Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 206(10): 2441-2452, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33941658

RESUMO

Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1ß contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1ß secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1ß-dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1ß secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1ß production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1ß production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1ß-dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R-deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1ß secretion outside the gut.


Assuntos
Infecções por Bacteroides/imunologia , Bacteroides fragilis/imunologia , Fibras na Dieta/efeitos adversos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Peritonite/imunologia , Animais , Infecções por Bacteroides/microbiologia , Dieta , Fibras na Dieta/administração & dosagem , Modelos Animais de Doenças , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Peritonite/microbiologia , Receptores de Interleucina-1/deficiência , Receptores de Interleucina-1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
2.
Clin Transl Immunology ; 5(5): e82, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27350881

RESUMO

It is now convincingly clear that diet is one of the most influential lifestyle factors contributing to the rise of inflammatory diseases and autoimmunity in both developed and developing countries. In addition, the modern 'Western diet' has changed in recent years with increased caloric intake, and changes in the relative amounts of dietary components, including lower fibre and higher levels of fat and poor quality of carbohydrates. Diet shapes large-bowel microbial ecology, and this may be highly relevant to human diseases, as changes in the gut microbiota composition are associated with many inflammatory diseases. Recent studies have demonstrated a remarkable role for diet, the gut microbiota and their metabolites-the short-chain fatty acids (SCFAs)-in the pathogenesis of several inflammatory diseases, such as asthma, arthritis, inflammatory bowel disease, colon cancer and wound-healing. This review summarizes how diet, microbiota and gut microbial metabolites (particularly SCFAs) can modulate the progression of inflammatory diseases and autoimmunity, and reveal the molecular mechanisms (metabolite-sensing G protein-coupled receptor (GPCRs) and inhibition of histone deacetylases (HDACs)). Therefore, considerable benefit could be achieved simply through the use of diet, probiotics and metabolites for the prevention and treatment of inflammatory diseases and autoimmunity.

3.
Eur J Immunol ; 44(4): 983-93, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24435807

RESUMO

Targeting the BAFF/APRIL system has shown to be effective in preventing T-cell dependent autoimmune disease in the NOD mouse, a spontaneous model of type 1 diabetes. In this study we generated BAFF-deficient NOD mice to examine how BAFF availability would influence T-cell responses in vivo and the development of spontaneous diabetes. BAFF-deficient NOD mice which lack mature B cells, were protected from diabetes and showed delayed rejection of an allogeneic islet graft. Diabetes protection correlated with a failure to expand pathogenic IGRP-reactive CD8(+) T cells, which were maintained in the periphery at correspondingly low levels. Adoptive transfer of IGRP-reactive CD8(+) T cells with B cells into BAFF-deficient NOD mice enhanced IGRP-reactive CD8(+) T-cell expansion. Furthermore, when provoked with cyclophosphamide, or transferred to a secondary lymphopenic host, the latent pool of self-reactive T cells resident in BAFF-deficient NOD mice could elicit beta cell destruction. We conclude that lack of BAFF prevents the procurement of B-cell-dependent help necessary for the emergence of destructive diabetes. Indeed, treatment of NOD mice with the BAFF-blocking compound, BR3-Fc, resulted in a delayed onset and reduced incidence of diabetes.


Assuntos
Autoimunidade/imunologia , Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Autoimunidade/genética , Fator Ativador de Células B/genética , Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Citometria de Fluxo , Glucose-6-Fosfatase/imunologia , Glucose-6-Fosfatase/metabolismo , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/genética , Sobrevivência de Enxerto/imunologia , Imunofenotipagem , Transplante das Ilhotas Pancreáticas/métodos , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Linfócitos T/metabolismo , Fatores de Tempo
4.
J Immunol ; 191(1): 97-109, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23740954

RESUMO

Marginal zone (MZ) B cells are an innate-like population that oscillates between MZ and follicular areas of the splenic white pulp. Differentiation of B cells into the MZ subset is governed by BCR signal strength and specificity, NF-κB activation through the B cell-activating factor belonging to the TNF family (BAFF) receptor, Notch2 signaling, and migration signals mediated by chemokine, integrin, and sphingosine-1-phosphate receptors. An imbalance in splenic B cell development resulting in expansion of the MZ subset has been associated with autoimmune pathogenesis in various murine models. One example is the NOD inbred mouse strain, in which MZ B cell expansion has been linked to development of type 1 diabetes and Sjögren's syndrome. However, the cause of MZ B cell expansion in this strain remains poorly understood. We have determined that increased MZ B cell development in NOD mice is independent of T cell autoimmunity, BCR specificity, BCR signal strength, and increased exposure to BAFF. Rather, mixed bone marrow chimeras showed that the factor(s) responsible for expansion of the NOD MZ subset is B cell intrinsic. Analysis of microarray expression data indicated that NOD MZ and precursor transitional 2-MZ subsets were particularly dysregulated for genes controlling cellular trafficking, including Apoe, Ccbp2, Cxcr7, Lgals1, Pla2g7, Rgs13, S1pr3, Spn, Bid, Cd55, Prf1, and Tlr3. Furthermore, these B cell subsets exhibited an increased steady state dwell time within splenic MZ areas. Our data therefore reveal that precursors of mature B cells in NOD mice exhibit an altered migration set point, allowing increased occupation of the MZ, a niche favoring MZ B cell differentiation.


Assuntos
Fator Ativador de Células B/fisiologia , Subpopulações de Linfócitos B/imunologia , Diferenciação Celular/imunologia , Receptor Notch2/fisiologia , Receptores de Antígenos de Linfócitos B/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Baço/imunologia , Animais , Fator Ativador de Células B/deficiência , Receptor do Fator Ativador de Células B/fisiologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/patologia , Diferenciação Celular/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Pró-Proteína Convertases/fisiologia , Serina Endopeptidases/fisiologia , Baço/metabolismo , Baço/patologia
5.
Diabetes ; 61(11): 2893-905, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22829452

RESUMO

For autoimmune conditions like type 1 diabetes to progress, self-reactive CD8⁺ T cells would need to interact with peptide-antigen cross-presented on the surface of antigen-presenting cells in a major histocompatibility complex (MHC) class I-restricted fashion. However, the mechanisms by which autoantigen is cross-presented remain to be identified. In this study, we show cross-presentation of islet-derived autoantigens by B cells. B cells engage self-reactive CD8⁺ T cells in the pancreatic lymph node, driving their proliferative expansion and differentiation into granzyme B⁺interferon-γ⁺lysosomal-associated membrane protein 1⁺ effector cells. B-cell cross-presentation of insulin required proteolytic cleavage and endosomal localization and was sensitive to inhibitors of protein trafficking. Absent B-cell MHC class I, or B-cell receptor restriction to an irrelevant specificity, blunted the expansion of self-reactive CD8⁺ T cells, suggesting B-cell antigen capture and presentation are critical in vivo events for CD8 activation. Indeed, the singular loss of B-cell MHC class I subverted the conversion to clinical diabetes in NOD mice, despite the presence of a pool of activated, and B cell-dependent, interleukin-21-expressing Vß4⁺CD4⁺ T cells. Thus, B cells govern the transition from clinically silent insulitis to frank diabetes by cross-presenting autoantigen to self-reactive CD8⁺ T cells.


Assuntos
Apresentação de Antígeno , Células Apresentadoras de Antígenos/imunologia , Autoantígenos/efeitos adversos , Linfócitos B/imunologia , Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/patologia , Autoantígenos/metabolismo , Linfócitos B/metabolismo , Linfócitos B/patologia , Transplante de Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Células Cultivadas , Quimera , Técnicas de Cocultura , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Progressão da Doença , Feminino , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Pâncreas/imunologia , Pâncreas/metabolismo , Pâncreas/patologia , Estado Pré-Diabético/imunologia , Estado Pré-Diabético/metabolismo , Estado Pré-Diabético/patologia
6.
Diabetes ; 58(7): 1568-77, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19336675

RESUMO

OBJECTIVE: Tumor necrosis factor ligand family members B-cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) can exert powerful effects on B-cell activation and development, type 1 T-helper cell (Th1) immune responses, and autoimmunity. We examined the effect of blocking BAFF and APRIL on the development of autoimmune diabetes. RESEARCH DESIGN AND METHODS: Female NOD mice were administered B-cell maturation antigen (BCMA)-Fc from 9 to 15 weeks of age. Diabetes incidence, islet pathology, and T- and B-cell populations were examined. RESULTS: BCMA-Fc treatment reduced the severity of insulitis and prevented diabetes development in NOD mice. BCMA-Fc-treated mice showed reduced follicular, marginal-zone, and T2MZ B-cells. B-cell reduction was accompanied by decreased frequencies of pathogenic CD4(+)CD40(+) T-cells and reduced Th1 cytokines IL-7, IL-15, and IL-17. Thus, T-cell activation was blunted with reduced B-cells. However, BCMA-Fc-treated mice still harbored detectable diabetogenic T-cells, suggesting that regulatory mechanisms contributed to diabetes prevention. Indeed, BCMA-Fc-treated mice accumulated increased CD4(+)CD25(+) regulatory T-cells (Tregs) with age. CD4(+)CD25(+) cells were essential for maintaining euglycemia because their depletion abrogated BCMA-Fc-mediated protection. BCMA-Fc did not directly affect Treg homeostasis given that CD4(+)CD25(+)Foxp3(+) T-cells did not express TACI or BR3 receptors and that CD4(+)CD25(+)Foxp3(+) T-cell frequencies were equivalent in wild-type, BAFF(-/-), TACI(-/-), BCMA(-/-), and BR3(-/-) mice. Rather, B-cell depletion resulted in CD4(+)CD25(+) T-cell-mediated protection from diabetes because anti-CD25 monoclonal antibody treatment precipitated diabetes in both diabetes-resistant NOD.microMT(-/-) and BCMA-Fc-treated mice. CONCLUSIONS: BAFF/APRIL blockade prevents diabetes. BCMA-Fc reduces B-cells, subsequently blunting autoimmune activity and allowing endogenous regulatory mechanisms to preserve a prehyperglycemic state.


Assuntos
Autoimunidade , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Subunidade alfa de Receptor de Interleucina-2/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD/imunologia , Antígeno de Maturação de Linfócitos B/farmacologia , Feminino , Interleucina-15/imunologia , Interleucina-17/imunologia , Interleucina-7/imunologia , Ativação Linfocitária , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos NOD
7.
J Immunol ; 182(2): 793-801, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19124722

RESUMO

The cytokine B cell activation factor of the TNF family (BAFF) is considered to perform a proinflammatory function. This paradigm is particularly true for B cell-dependent immune responses; however the exact role for BAFF in regulating T cell immunity is ill-defined. To directly assess the effect of BAFF upon T cells, we analyzed T cell-dependent immune responses in BAFF-transgenic (Tg) mice. We found that T cell responses in BAFF-Tg mice are profoundly compromised, as indicated by their acceptance of islet allografts and delayed skin graft rejection. However, purified BAFF-Tg effector T cells could reject islet allografts with a normal kinetic, suggesting that the altered response did not relate to a defect in T cell function per se. Rather, we found that BAFF-Tg mice harbored an increased number of peripheral CD4+Foxp3+ T cells. A large proportion of the BAFF-expanded CD4+CD25+Foxp3+ regulatory T cells (Tregs) were CD62LlowCD103high and ICAM-1high, a phenotype consistent with an ability to home to inflammatory sites and prevent T cell effector responses. Indeed, depletion of the endogenous BAFF-Tg Tregs allowed allograft rejection to proceed, demonstrating that the increased Tregs were responsible for preventing alloimmunity. The ability of BAFF to promote Treg expansion was not T cell intrinsic, as Tregs did not express high levels of BAFF receptor 3, nor did excessive BAFF trigger NF-kappaB2 processing in Tregs. In contrast, we found that BAFF engendered Treg expansion through an indirect, B cell-dependent mechanism. Thus, under certain conditions, BAFF can play a surprising anti-inflammatory role in T cell biology by promoting the expansion of Treg cells.


Assuntos
Fator Ativador de Células B/genética , Proliferação de Células , Fatores de Transcrição Forkhead/biossíntese , Rejeição de Enxerto/prevenção & controle , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/genética , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transplante de Pele/imunologia , Tolerância ao Transplante/genética
8.
J Autoimmun ; 31(3): 301-5, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18809297

RESUMO

Type I diabetes mellitus is an autoimmune disease mediated by a selective immune-mediated destruction of the insulin containing beta cells within the pancreatic islets of Langerhans. T cells reactive to beta cell-derived antigens are critical for the pathogenesis of type I diabetes, indeed treatments that target T cells are currently in clinical trials. CD8+ T cells may play a particularly crucial role in the onset of hyperglycaemia, as they can mediate beta cell destruction late in the pathogenesis of diabetes. However, the precise steps by which beta cell-reactive CD8+ T cells are activated are poorly understood. In this review we speculate on the possibility that B cells are essential for the activation and expansion of pathogenic CD8+ T cells that cause final beta cell destruction. We also discuss the involvement of different B cell subsets in the aetiology of diabetes.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Comunicação Celular/imunologia , Diabetes Mellitus Tipo 1/imunologia , Animais , Subpopulações de Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo
9.
Diabetes ; 55(9): 2491-501, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936197

RESUMO

Apoptotic beta-cell death is central to the pathogenesis of type 1 diabetes and may be important in islet graft rejection. Despite this, genetic control of beta-cell apoptosis is only poorly understood. We report that inhibition of gene transcription sensitized beta-cells to tumor necrosis factor (TNF)-alpha-induced apoptosis, indicating the presence of a regulated antiapoptotic response. Using oligonucleotide microarrays and real-time PCR, we identified TNFAIP3/A20 as the most highly regulated antiapoptotic gene expressed in cytokine-stimulated human and mouse islets. Cytokine induction of A20 mRNA in primary islets and insulinoma cells was rapid and observed within 1 h, consistent with A20 being an immediate early response gene in beta-cells. Regulation of A20 was nuclear factor-kappaB (NF-kappaB)-dependent, two NF-kappaB sites within the A20 promoter were found to be necessary and sufficient for A20 expression in beta-cells. Activation of NF-kappaB by TNF receptor-associated factor (TRAF) 2, TRAF6, NF-kappaB-inducing kinase, or protein kinase D, which transduce signals downstream of Toll-like receptors, TNF receptors, and free radicals, respectively, were all potent activators of the A20 promoter. Moreover, A20 expression was induced in transplanted islets in vivo. Finally, A20 expression was sufficient to protect beta-cells from TNF-induced apoptosis. These data demonstrate that A20 is the cardinal antiapoptotic gene in beta-cells. Further, A20 expression is NF-kappaB dependent, thus linking islet proinflammatory gene responses with protection from apoptosis.


Assuntos
Apoptose , Células Secretoras de Insulina/citologia , NF-kappa B/fisiologia , Proteínas/fisiologia , Animais , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Cisteína Endopeptidases , Proteínas de Ligação a DNA , Perfilação da Expressão Gênica , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1 , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Proteínas Nucleares , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/farmacologia , Fator 6 Associado a Receptor de TNF/farmacologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Quinase Induzida por NF-kappaB
10.
Cytokine ; 30(6): 359-65, 2005 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-15935956

RESUMO

There is evidence that severe dengue disease is associated with alterations of the microvascular endothelium. We examined the hypothesis that activation and damage of microvascular endothelial cells (EC) could be induced by inflammatory mediators present in dengue patient's sera. We cultured human microvascular EC (HMEC-1) in vitro with sera from patients with acute dengue infection. Sera from patients with acute dengue induced an increase in ICAM-1 expression on HMEC-1. This effect was greater with samples from the acute febrile phase than with samples from the convalescent phase of the disease. Acute dengue sera had elevated levels of TNF-alpha and the endothelial activating effect of acute dengue sera was inhibited up to 80% by pre-treatment with monoclonal antibodies against TNF-alpha. Furthermore, acute dengue sera induced apoptosis in HMEC-1. These findings support the pathophysiologic significance of microvascular EC and serum inflammatory mediators in dengue.


Assuntos
Apoptose , Dengue/metabolismo , Células Endoteliais/virologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Inflamação , Fator de Necrose Tumoral alfa/fisiologia , Sobrevivência Celular , Células Cultivadas , Dengue/imunologia , Endotélio Vascular/virologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Microcirculação , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
11.
Cytokine ; 22(5): 142-8, 2003 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-12842762

RESUMO

Interleukin-18 (IL-18) is a newly identified cytokine with proinflammatory activity. Numerous studies have shown that proinflammatory cytokines may regulate endothelial cells (EC) apoptosis mediated by members of the tumor necrosis factor (TNF) family, such as TNF-alpha and Fas. In this study we hypothesized that IL-18 may regulate the susceptibility of liver endothelial cells (LEC) to apoptosis induced by TNF and Fas. IL-18 increased the susceptibility of LEC to undergo apoptosis mediated by TNF but not by Fas. Since TNF-induced apoptosis is mediated by the type I TNF receptor (TNFRI), we investigated up-regulation of this receptor in IL-18-treated LEC. IL-18 induced up-regulation of the TNFRI on the surface of LEC. Partial blocking of LEC apoptosis induced by IL-18 and TNF was observed when the cells were pretreated with the broad-spectrum inhibitor of caspases z-VAD-fmk, suggesting involvement of the caspase pathway in apoptosis induced by these cytokines in these cells. Our results show that IL-18 differentially regulates apoptosis mediated by the death-inducing factors, TNF and Fas. To our knowledge, this is the first report that IL-18 may regulate endothelial cell apoptosis mediated by TNF. These results may have clinical implications in those clinical hepatic conditions associated with high levels of IL-18 and TNF.


Assuntos
Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Interleucina-18/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Receptor fas/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Cricetinae , Células Endoteliais/citologia , Citometria de Fluxo , Fluorescência , Expressão Gênica , Camundongos , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator de Necrose Tumoral alfa/genética , Receptor fas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA