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.
Diabetes ; 64(9): 3229-38, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25948683

RESUMO

Because regulatory T-cell (Treg) development can be induced by the same agonist self-antigens that induce negative selection, perturbation of apoptosis will affect both negative selection and Treg development. But how the processes of thymocyte deletion versus Treg differentiation bifurcate and their relative importance for tolerance have not been studied in spontaneous organ-specific autoimmune disease. We addressed these questions by removing a critical mediator of thymocyte deletion, BIM, in the NOD mouse model of autoimmune diabetes. Despite substantial defects in the deletion of autoreactive thymocytes, BIM-deficient NOD (NODBim(-/-)) mice developed less insulitis and were protected from diabetes. BIM deficiency did not impair effector T-cell function; however, NODBim(-/-) mice had increased numbers of Tregs, including those specific for proinsulin, in the thymus and peripheral lymphoid tissues. Increased levels of Nur77, CD5, GITR, and phosphorylated IκB-α in thymocytes from NODBim(-/-) mice suggest that autoreactive cells receiving strong T-cell receptor signals that would normally delete them escape apoptosis and are diverted into the Treg pathway. Paradoxically, in the NOD model, reduced thymic deletion ameliorates autoimmune diabetes by increasing Tregs. Thus, modulating apoptosis may be one of the ways to increase antigen-specific Tregs and prevent autoimmune disease.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Deleção Clonal/genética , Diabetes Mellitus Tipo 1/genética , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Linfócitos T Reguladores/imunologia , Timócitos/imunologia , Animais , Proteínas Reguladoras de Apoptose/imunologia , Proteína 11 Semelhante a Bcl-2 , Antígenos CD5/metabolismo , Deleção Clonal/imunologia , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Proteínas I-kappa B/metabolismo , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Inibidor de NF-kappaB alfa , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/imunologia , Timócitos/metabolismo
2.
J Immunol ; 187(4): 1702-12, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21734073

RESUMO

TNF has been implicated in the pathogenesis of type 1 diabetes. When administered early in life, TNF accelerates and increases diabetes in NOD mice. However, when administered late, TNF decreases diabetes incidence and delays onset. TNFR1-deficient NOD mice were fully protected from diabetes and only showed mild peri-insulitis. To further dissect how TNFR1 deficiency affects type 1 diabetes, these mice were crossed to ß cell-specific, highly diabetogenic TCR transgenic I-A(g7)-restricted NOD4.1 mice and Kd-restricted NOD8.3 mice. TNFR1-deficient NOD4.1 and NOD8.3 mice were protected from diabetes and had significantly less insulitis compared with wild type NOD4.1 and NOD8.3 controls. Diabetic NOD4.1 mice rejected TNFR1-deficient islet grafts as efficiently as control islets, confirming that TNFR1 signaling is not directly required for ß cell destruction. Flow cytometric analysis showed a significant increase in the number of CD4(+)CD25(+)Foxp3(+) T regulatory cells in TNFR1-deficient mice. TNFR1-deficient T regulatory cells were functionally better at suppressing effector cells than were wild type T regulatory cells both in vitro and in vivo. This study suggests that blocking TNF signaling may be beneficial in increasing the function of T regulatory cells and suppression of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Células Secretoras de Insulina/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Diabetes Mellitus Tipo 1/genética , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/genética , Transplante Homólogo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
3.
Apoptosis ; 16(8): 822-30, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21644000

RESUMO

Type 1 diabetes is caused by death of insulin-producing pancreatic beta cells. Beta-cell apoptosis induced by FasL may be important in type 1 diabetes in humans and in the non-obese diabetic (NOD) mouse model. Deficiency of the pro-apoptotic BH3-only molecule Bid protects beta cells from FasL-induced apoptosis in vitro. We aimed to test the requirement for Bid, and the significance of Bid-dependent FasL-induced beta-cell apoptosis in type 1 diabetes. We backcrossed Bid-deficient mice, produced by homologous recombination and thus without transgene overexpression, onto a NOD genetic background. Genome-wide single nucleotide polymorphism analysis demonstrated that diabetes-related genetic regions were NOD genotype. Transferred beta cell antigen-specific CD8+ T cells proliferated normally in the pancreatic lymph nodes of Bid-deficient mice. Moreover, Bid-deficient NOD mice developed type 1 diabetes and insulitis similarly to wild-type NOD mice. Our data indicate that beta-cell apoptosis in type 1 diabetes can proceed without Fas-induced killing mediated by the BH3-only protein Bid.


Assuntos
Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Ilhotas Pancreáticas/imunologia , Animais , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/deficiência , Antígenos CD4/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/fisiologia , Proliferação de Células , Células Cultivadas , Fragmentação do DNA , Diabetes Mellitus Tipo 1/imunologia , Proteína Ligante Fas/farmacologia , Proteína Ligante Fas/fisiologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Sistema Imunitário/citologia , Imunofenotipagem , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Interferon gama/farmacologia , Interferon gama/fisiologia , Interleucina-1beta/farmacologia , Interleucina-1beta/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/fisiologia , Receptor fas/metabolismo
4.
Eur J Immunol ; 41(8): 2238-47, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21674480

RESUMO

The presentation of islet antigens in the pancreatic LNs (PLNs) of mice is a developmentally regulated process. It has been hypothesized that, during physiological tissue remodeling, a wave of neonatal ß-cell apoptosis may initiate diabetes in autoimmune-prone strains of mice. If true, increasing or decreasing physiological ß-cell apoptosis in neonatal NOD mice should alter the time-course of antigen presentation in the PLNs. We used transgenic over-expression of either an anti-apoptotic protein (Bcl-2) or a toxic transgene (rat insulin promoter-Kb) in mouse ß cells to reduce or increase neonatal ß-cell apoptosis, respectively. Neither intervention affected the timing of antigen presentation in the PLNs or the initiation of islet infiltration. This suggests that under physiological conditions and in the absence of inflammation, neonatal ß-cell apoptosis in NOD mice is not the trigger for antigen presentation in the draining LNs.


Assuntos
Apoptose/imunologia , Diabetes Mellitus Tipo 1/imunologia , Inflamação/imunologia , Células Secretoras de Insulina/imunologia , Transferência Adotiva , Animais , Animais Recém-Nascidos , Diabetes Mellitus Tipo 1/metabolismo , Marcação In Situ das Extremidades Cortadas , Inflamação/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Microscopia Confocal , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo
5.
Diabetes ; 59(3): 644-52, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19959756

RESUMO

OBJECTIVE: High concentrations of circulating glucose are believed to contribute to defective insulin secretion and beta-cell function in diabetes and at least some of this effect appears to be caused by glucose-induced beta-cell apoptosis. In mammalian cells, apoptotic cell death is controlled by the interplay of proapoptotic and antiapoptotic members of the Bcl-2 family. We investigated the apoptotic pathway induced in mouse pancreatic islet cells after exposure to high concentrations of the reducing sugars ribose and glucose as a model of beta-cell death due to long-term metabolic stress. RESEARCH DESIGN AND METHODS: Islets isolated from mice lacking molecules implicated in cell death pathways were exposed to high concentrations of glucose or ribose. Apoptosis was measured by analysis of DNA fragmentation and release of mitochondrial cytochrome c. RESULTS: Deficiency of interleukin-1 receptors or Fas did not diminish apoptosis, making involvement of inflammatory cytokine receptor or death receptor signaling in glucose-induced apoptosis unlikely. In contrast, overexpression of the prosurvival protein Bcl-2 or deficiency of the apoptosis initiating BH3-only proteins Bim or Puma, or the downstream apoptosis effector Bax, markedly reduced glucose- or ribose-induced killing of islets. Loss of other BH3-only proteins Bid or Noxa, or the Bax-related effector Bak, had no impact on glucose-induced apoptosis. CONCLUSIONS: These results implicate the Bcl-2 regulated apoptotic pathway in glucose-induced islet cell killing and indicate points in the pathway at which interventional strategies can be designed.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Diabetes Mellitus Tipo 2/patologia , Glucose/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Células Cultivadas , Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Receptores de Interleucina-1/genética , Ribose/toxicidade , Estresse Fisiológico/efeitos dos fármacos , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Proteína Killer-Antagonista Homóloga a bcl-2/química , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/química , Proteína X Associada a bcl-2/genética , Receptor fas/genética
6.
J Virol ; 82(13): 6139-49, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18417562

RESUMO

Infection modulates type 1 diabetes, a common autoimmune disease characterized by the destruction of insulin-producing islet beta cells in the pancreas. Childhood rotavirus infections have been associated with exacerbations in islet autoimmunity. Nonobese diabetic (NOD) mice develop lymphocytic islet infiltration (insulitis) and then clinical diabetes, whereas NOD8.3 TCR mice, transgenic for a T-cell receptor (TCR) specific for an important islet autoantigen, show more rapid diabetes onset. Oral infection of infant NOD mice with the monkey rotavirus strain RRV delays diabetes development. Here, the effect of RRV infection on diabetes development once insulitis is established was determined. NOD and NOD8.3 TCR mice were inoculated with RRV aged > or = 12 and 5 weeks, respectively. Diabetes onset was significantly accelerated in both models (P < 0.024), although RRV infection was asymptomatic and confined to the intestine. The degree of diabetes acceleration was related to the serum antibody titer to RRV. RRV-infected NOD mice showed a possible trend toward increased insulitis development. Infected males showed increased CD8(+) T-cell proportions in islets. Levels of beta-cell major histocompatibility complex class I expression and islet tumor necrosis factor alpha mRNA were elevated in at least one model. NOD mouse exposure to mouse rotavirus in a natural experiment also accelerated diabetes. Thus, rotavirus infection after beta-cell autoimmunity is established affects insulitis and exacerbates diabetes. A possible mechanism involves increased exposure of beta cells to immune recognition and activation of autoreactive T cells by proinflammatory cytokines. The timing of infection relative to mouse age and degree of insulitis determines whether diabetes onset is delayed, unaltered, or accelerated.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Ilhotas Pancreáticas/imunologia , Infecções por Rotavirus/complicações , Fatores Etários , Análise de Variância , Animais , Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Citometria de Fluxo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia
7.
Diabetes ; 57(5): 1284-92, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18252892

RESUMO

OBJECTIVE: Apoptosis of pancreatic beta-cells is critical in both diabetes development and failure of islet transplantation. The role in these processes of pro- and antiapoptotic Bcl-2 family proteins, which regulate apoptosis by controlling mitochondrial integrity, remains poorly understood. We investigated the role of the BH3-only protein Bid and the multi-BH domain proapoptotic Bax and Bak, as well as prosurvival Bcl-2, in beta-cell apoptosis. RESEARCH DESIGN AND METHODS: We isolated islets from mice lacking Bid, Bax, or Bak and those overexpressing Bcl-2 and exposed them to Fas ligand, tumor necrosis factor (TNF)-alpha, and proinflammatory cytokines or cytotoxic stimuli that activate the mitochondrial apoptotic pathway (staurosporine, etoposide, gamma-radiation, tunicamycin, and thapsigargin). Nuclear fragmentation was measured by flow cytometry. RESULTS: Development and function of islets were not affected by loss of Bid, and Bid-deficient islets were as susceptible as wild-type islets to cytotoxic stimuli that cause apoptosis via the mitochondrial pathway. In contrast, Bid-deficient islets and those overexpressing antiapoptotic Bcl-2 were protected from Fas ligand-induced apoptosis. Bid-deficient islets were also resistant to apoptosis induced by TNF-alpha plus cycloheximide and were partially resistant to proinflammatory cytokine-induced death. Loss of the multi-BH domain proapoptotic Bax or Bak protected islets partially from death receptor-induced apoptosis. CONCLUSIONS: These results demonstrate that Bid is essential for death receptor-induced apoptosis of islets, similar to its demonstrated role in hepatocytes. This indicates that blocking Bid activity may be useful for protection of islets from immune-mediated attack and possibly also in other pathological states in which beta-cells are destroyed.


Assuntos
Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Receptores de Morte Celular/fisiologia , Animais , Morte Celular , Citocromos c/análise , Teste de Tolerância a Glucose , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/imunologia , Interferons/farmacologia , Interleucina-1beta/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia
8.
Diabetes ; 55(9): 2412-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936188

RESUMO

Cytotoxic T-cells are the major mediators of beta-cell destruction in type 1 diabetes, but the molecular mechanisms are not definitively established. We have examined the contribution of perforin and Fas ligand to beta-cell destruction using islet-specific CD8(+) T-cells from T-cell receptor transgenic NOD8.3 mice. NOD8.3 T-cells killed Fas-deficient islets in vitro and in vivo. Perforin-deficient NOD8.3 T-cells were able to destroy wild-type but not Fas-deficient islets in vitro. These results imply that NOD8.3 T-cells use both pathways and that Fas is required for beta-cell killing only when perforin is missing. Consistent with this theory, transgenic NOD8.3 mice with beta-cells that do not respond to Fas ligation were not protected from diabetes. We next investigated the mechanism of protection provided by overexpression of suppressor of cytokine signaling-1 (SOCS-1) in beta-cells of NOD8.3 mice. SOCS-1 islets remained intact when grafted into NOD8.3 mice and were less efficiently killed in vitro. However, addition of exogenous peptide rendered SOCS-1 islets susceptible to 8.3 T-cell-mediated lysis. Therefore, NOD8.3 T-cells use both perforin and Fas pathways to kill beta-cells and the surprising blockade of NOD8.3 T-cell-mediated beta-cell death by SOCS-1 overexpression may be due in part to reduced target cell recognition.


Assuntos
Células Secretoras de Insulina/citologia , Glicoproteínas de Membrana/fisiologia , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Linfócitos T Citotóxicos/fisiologia , Receptor fas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linfócitos T CD8-Positivos/fisiologia , Diabetes Mellitus Tipo 1/fisiopatologia , Proteína de Domínio de Morte Associada a Fas , Glucose-6-Fosfatase/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Perforina , Proteínas Citotóxicas Formadoras de Poros , Proteínas/fisiologia , Proteína 1 Supressora da Sinalização de Citocina
9.
J Immunol ; 175(1): 293-301, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15972661

RESUMO

In type 1 diabetes, many effector mechanisms damage the beta cell, a key one being perforin/granzyme B production by CD8(+) T cells. The death receptor pathway has also been implicated in beta cell death, and we have therefore generated NOD mice that express a dominant-negative form of the Fas-associated death domain protein (FADD) adaptor to block death receptor signaling in beta cells. Islets developed normally in these animals, indicating that FADD is not necessary for beta cell development as it is for vasculogenesis. beta cells from the transgenic mice were resistant to killing via the Fas pathway in vitro. In vivo, a reduced incidence of diabetes was found in mice with higher levels of dominant-negative FADD expression. This molecule also blocked signals from the IL-1R in culture, protecting isolated islets from the toxic effects of cytokines and also marginally reducing the levels of Fas up-regulation. These data support a role for death receptors in beta cell destruction in NOD mice, but blocking the perforin/granzyme pathway would also be necessary for dominant-negative FADD to have a beneficial clinical effect.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Diabetes Mellitus Tipo 1/prevenção & controle , Ilhotas Pancreáticas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Proteína Ligante Fas , Proteína de Domínio de Morte Associada a Fas , Feminino , Expressão Gênica , Humanos , Técnicas In Vitro , Interferon gama/farmacologia , Interleucina-1/farmacologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Proteínas Recombinantes/farmacologia
10.
Transplantation ; 77(10): 1610-3, 2004 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15239630

RESUMO

One major limitation in pancreatic islet transplantation is availability of donor tissue. Donor shortage is exacerbated by islet apoptosis from the stresses of islet isolation and transplantation. Furthermore, the side effects of immunosuppressive drugs preclude transplants into patients whose diabetes is controlled by parenteral insulin. We hypothesised that over-expressing anti-apoptotic Bcl-2 or secretion of immunomodulatory CTLA4Ig molecules in islet beta cells would enhance survival of transplanted islets while minimizing systemic side effects. Over-expression of Bcl-2 neither significantly increased preservation of islet cell mass after transplantation into immunocompromised recipients nor decreased cytokine-mediated apoptosis in vitro. Although Bcl-2 over-expression alone was insufficient in protecting islet allografts from rejection, its beneficence was shown by the enhancement of protection when the adaptive immune response was inhibited by locally produced CTLA4Ig. Thus, the combination of anti-apoptotic and immunosuppressive intervention has additive or synergistic efficacy and may reduce the level of systemic immunosuppression or quantity of donor tissue required.


Assuntos
Ciclina D1/farmacologia , Diabetes Mellitus/fisiopatologia , Diabetes Mellitus/cirurgia , Imunoconjugados/metabolismo , Transplante das Ilhotas Pancreáticas , Abatacepte , Animais , Apoptose/efeitos dos fármacos , Ciclina D1/genética , Citoproteção , Diabetes Mellitus/metabolismo , Sobrevivência de Enxerto/efeitos dos fármacos , Imunoconjugados/genética , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos/genética , Transplante Homólogo
11.
J Immunol ; 172(9): 5420-6, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15100283

RESUMO

We have dissected the helper requirements for converting a tolerogenic CD8 T cell response into one capable of causing destruction of the pancreatic islets. Injection of naive OVA-specific CD8 T cells into transgenic mice expressing OVA in the pancreas only resulted in islet destruction when activated CD4 Th cells were coinjected. This requirement for activated CD4 T cell help for induction of primary CD8 T cell-mediated immunity to tissue Ags contrasts recent reports suggesting that help is only important for CTL memory. Our findings show that signaling of CD40 on the dendritic cell presenting to CD8 T cells is important, but not sufficient, for induction of diabetes. Furthermore, once helpers are activated, they need not recognize Ag on the dendritic cells they license. This provides insight into the helper requirements for adoptive transfer immunotherapy of tumors and suggests key points for inhibition of CTL-mediated autoimmunity.


Assuntos
Autoantígenos/fisiologia , Comunicação Celular/imunologia , Citotoxicidade Imunológica , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Ativação Linfocitária/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/genética , Ligante de CD40/genética , Ligante de CD40/fisiologia , Comunicação Celular/genética , Divisão Celular/genética , Divisão Celular/imunologia , Células Cultivadas , Citotoxicidade Imunológica/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/imunologia , Ovalbumina/metabolismo , Linfócitos T Citotóxicos/transplante , Linfócitos T Auxiliares-Indutores/metabolismo
12.
J Immunol ; 172(9): 5714-21, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15100317

RESUMO

In type 1 diabetes, cytokine action on beta cells potentially contributes to beta cell destruction by direct cytotoxicity, inducing Fas expression, and up-regulating class I MHC and chemokine expression to increase immune recognition. To simultaneously block beta cell responsiveness to multiple cytokines, we overexpressed suppressor of cytokine signaling-1 (SOCS-1). This completely prevented progression to diabetes in CD8(+) TCR transgenic nonobese diabetic (NOD) 8.3 mice without affecting pancreas infiltration and partially prevented diabetes in nontransgenic NOD mice. SOCS-1 appeared to protect at least in part by inhibiting TNF- and IFN-gamma-induced Fas expression on beta cells. Fas expression was up-regulated on beta cells in vivo in prediabetic NOD8.3 mice, and this was inhibited by SOCS-1. Additionally, IFN-gamma-induced class I MHC up-regulation and TNF- and IFN-gamma-induced IL-15 expression by beta cells were inhibited by SOCS-1, which correlated with suppressed 8.3 T cell proliferation in vitro. Despite this, 8.3 T cell priming in vivo appeared unaffected. Therefore, blocking beta cell responses to cytokines impairs recognition by CD8(+) T cells and blocks multiple mechanisms of beta cell destruction, but does not prevent T cell priming and recruitment to the islets. Our findings suggest that increasing SOCS-1 expression may be useful as a strategy to block CD8(+) T cell-mediated type 1 diabetes as well as to more generally prevent cytokine-dependent tissue destruction in inflammatory diseases.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Citotoxicidade Imunológica , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/prevenção & controle , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Transferência Adotiva , Animais , Proteínas de Transporte/fisiologia , Morte Celular/genética , Morte Celular/imunologia , Células Cultivadas , Citotoxicidade Imunológica/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Progressão da Doença , Regulação para Baixo/genética , Feminino , Antígenos de Histocompatibilidade Classe I/biossíntese , Insulina/genética , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Interferon gama/farmacologia , Interleucina-15/antagonistas & inibidores , Interleucina-15/biossíntese , Ilhotas Pancreáticas/metabolismo , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Regiões Promotoras Genéticas , Ratos , Receptores de Antígenos de Linfócitos T/genética , Proteínas Repressoras/fisiologia , Baço/citologia , Baço/imunologia , Baço/transplante , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina , Transgenes/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/biossíntese
13.
Diabetes ; 53(1): 113-21, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14693705

RESUMO

Proinflammatory cytokines are believed to be important in pancreatic beta-cell destruction in the development of type 1 diabetes. They act by upregulation of genes including Fas and inducible nitric oxide synthase (iNOS), which have both been shown to lead to beta-cell death in vitro. We used mice deficient in the interleukin (IL)-1 receptor (IL-1R) to assess the contribution of IL-1 to different models of diabetes. IL-1R-deficient islets were protected from the damaging effects of tumor necrosis factor (TNF) and interferon (IFN)-gamma in vitro, and beta-cell expression of iNOS was reduced, suggesting that IL-1 mediates the induction of iNOS by TNF and IFN-gamma. IL-1 action was not required for induction of class I major histocompatibility complex or Fas by TNF and IFN-gamma. IL-1R-deficient nonobese diabetic (NOD) mice developed diabetes significantly slower than wild-type mice. IL-1R deficiency did not affect diabetes in 8.3 TCR transgenic NOD mice but prolonged the time to diabetes in BDC2.5 TCR transgenic NOD mice. We conclude that IL-1R deficiency slows progression to diabetes in NOD mice but on its own does not prevent diabetes.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Receptores de Interleucina-1/deficiência , Receptores de Interleucina-1/imunologia , Animais , Morte Celular , Cruzamentos Genéticos , Citocinas/farmacologia , Diabetes Mellitus Tipo 1/genética , Progressão da Doença , Regulação da Expressão Gênica/imunologia , Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Interleucina-1/biossíntese , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Interleucina-1/genética
14.
Int Immunol ; 15(6): 679-90, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12750352

RESUMO

Fas and TNFRI, two members of the tumor necrosis factor receptor family with an intracellular death domain, each play critical roles in apoptotic death of lymphocytes and certain other cell types. We determined the overlapping functions of Fas and TNFRI by breeding non-obese diabetic (NOD) mutant mice that lacked both receptors. NODlpr mice developed extensive lymphadenopathy, splenomegaly, CD4(-)CD8(-) B220(+) alpha beta TCR(+) T cells and autoimmune kidney disease. This pathology was not modified by concomitant deficiency in TNFRI as was reported for lpr mice on a B6 background. NODlpr mice lacking CD8(+) T cells, because of a null mutation in beta(2)-microglobulin (beta(2)m), also developed a similar disease profile to NODlpr animals, but the CD4(-)CD8(-) B220(+) alpha beta TCR(+) T cells now derived from a CD4(+) T cell lineage. These results demonstrate that, as in the autoimmune-prone MRL stain, the NOD genetic background promotes lupus nephritis-like pathology and extensive lymphadenopathy when lpr is present. Loss of TNFRI does not exacerbate the pathology caused by deficiency in Fas and loss of beta(2)m does not reduce it.


Assuntos
Doenças Autoimunes/genética , Nefropatias/genética , Doenças Linfáticas/genética , Receptores do Fator de Necrose Tumoral/deficiência , Microglobulina beta-2/deficiência , Animais , Doenças Autoimunes/patologia , Proteína Ligante Fas , Feminino , Citometria de Fluxo , Nefropatias/imunologia , Nefropatias/patologia , Doenças Linfáticas/imunologia , Doenças Linfáticas/patologia , Ativação Linfocitária/genética , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA