced suppressive effect on EAMG and was by far more effective than each of the drugs administered separately at these low doses. The suppressive effect on EAMG was accompanied by decreased humoral and cellular responses to AChR as well as down-regulated mRNA expression levels of Th1 cytokines and IL-10 in lymph node cells and of PDE-4 and cathepsin-l in the muscle. This study demonstrates the potential of PTX as a steroid-sparing agent in the management of myasthenia gravis.


Assuntos
Glucocorticoides/uso terapêutico , Metilprednisolona/uso terapêutico , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Pentoxifilina/uso terapêutico , Inibidores de Fosfodiesterase/uso terapêutico , Animais , Anticorpos/sangue , Peso Corporal/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Miastenia Gravis Autoimune Experimental/sangue , Miastenia Gravis Autoimune Experimental/patologia , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Índice de Gravidade de Doença
8.
J Neuroimmunol ; 194(1-2): 89-96, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18178258

RESUMO

Intravenous immunoglobulin (IVIG) treatment is beneficially used in autoimmune disorders including myasthenia gravis (MG) although its mode of action and active components are still not fully identified. In an attempt to isolate from IVIG a disease-specific suppressive fraction, IVIG was passed on columns of IgG from rats with experimental autoimmune MG (EAMG) or from MG patients. These chromatographies resulted in depletion of the suppressive activity of IVIG on rat EAMG whereas the minute amounts of IgG fractions eluted from the EAMG- or MG-specific columns retained the immunosuppressive activity of IVIG. These results demonstrate that a minor disease-specific immunoglobulin fraction present in IVIG is essential for its suppressive activity.


Assuntos
Anticorpos Anti-Idiotípicos/isolamento & purificação , Imunoglobulinas Intravenosas/uso terapêutico , Imunossupressores/uso terapêutico , Miastenia Gravis Autoimune Experimental/terapia , Animais , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Anti-Idiotípicos/uso terapêutico , Especificidade de Anticorpos , Autoanticorpos/imunologia , Cromatografia de Afinidade , Adjuvante de Freund/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulinas Intravenosas/química , Técnicas de Imunoadsorção , Imunossupressores/isolamento & purificação , Miastenia Gravis/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Ratos , Especificidade da Espécie
9.
Ann N Y Acad Sci ; 1132: 244-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18567875

RESUMO

Intravenous immunoglobulin (IVIG) administration has been beneficially used for the treatment of a variety of autoimmune diseases including myasthenia gravis (MG). We have demonstrated that IVIG administration in experimental autoimmune MG (EAMG) results in suppression of disease that is accompanied by decreased Th1 cell and B cell proliferation. Chromatography of pooled human immunoglobulins (IVIG) on immobilized IgG, isolated from rats with EAMG or from MG patients, results in a depletion of the suppressive activity of the IVIG. Moreover, reconstitution of the activity-depleted IVIG with the eluted minute IVIG fractions that had been adsorbed onto the EAMG- or MG-specific columns recovers the depleted immunosuppressive activity. This study supports the notion that the therapeutic effect of IVIG is mediated by an antigen-specific anti-immunoglobulin (anti-idiotypic) activity that is essential for its suppressive activity.


Assuntos
Especificidade de Anticorpos/imunologia , Imunoglobulinas Intravenosas/imunologia , Imunoglobulinas Intravenosas/uso terapêutico , Imunossupressores/imunologia , Imunossupressores/uso terapêutico , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/terapia , Animais , Humanos , Imunoterapia , Ratos , Receptores Colinérgicos/imunologia
10.
Isr Med Assoc J ; 10(1): 58-60, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18300576

RESUMO

BACKGROUND: [corrected] Intravenous immunoglobulin administration has been beneficially used for the treatment of a variety of autoimmune diseases including myasthenia gravis, although its mode of action and active components have not yet been fully identified. OBJECTIVES: To isolate from IVIg a disease-specific fraction involved in the therapeutic activity in myasthenia and to identify its properties and function. RESULTS: IVIg administration in experimental autoimmune MG results in suppression of disease that is accompanied by decreased Th1 cell and B cell proliferation. Chromatography of IVIg on columns of IgG from rats with EAMG or from MG patients resulted in depletion of the suppressive activity that IVIg has on rat EAMG. Moreover, the minute amounts of IgG fractions eluted from the EAMG or MG-specific columns retained the immunosuppressive activity of IVIg. CONCLUSIONS: Our study supports the notion that the therapeutic effect of IVIg is mediated by a minor disease-specific immunoglobulin fraction that is present in IVIg and is essential for its therapeutic activity.


Assuntos
Imunoglobulinas Intravenosas/farmacologia , Fatores Imunológicos/farmacologia , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Animais , Linfócitos B/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fracionamento Químico , Cromatografia , Humanos , Ratos , Células Th1/efeitos dos fármacos
11.
FASEB J ; 20(2): 374-6, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16365386

RESUMO

Myasthenia gravis (MG) and experimental autoimmune MG (EAMG) are T cell-dependent antibody-mediated autoimmune disorders, in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. DNA microarray analysis revealed increased levels of several phosphodiesterase (PDE) subtypes in lymph node cells (LNC) and muscles of EAMG rats compared with healthy controls. Quantitative real-time PCR analysis indicated that EAMG is characterized by an increase of PDE subtypes 1, 3, 4, and 7 in LNC and of PDE subtypes 2, 3, 4, and 7 in muscles. Pentoxifylline (PTX), a general PDE inhibitor, inhibited the progression of EAMG when treatment started at either the acute or chronic stages of disease. This suppression was associated with down-regulation of humoral and cellular AChR-specific responses, as well as down-regulation of PDE4, TNF-alpha, IL-18, IL-12, and IL-10 in LNC and of PDEs 1, 4, 7, and TNF-alpha in muscles. The expression of Foxp3, a transcription factor essential for CD4+CD25+ regulatory T cell function, was increased in splenocytes although the number of these cells remained unchanged. PTX also reduced the expression of the endopeptidase cathepsin-l, a marker of muscle damage, in EAMG muscles. This study demonstrates the involvement of PDE regulation in EAMG pathogenesis and suggests that PDE inhibitors may be considered for immunotherapy of MG.


Assuntos
Miastenia Gravis/tratamento farmacológico , Miastenia Gravis/enzimologia , Pentoxifilina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Animais , Catepsinas/metabolismo , Linhagem Celular , Citocinas/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Humanos , Diester Fosfórico Hidrolases/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/metabolismo , Linfócitos T Reguladores/metabolismo
12.
Ann N Y Acad Sci ; 1110: 550-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17911471

RESUMO

Intravenous immunoglobulin (IVIG) administration has been beneficially used for the treatment of a variety of autoimmune diseases including myasthenia gravis (MG). We have demonstrated that IVIG administration in experimental autoimmune MG (EAMG) results in suppression of disease that is accompanied by decreased Th1 cell and B cell proliferation. Chromatography of pooled human immunoglobulins (IVIGs) on immobilized IgG, isolated from rats with EAMG, results in a complete depletion of the suppressive activity of the IVIG. Moreover, the eluate from this EAMG-specific antibody column retains the immunosuppressive activity of IVIG. This study supports the notion that the therapeutic effect of IVIGs is mediated by an antigen-specific anti-immunoglobulin (anti-idiotypic) activity that is essential for its suppressive activity.


Assuntos
Imunoglobulina G/isolamento & purificação , Imunoglobulina G/uso terapêutico , Imunoglobulinas Intravenosas/imunologia , Imunoglobulinas Intravenosas/uso terapêutico , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/terapia , Animais , Humanos , Imunoglobulina G/imunologia , Imunoterapia , Miastenia Gravis Autoimune Experimental/patologia , Ratos
13.
Ann N Y Acad Sci ; 1107: 111-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17804538

RESUMO

DNA microarray technology was used to identify new potential drug targets for myasthenia gravis (MG), to delineate genes involved in the pathogenesis of the disease and to possibly target their protein products for immunotherapy. In this study we compared the gene expression in lymph node cells (LNC) and muscles of rats with experimental autoimmune MG (EAMG) to those of control, healthy rats. Of the genes that were found to be deregulated in EAMG, we chose to elaborate on two gene systems: (a) The chemokine IFN-gamma-inducible protein 10 (IP-10, CXCL10), and its receptor (CXCR3) and (b) phosphodiesterases.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Miastenia Gravis/tratamento farmacológico , Miastenia Gravis/genética , Análise de Sequência com Séries de Oligonucleotídeos , Corticosteroides/uso terapêutico , Animais , Quimiocina CXCL10 , Quimiocinas CXC/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Miastenia Gravis/metabolismo , Miastenia Gravis/patologia , Inibidores de Fosfodiesterase/uso terapêutico , Receptores CXCR3 , Receptores de Quimiocinas/genética
14.
J Neuroimmunol ; 176(1-2): 187-97, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16730380

RESUMO

Intravenous immunoglobulin (IVIG) administration has been beneficially used in the treatment of several autoimmune disorders including myasthenia gravis (MG), although its mechanism of action is still not clear. To study the optimal conditions of IVIG treatment and delineate its mechanism of action we established a suitable model in rat experimental autoimmune MG (EAMG). We show that IVIG has a suppressive effect on the clinical symptoms of ongoing EAMG that is associated with decreased AChR-specific cellular and humoral immune reactivity. Costimulatory factors and cytokine profile analyses suggest that IVIG immunomodulation in EAMG involves suppression of B and Th1-type T cell responses with no generation of T-regulatory cells. Our data contribute to the understanding of the immunological mechanisms underlying IVIG treatment in MG and in other autoimmune disorders.


Assuntos
Imunoglobulinas Intravenosas/uso terapêutico , Miastenia Gravis Autoimune Experimental/prevenção & controle , Animais , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Interleucina-10/fisiologia , Interleucina-4/fisiologia , Ativação Linfocitária , Miastenia Gravis Autoimune Experimental/imunologia , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Receptores de Interleucina-2/genética , Fator de Crescimento Transformador beta/genética
15.
Oncotarget ; 7(7): 7550-62, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26771137

RESUMO

Abnormal overexpression of CXCL13 is observed in many inflamed tissues and in particular in autoimmune diseases. Myasthenia gravis (MG) is a neuromuscular disease mainly mediated by anti-acetylcholine receptor autoantibodies. Thymic hyperplasia characterized by ectopic germinal centers (GCs) is a common feature in MG and is correlated with high levels of anti-AChR antibodies. We previously showed that the B-cell chemoattractant, CXCL13 is overexpressed by thymic epithelial cells in MG patients. We hypothesized that abnormal CXCL13 expression by the thymic epithelium triggered B-cell recruitment in MG. We therefore created a novel transgenic (Tg) mouse with a keratin 5 driven CXCL13 expression. The thymus of Tg mice overexpressed CXCL13 but did not trigger B-cell recruitment. However, in inflammatory conditions, induced by Poly(I:C), B cells strongly migrated to the thymus. Tg mice were also more susceptible to experimental autoimmune MG (EAMG) with stronger clinical signs, higher titers of anti-AChR antibodies, increased thymic B cells, and the development of germinal center-like structures. Consequently, this mouse model finally mimics the thymic pathology observed in human MG. Our data also demonstrated that inflammation is mandatory to reveal CXCL13 ability to recruit B cells and to induce tertiary lymphoid organ development.


Assuntos
Linfócitos B/patologia , Quimiocina CXCL13/fisiologia , Inflamação/complicações , Miastenia Gravis Autoimune Experimental/patologia , Hiperplasia do Timo/fisiopatologia , Animais , Linfócitos B/metabolismo , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Citometria de Fluxo , Centro Germinativo/metabolismo , Centro Germinativo/patologia , Humanos , Técnicas Imunoenzimáticas , Inflamação/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miastenia Gravis Autoimune Experimental/etiologia , Miastenia Gravis Autoimune Experimental/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
FASEB J ; 18(13): 1600-2, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15319371

RESUMO

Neuro-immune interactions enable mutual regulation of the nervous and immune systems. To date, evidence exists for manipulations of immune cells by neurotransmitters in the periphery. In this study, we suggest the existence of a pathway by which the brain affects immune cells. The pathway we describe here is mediated by dopamine receptors expressed on activated T cells, termed blasts. Blasts can cross the blood brain barrier regardless of antigen specificity and can therefore encounter neurotransmitters in the brain. We show that blasts have a unique response to dopaminergic activation, which has no counterpart in resting T cells. Dopaminergic activation of blasts induces a Th1 bias in their cytokine profile and causes changes in surface marker expression. We further suggest that these changes can subsequently be transferred to peripheral T cells. We have tested this pathway in two in vivo systems: in rats exogenously administered with L-dopa, and in schizophrenia, which is characterized by a central nervous system-restricted increase in dopamine. In both models, peripheral T cells exhibit similar features to those of dopaminergically activated blasts. The existence of such a pathway by which the brain can regulate immune cells opens a conceptually new direction in neuro-immune interactions.


Assuntos
Dopamina/metabolismo , Ativação Linfocitária , Receptores Dopaminérgicos/metabolismo , Linfócitos T/imunologia , Animais , Fatores Biológicos/metabolismo , Fatores Biológicos/farmacologia , Células Cultivadas , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Levodopa/farmacologia , Quimpirol/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Solubilidade , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Células Th2/metabolismo
17.
FASEB J ; 17(13): 1948-50, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12897067

RESUMO

Recent studies have suggested that the alpha7 nicotinic acetylcholine receptor (alpha7 AChR) may play a role in the pathogenesis of schizophrenia. In search for peripheral biological markers for schizophrenia we have investigated alpha7 mRNA levels in peripheral blood lymphocytes (PBLs) of schizophrenic patients and healthy controls. Peripheral blood samples were collected from medicated and non-medicated (drug naive) schizophrenic patients as well as from healthy (non-mentally ill) smokers and non-smokers. RNA was prepared from isolated lymphocytes. Polymerase chain reaction products specific for human alpha7 AChR were quantified by densitometry using Scion image-analysis (shared NIH software). We observed a significant decrease of alpha7 mRNA levels on PBLs of schizophrenic patients compared with controls. The decrease in alpha7 mRNA levels was not a result of medication management, because non-medicated schizophrenic patients displayed the same level of reduction in alpha7 mRNA as did patients receiving medication. In addition, we exclude the possibility that the observed decrease in alpha7 mRNA levels resulted from nicotine consumption in smoking, because healthy smokers exhibited the same levels of alpha7 mRNA as non-smokers. We propose that alpha7 AChR may be involved in the pathophysiology of the disease and may serve as a reliable peripheral biological marker in schizophrenia.


Assuntos
Receptores Nicotínicos/metabolismo , Esquizofrenia/metabolismo , Adolescente , Adulto , Idoso , Biomarcadores/análise , Feminino , Regulação da Expressão Gênica , Humanos , Linfócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , RNA Mensageiro/metabolismo , Receptores Nicotínicos/genética , Esquizofrenia/genética , Fumar , Receptor Nicotínico de Acetilcolina alfa7
18.
Acta Neuropathol Commun ; 3: 1, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25627031

RESUMO

INTRODUCTION: Anti-acetylcholine receptor (AChR) autoantibodies target muscles in spontaneous human myasthenia gravis (MG) and its induced experimental autoimmune model MG (EAMG). The aim of this study was to identify novel functional mechanisms occurring in the muscle pathology of myasthenia. RESULTS: A transcriptome analysis performed on muscle tissue from MG patients (compared with healthy controls) and from EAMG rats (compared with control rats) revealed a deregulation of genes associated with the Interleukin-6 (IL-6) and Insulin-Like Growth Factor 1 (IGF-1) pathways in both humans and rats. The expression of IL-6 and its receptor IL-6R transcripts was found to be altered in muscles of EAMG rats and mice compared with control animals. In muscle biopsies from MG patients, IL-6 protein level was higher than in control muscles. Using cultures of human muscle cells, we evaluated the effects of anti-AChR antibodies on IL-6 production and on the phosphorylation of Protein Kinase B (PKB/Akt). Most MG sera and some monoclonal anti-AChR antibodies induced a significant increase in IL-6 production by human muscle cells. Furthermore, Akt phosphorylation in response to insulin was decreased in the presence of monoclonal anti-AChR antibodies. CONCLUSIONS: Anti-AChR antibodies alter IL-6 production by muscle cells, suggesting a putative novel functional mechanism of action for the anti-AChR antibodies. IL-6 is a myokine with known effects on signaling pathways such as Akt/mTOR (mammalian Target of Rapamycin). Since Akt plays a key role in multiple cellular processes, the reduced phosphorylation of Akt by the anti-AChR antibodies may have a significant impact on the muscle fatigability observed in MG patients.


Assuntos
Interleucina-6/metabolismo , Músculo Esquelético/metabolismo , Miastenia Gravis/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adolescente , Adulto , Animais , Anticorpos/farmacologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Criança , Modelos Animais de Doenças , Feminino , Adjuvante de Freund/toxicidade , Perfilação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Interleucina-6/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Miastenia Gravis/etiologia , Miastenia Gravis/genética , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Fosforilação , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Receptores de Interleucina-6/metabolismo , Adulto Jovem
19.
J Interferon Cytokine Res ; 22(3): 321-8, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12034039

RESUMO

Interleukin-18 binding protein (IL-18BP) is a constitutively expressed and secreted protein that lacks a transmembrane domain. IL-18BP binds specifically to mature IL-18 and inhibits its activity. To study the immunomodulating role of IL-18BP in models of autoimmune diseases in rats, we cloned and characterized rat IL-18BP. Rat IL-18BP has 193 amino acid residues and is highly homologous to human and mouse IL-18BP. Recombinant rat IL-18BP binds to rat IL-18, reacts with antibodies to human or mouse IL-18BP, and inhibits IL-18-dependent interferon-gamma (IFN-gamma) production in vitro. Thus, rat IL-18BP can be employed to antagonize the proinflammatory responses induced by endogenous IL-18 in rat models of autoimmune diseases.


Assuntos
Glicoproteínas/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Clonagem Molecular , Relação Dose-Resposta a Droga , Escherichia coli/genética , Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Interferon gama/biossíntese , Interferon gama/efeitos dos fármacos , Interleucina-12/farmacologia , Interleucina-18/metabolismo , Dados de Sequência Molecular , Ratos , Ratos Endogâmicos Lew , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Homologia de Sequência de Aminoácidos
20.
J Neuroimmunol ; 140(1-2): 153-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12864983

RESUMO

The acetylcholine receptor (AChR) is the major autoantigen in the antibody-mediated disease myasthenia gravis (MG) and its animal model experimental autoimmune myasthenia gravis (EAMG). This study demonstrates that rats immunized with a recombinant fragment corresponding to the normally exposed extracellular region of the rat AChR alpha-subunit first develop antibodies to the injected extracellular portion only, but later develop antibodies to intracellular cytoplasmic epitopes of AChR. The presence of autoantibodies to intracellular epitopes seems to be correlated with development of clinical signs of disease. We propose that a similar process of epitope spreading may take place in the natural course of myasthenia.


Assuntos
Autoantígenos/imunologia , Citoplasma/imunologia , Tolerância Imunológica , Epitopos Imunodominantes/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Colinérgicos/imunologia , Animais , Autoanticorpos/biossíntese , Autoantígenos/administração & dosagem , Autoantígenos/genética , Citoplasma/genética , Espaço Extracelular/genética , Espaço Extracelular/imunologia , Feminino , Humanos , Tolerância Imunológica/genética , Epitopos Imunodominantes/administração & dosagem , Epitopos Imunodominantes/genética , Injeções Subcutâneas , Miastenia Gravis Autoimune Experimental/genética , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Subunidades Proteicas/administração & dosagem , Subunidades Proteicas/genética , Subunidades Proteicas/imunologia , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/administração & dosagem , Receptores Colinérgicos/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Torpedo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA