Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 8(11): e74450, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24265670

RESUMO

INTRODUCTION: We developed an experimental autoimmune myositis (EAM) mouse model of polymyositis where we outlined the role of regulatory T (Treg) cells. Rapamycin, this immunosuppressant drug used to prevent rejection in organ transplantation, is known to spare Treg. Our aim was to test the efficacy of rapamycin in vivo in this EAM model and to investigate the effects of the drug on different immune cell sub-populations. METHODS: EAM is induced by 3 injections of myosin emulsified in CFA. Mice received rapamycin during 25 days starting one day before myosin immunization (preventive treatment), or during 10 days following the last myosin immunization (curative treatment). RESULTS: Under preventive or curative treatment, an increase of muscle strength was observed with a parallel decrease of muscle inflammation, both being well correlated (R(2) = -0.645, p<0.0001). Rapamycin induced a general decrease in muscle of CD4 and CD8 T cells in lymphoid tissues, but spared B cells. Among T cells, the frequency of Treg was increased in rapamycin treated mice in draining lymph nodes (16.9 ± 2.2% vs. 9.3 ± 1.4%, p<0.001), which were mostly activated regulatory T cells (CD62L(low)CD44(high): 58.1 ± 5.78% vs. 33.1 ± 7%, treated vs. untreated, p<0.001). In rapamycin treated mice, inhibition of proliferation (Ki-67(+)) is more important in effector T cells compared to Tregs cells (p<0.05). Furthermore, during preventive treatment, rapamycin increased the levels of KLF2 transcript in CD44(low) CD62L(high) naive T cell and in CD62L(low) CD44(high) activated T cell. CONCLUSIONS: Rapamycin showed efficacy both as curative and preventive treatment in our murine model of experimental myositis, in which it induced an increase of muscle strength with a parallel decrease in muscle inflammation. Rapamycin administration was also associated with a decrease in the frequency of effector T cells, an increase in Tregs, and, when administered as preventive treatment, an upregulation of KFL2 in naive and activated T cells.


Assuntos
Imunossupressores/farmacologia , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Sirolimo/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Feminino , Imunossupressores/administração & dosagem , Imunossupressores/efeitos adversos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Contagem de Linfócitos , Linfopenia/induzido quimicamente , Linfopenia/patologia , Camundongos , Força Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Doença Autoimune do Sistema Nervoso Experimental/metabolismo , Doença Autoimune do Sistema Nervoso Experimental/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Sirolimo/administração & dosagem , Sirolimo/efeitos adversos , Linfócitos T Reguladores/metabolismo
2.
Arthritis Rheum ; 60(10): 3118-27, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19790078

RESUMO

OBJECTIVE: Polymyositis and dermatomyositis are chronic inflammatory muscle diseases. Retinoids are compounds that bind to the retinoic acid binding site of retinoic acid receptors and have biologic activities similar to those of vitamin A. Recent studies indicate that retinoids promote Th2 differentiation and suppress Th1 and Th17 differentiation in vitro. The present study was undertaken to examine the effects of a synthetic retinoid, Am80, on experimental autoimmune myositis as well as on Th phenotype development and antibody production. METHODS: Experimental autoimmune myositis was induced in SJL/J mice by immunization with rabbit myosin. Am80 was administered orally once daily. Its effects were evaluated by measurement of the numbers of infiltrating inflammatory cells, production of inflammatory cytokines in muscle, production of Th-specific cytokines by myosin-stimulated splenic T cells, and production of antimyosin antibodies in serum. RESULTS: In mice with experimental autoimmune myositis, orally administered Am80 significantly reduced the number of infiltrating inflammatory cells and the expression of tumor necrosis factor alpha and interleukin-1beta (IL-1beta) in muscle. Moreover, Am80 increased production of interferon-gamma, IL-4, and IL-10, but not IL-17, by myosin-stimulated splenic T cells of mice with experimental autoimmune myositis, suggesting that it could enhance differentiation into Th1 and Th2, but not Th17, in vivo. Am80 also decreased serum levels of IgG2a and IgG2b antimyosin antibodies, but did not affect levels of IgG1 antimyosin antibodies. In addition, it suppressed chemokine expression and activator protein 1 activity in myoblasts in vitro. CONCLUSION: The synthetic retinoid Am80 has an inhibitory effect on experimental autoimmune myositis. It might regulate the development of Th phenotype and antibody production in vivo, in addition to its effects on cytokine and chemokine production.


Assuntos
Anticorpos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Doença Autoimune do Sistema Nervoso Experimental/prevenção & controle , Retinoides/farmacologia , Linfócitos T Auxiliares-Indutores/patologia , Animais , Benzoatos/farmacologia , Modelos Animais de Doenças , Imunoglobulina G/metabolismo , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Miosinas/imunologia , Doença Autoimune do Sistema Nervoso Experimental/tratamento farmacológico , Retinoides/uso terapêutico , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/metabolismo , Tetra-Hidronaftalenos/farmacologia , Células Th1/efeitos dos fármacos , Células Th1/metabolismo , Células Th1/patologia , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Células Th2/patologia , Fator de Transcrição AP-1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
J Neuroimmunol ; 215(1-2): 43-8, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19748134

RESUMO

Glatiramer acetate (GA), a synthetic random amino acid copolymer, poly(Y, E, A, K)n, is widely used for treatment of multiple sclerosis. It inhibits experimental autoimmune encephalomyelitis (EAE) in mice by competition with the antigen and by induction of regulatory T cells. A novel copolymer, poly (F, Y, A, K)n , designated FYAK, was more effective than GA in its immunomodulatory activity in EAE. Here, FYAK and GA were compared in the amelioration of another disease model in mice, experimental autoimmune uveoretinitis (EAU). When tested by co-immunization with an uveitogenic antigen, FYAK was superior to GA in its capacity to inhibit EAU induction, as well as immune processes related to this condition. Further, regulatory T-cell lines specific to FYAK were more immunosuppressive than GA-specific lines in the EAU model. The superiority of FYAK-specific lines was accompanied by higher production of Th2 cytokines. These data thus demonstrate that FYAK, a novel copolymer, is superior to GA in its capacity to inhibit immunopathogenic processes in a non-central nervous system tissue.


Assuntos
Aminoácidos/uso terapêutico , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Doença Autoimune do Sistema Nervoso Experimental/prevenção & controle , Peptídeos/uso terapêutico , Uveíte/imunologia , Uveíte/prevenção & controle , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Feminino , Acetato de Glatiramer , Camundongos , Polímeros/uso terapêutico
4.
Brain ; 131(Pt 5): 1197-208, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18184663

RESUMO

Anti-GQ1b ganglioside antibodies are the serological hallmark of the Miller Fisher syndrome (MFS) variant of the paralytic neuropathy, Guillain-Barré syndrome, and are believed to be the principal pathogenic mediators of the disease. In support of this, we previously showed in an in vitro mouse model of MFS that anti-GQ1b antibodies were able to bind and disrupt presynaptic motor nerve terminals at the neuromuscular junction (NMJ) as one of their target sites, thereby causing muscle paralysis. This injury only occurred through activation of complement, culminating in the formation and deposition of membrane attack complex (MAC, C5b-9) in nerve membranes. Since this step is crucial to the neuropathic process and an important convergence point for antibody and complement mediated membrane injury in general, it forms an attractive pharmacotherapeutic target. Here, we assessed the efficacy of the humanized monoclonal antibody eculizumab, which blocks the formation of human C5a and C5b-9, in preventing the immune-mediated motor neuropathy exemplified in this model. Eculizumab completely prevented electrophysiological and structural lesions at anti-GQ1b antibody pre-incubated NMJs in vitro when using normal human serum (NHS) as a complement source. In a novel in vivo mouse model of MFS generated through intraperitoneal injection of anti-GQ1b antibody and NHS, mice developed respiratory paralysis due to transmission block at diaphragm NMJs, resulting from anti-GQ1b antibody binding and complement activation. Intravenous injection of eculizumab effectively prevented respiratory paralysis and associated functional and morphological hallmarks of terminal motor neuropathy. We show that eculizumab protects against complement-mediated damage in murine MFS, providing the rationale for undertaking clinical trials in this disease and other antibody-mediated neuropathies in which complement activation is believed to be involved.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Síndrome de Miller Fisher/prevenção & controle , Doença Autoimune do Sistema Nervoso Experimental/prevenção & controle , Animais , Anticorpos Monoclonais Humanizados , Ativação do Complemento/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Gangliosídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Síndrome de Miller Fisher/imunologia , Síndrome de Miller Fisher/fisiopatologia , Contração Muscular , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Doença Autoimune do Sistema Nervoso Experimental/fisiopatologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/fisiopatologia , Paralisia Respiratória/imunologia , Paralisia Respiratória/fisiopatologia , Paralisia Respiratória/prevenção & controle , Sinapses/ultraestrutura , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...