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.
J Toxicol Sci ; 45(4): 219-226, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32238696

RESUMO

Tumor necrosis factor receptor-associated factor 2 (TRAF2) is an essential component of tumor necrosis factor-α (TNF-α) signaling that regulates nuclear factor-κB (NF-κB) and c-Jun N-terminal kinase (JNK) pathways, and compelling evidence has demonstrated that TRAF2 suppresses TNF-α-induced cytotoxicity. On the other hand, it has been reported that oxidative stress-induced cytotoxicity is potentiated by TRAF2, indicating that TRAF2 both positively and negatively regulates stress-induced cytotoxicity in a context-specific manner. However, the causal role of TRAF2 in DNA damage response (DDR) remains to be explored. In this study, we assessed the function of TRAF2 in DDR induced by cisplatin, a representative DNA-damaging agent, and found that TRAF2 exerts pro-apoptotic activity through p53-dependent mechanisms at least in human fibrosarcoma cell line HT1080. TRAF2 deficient cells exhibit significant resistance to cell death induced by cisplatin, accompanied by the reduction of both p53 protein level and caspase-3 activation. Moreover, cisplatin-induced JNK activation was attenuated in TRAF2-deficient cells, and pharmacological inhibition of JNK signaling suppressed p53 stabilization. These results suggest that TRAF2 promotes p53-dependent apoptosis by activating the JNK signaling cascade in HT1080 cells. Thus, our data demonstrate a novel function of TRAF2 in cisplatin-induced DDR as a pro-apoptotic protein.


Assuntos
Antineoplásicos/farmacologia , Apoptose/genética , Cisplatino/farmacologia , Fator 2 Associado a Receptor de TNF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA , Fibrossarcoma/genética , Fibrossarcoma/patologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/genética , Transdução de Sinais/genética , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/genética , Fator de Necrose Tumoral alfa
2.
Cell ; 178(3): 585-599.e15, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31303383

RESUMO

New opportunities are needed to increase immune checkpoint blockade (ICB) benefit. Whereas the interferon (IFN)γ pathway harbors both ICB resistance factors and therapeutic opportunities, this has not been systematically investigated for IFNγ-independent signaling routes. A genome-wide CRISPR/Cas9 screen to sensitize IFNγ receptor-deficient tumor cells to CD8 T cell elimination uncovered several hits mapping to the tumor necrosis factor (TNF) pathway. Clinically, we show that TNF antitumor activity is only limited in tumors at baseline and in ICB non-responders, correlating with its low abundance. Taking advantage of the genetic screen, we demonstrate that ablation of the top hit, TRAF2, lowers the TNF cytotoxicity threshold in tumors by redirecting TNF signaling to favor RIPK1-dependent apoptosis. TRAF2 loss greatly enhanced the therapeutic potential of pharmacologic inhibition of its interaction partner cIAP, another screen hit, thereby cooperating with ICB. Our results suggest that selective reduction of the TNF cytotoxicity threshold increases the susceptibility of tumors to immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoterapia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Interferon gama/metabolismo , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/mortalidade , Neoplasias/terapia , RNA Guia de Cinetoplastídeos/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/genética , Fator de Necrose Tumoral alfa/farmacologia , Receptor de Interferon gama
3.
Circulation ; 136(8): 729-742, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28572508

RESUMO

BACKGROUND: Programmed cell death, including apoptosis, mitochondria-mediated necrosis, and necroptosis, is critically involved in ischemic cardiac injury, pathological cardiac remodeling, and heart failure progression. Whereas apoptosis and mitochondria-mediated necrosis signaling is well established, the regulatory mechanisms of necroptosis and its significance in the pathogenesis of heart failure remain elusive. METHODS: We examined the role of tumor necrosis factor receptor-associated factor 2 (Traf2) in regulating myocardial necroptosis and remodeling using genetic mouse models. We also performed molecular and cellular biology studies to elucidate the mechanisms by which Traf2 regulates necroptosis signaling. RESULTS: We identified a critical role for Traf2 in myocardial survival and homeostasis by suppressing necroptosis. Cardiac-specific deletion of Traf2 in mice triggered necroptotic cardiac cell death, pathological remodeling, and heart failure. Plasma tumor necrosis factor α level was significantly elevated in Traf2-deficient mice, and genetic ablation of TNFR1 largely abrogated pathological cardiac remodeling and dysfunction associated with Traf2 deletion. Mechanistically, Traf2 critically regulates receptor-interacting proteins 1 and 3 and mixed lineage kinase domain-like protein necroptotic signaling with the adaptor protein tumor necrosis factor receptor-associated protein with death domain as an upstream regulator and transforming growth factor ß-activated kinase 1 as a downstream effector. It is important to note that genetic deletion of RIP3 largely rescued the cardiac phenotype triggered by Traf2 deletion, validating a critical role of necroptosis in regulating pathological remodeling and heart failure propensity. CONCLUSIONS: These results identify an important Traf2-mediated, NFκB-independent, prosurvival pathway in the heart by suppressing necroptotic signaling, which may serve as a new therapeutic target for pathological remodeling and heart failure.


Assuntos
Apoptose/fisiologia , Miócitos Cardíacos/metabolismo , Fator 2 Associado a Receptor de TNF/deficiência , Remodelação Ventricular/fisiologia , Animais , Animais Recém-Nascidos , Cardiotônicos/metabolismo , Morte Celular/fisiologia , Células Cultivadas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Necrose/patologia , Necrose/prevenção & controle , Ratos , Ratos Sprague-Dawley , Fator 2 Associado a Receptor de TNF/genética
4.
J Immunol ; 189(2): 1053-61, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22711886

RESUMO

We have previously shown that transgenic (tg) mice expressing in B lymphocytes both BCL-2 and a TNFR-associated factor 2 (TRAF2) mutant lacking the really interesting new gene and zinc finger domains (TRAF2DN) develop small lymphocytic lymphoma and chronic lymphocytic leukemia with high incidence (Zapata et al. 2004. Proc. Nat. Acad. Sci. USA 101: 16600-16605). Further analysis of the expression of TRAF2 and TRAF2DN in purified B cells demonstrated that expression of both endogenous TRAF2 and tg TRAF2DN was negligible in Traf2DN-tg B cells compared with wild-type mice. This was the result of proteasome-dependent degradation, and rendered TRAF2DN B cells as bona fide TRAF2-deficient B cells. Similar to B cells with targeted Traf2 deletion, Traf2DN-tg mice show expanded marginal zone B cell population and have constitutive p100 NF-κB2 processing. Also, TRAF3, X-linked inhibitor of apoptosis, and Bcl-X(L) expression levels were increased, whereas cellular inhibitors of apoptosis 1 and 2 levels were drastically reduced compared with those found in wild-type B cells. Moreover, consistent with previous results, we also show that TRAF2 was required for efficient JNK and ERK activation in response to CD40 engagement. However, TRAF2 was deleterious for BCR-mediated activation of these kinases. In contrast, TRAF2 deficiency had no effect on CD40-mediated p38 MAPK activation but significantly reduced BCR-mediated p38 activation. Finally, we further confirm that TRAF2 was required for CD40-mediated proliferation, but its absence relieved B cells of the need for B cell activating factor for survival. Altogether, our results suggest that TRAF2 deficiency cooperates with BCL-2 in promoting chronic lymphocytic leukemia/small lymphocytic lymphoma in mice, possibly by specifically enforcing marginal zone B cell accumulation, increasing X-linked inhibitor of apoptosis expression, and rendering B cells independent of B cell activating factor for survival.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Predisposição Genética para Doença , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/imunologia , Linfoma Folicular/genética , Linfoma Folicular/imunologia , Fator 2 Associado a Receptor de TNF/deficiência , Animais , Subpopulações de Linfócitos B/patologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Leucemia Linfocítica Crônica de Células B/metabolismo , Linfoma Folicular/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Estrutura Terciária de Proteína/genética , Fator 2 Associado a Receptor de TNF/biossíntese , Fator 2 Associado a Receptor de TNF/genética , Dedos de Zinco/genética
5.
Immunol Cell Biol ; 90(9): 881-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22546736

RESUMO

Highly coordinated expression of inflammatory and anti-inflammatory cytokines is crucial for maintaining homeostasis of the gut that is constantly exposed to large amounts of commensal bacteria. We have previously reported that tumor necrosis factor (TNF) receptor-associated factor (Traf)2(-/-) mice spontaneously develop severe colitis and that the development of colitis largely depends on TNFα-dependent apoptosis of colonic epithelial cells. However, the detailed molecular mechanisms underlying the immunological disorders of Traf2(-/-) mice are not fully understood. Here we show that interleukin (IL)-10-secreting neutrophils accumulated in peripheral blood and bone marrow (BM) cells from Traf2(-/-) mice compared with those from wild-type mice. Treatment of Traf2(-/-) mice with neutralizing antibody against TNFα or crossing Traf2(-/-) mice with Tnfr1(-/-) mice reduced the percentages of IL-10-secreting neutrophils, suggesting that the development of IL-10-secreting neutrophils largely depended on TNFα signals. Moreover, stimulation of BM cells from wild-type mice with lipopolysaccharide and Pam3CS(K)4, a ligand for Toll-like receptor 4 and 2, respectively, induced differentiation of BM cells into IL-10-secreting neutrophils. These results suggest that the development of IL-10-secreting neutrophils is not restricted to Traf2(-/-) mice, but could be generalized to wild-type mice under certain conditions such as inflammation. Finally, combined treatment of Traf2(-/-) mice with neutralizing antibodies against TNFα and IL-10, but not each antibody alone, substantially ameliorated colitis and prolonged survival. Together, abrogation of immunosuppressive conditions mediated by IL-10-secreting neutrophils might be an alternative strategy to treat chronic inflammatory diseases at least under certain conditions.


Assuntos
Interleucina-10/imunologia , Neutrófilos/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Fator 2 Associado a Receptor de TNF/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Colite/imunologia , Colite/metabolismo , Colite/prevenção & controle , Feminino , Citometria de Fluxo , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopeptídeos/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Análise de Sobrevida , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
6.
Exp Cell Res ; 317(13): 1841-50, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21621534

RESUMO

TNFα stimulation triggers both cell death and survival programs. Since dysregulated apoptosis or cell growth can cause inflammatory diseases, cancer, or autoimmune disorders, it is important to understand the molecular mechanism of controlling cell death and survival by TNFR downstream signaling molecules. In this study, we used normal diploid cells, mouse embryonic fibroblasts (MEFs), to mimic the general TNFα-resistant phenomenon seen under physiological conditions.We elucidated the TNFα-induced death signaling complexes in TNF α-resistant WT MEFs and TNFα-sensitive MEFs that were cFLIP-, RelA-, TRAF2- or RIP1-deficient. Consistent with TNFα-mediated killing, we detected TNFα-induced high molecular weight complexes containing caspase-8 and FADD by gel filtration in the deficient MEFs, especially in those devoid of cFLIP. In addition to the presence of caspase-8-FADD in the TNFα-induced-death complex in the deficient MEFs, we also detected an intermediate protein complex containing RIP1, TRAF2 and caspase-8.Moreover, we demonstrated a correlation between TNFα-sensitivity and death-inducing complex ability in two transformed cell lines, E1A- and Ras- transformed MEFs and PDGF-B-transformed NIH-3T3 cells with PDGF-B signaling inhibited by the tyrosine kinase inhibitor STI571. Taken together, our results suggest the involvement of cFLIP-, RelA-, RIP1-, or TRAF2-related mechanisms for preventing FADD-caspase-8 interaction in wild-type MEFs.


Assuntos
Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/deficiência , Caspase 8/metabolismo , Morte Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Proteína de Domínio de Morte Associada a Fas/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Proteínas Ativadoras de GTPase/deficiência , Camundongos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Células NIH 3T3 , Fator 2 Associado a Receptor de TNF/deficiência , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
7.
J Virol ; 84(13): 6605-14, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410275

RESUMO

Epstein-Barr Virus (EBV) latent membrane protein 1 (LMP1) is required for EBV B-lymphocyte transformation, transforms rodent fibroblasts, and can induce lymphoma and epithelial hyperplasia in transgenic mice. Two domains have been identified within the intracellular carboxy terminus that can activate NF-kappaB, C-terminus-activating region 1 (CTAR1) and CTAR2, through interactions with tumor necrosis receptor-associated factors (TRAFs). CTAR1 can activate both the canonical and noncanonical NF-kappaB pathways and has unique effects on cellular gene expression. The epidermal growth factor receptor (EGFR) is highly induced by LMP1-CTAR1 in epithelial cells through activation of a novel NF-kappaB form containing p50 homodimers and Bcl-3. To further understand the regulation of NF-kappaB in CTAR1-induced EGFR expression, we evaluated the ability of CTAR1 to induce EGFR in mouse embryonic fibroblasts (MEFs) defective for different NF-kappaB effectors. CTAR1-mediated EGFR induction required the NF-kappaB-inducing kinase (NIK) but not the IkappaB kinase (IKK) complex components that regulate canonical or noncanonical NF-kappaB pathways. CTAR1-mediated induction of nuclear p50 occurred in IKKbeta-, IKKgamma-, and NIK-defective MEFs, indicating that this induction is not dependent on the canonical or noncanonical NF-kappaB pathways. EGFR and nuclear p50 were expressed at high levels in TRAF2(-/-) fibroblasts and were not induced by CTAR1. In TRAF3(-/-) MEFs, CTAR1 induced nuclear p50 but did not affect basal levels of STAT3 serine phosphorylation or induce EGFR expression. EGFR was induced by LMP1 in TRAF6(-/-) MEFs. These findings suggest that this novel NF-kappaB pathway is differentially regulated by TRAF2 and TRAF3, and that distinct interactions of LMP1 and its effectors regulate LMP1-mediated gene expression.


Assuntos
Receptores ErbB/biossíntese , Herpesvirus Humano 4/fisiologia , Subunidade p50 de NF-kappa B/metabolismo , Proteínas da Matriz Viral/metabolismo , Ligação Viral , Animais , Fibroblastos/virologia , Quinase I-kappa B/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 3 Associado a Receptor de TNF/deficiência , Fator 3 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/deficiência , Fator 6 Associado a Receptor de TNF/metabolismo , Quinase Induzida por NF-kappaB
8.
J Biol Chem ; 284(31): 20531-9, 2009 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-19506082

RESUMO

Inhibitor of apoptosis (IAP) proteins are key regulators of intracellular signaling that interact with tumor necrosis factor (TNF) receptor superfamily members as well as proapoptotic molecules such as Smac/DIABLO and caspases. Whereas the X-linked IAP is an established caspase inhibitor, the protective mechanisms utilized by the cellular IAP (c-IAP) proteins are less clear because c-IAPs bind to but do not inhibit the enzymatic activities of caspases. In this study, c-IAPs are shown to be highly unstable molecules that undergo autoubiquitination. The autoubiquitination of c-IAP1 is blocked upon coexpression with TNF receptor-associated factor (TRAF) 2, and this is achieved by inhibition of the E3 ubiquitin ligase activity intrinsic to the RING of c-IAP1. Consistent with these observations, loss of TRAF2 results in a decrease in c-IAP1 levels. Stabilized c-IAP1 was found to sequester and prevent Smac/DIABLO from antagonizing X-linked IAP and protect against cell death. Therefore, this study describes an intriguing cytoprotective mechanism utilized by c-IAP1 and provides critical insight into how IAP proteins function to alter the apoptotic threshold.


Assuntos
Citoproteção , Proteínas Inibidoras de Apoptose/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Linhagem Celular , Humanos , Proteínas Inibidoras de Apoptose/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Proteínas Mitocondriais/metabolismo , Proteínas Mutantes/metabolismo , Ligação Proteica , Biossíntese de Proteínas , Estabilidade Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/deficiência , Ubiquitinação , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Proteína X Associada a bcl-2/metabolismo
9.
J Immunol ; 179(7): 4645-53, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17878362

RESUMO

Members of the TNFR family play critical roles in the regulation of the immune system. One member of the family critical for efficient activation of T-dependent humoral immune responses is CD40, a cell surface protein expressed by B cells and other APC. The cytoplasmic domain of CD40 interacts with several members of the TNFR-associated factor (TRAF) family, which link CD40 to intracellular signaling pathways. TRAF2 and 6 appear to play particularly important roles in CD40 signaling. Previous studies suggest that the two molecules have certain overlapping roles in signaling, but that unique roles for each molecule also exist. To better define the roles of TRAF2 and TRAF6 in CD40 signaling, we used somatic cell gene targeting to generate TRAF-deficient mouse B cell lines. A20.2J cells deficient in TRAF6 exhibit marked defects in CD40-mediated JNK activation and the up-regulation of CD80. Our previous experiments with TRAF2-deficient B cell lines suggest that TRAF6 and TRAF2 may have redundant roles in CD40-mediated NF-kappaB activation. Consistent with this hypothesis, we found CD40-mediated activation of NF-kappaB intact in TRAF6-deficient cells and defective in cells lacking both TRAF2 and TRAF6. Interestingly, we found that TRAF6 mutants defective in CD40 binding were able to restore CD40-mediated JNK activation and CD80 up-regulation in TRAF6-deficient cells, indicating that TRAF6 may be able to contribute to certain CD40 signals without directly binding CD40.


Assuntos
Antígenos CD40/imunologia , Transdução de Sinais/imunologia , Fator 6 Associado a Receptor de TNF/imunologia , Fator 6 Associado a Receptor de TNF/metabolismo , Animais , Antígeno B7-1/metabolismo , Linhagem Celular , Ativação Enzimática , Vetores Genéticos/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Regulação para Cima
10.
J Immunol ; 178(4): 2272-7, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17277132

RESUMO

Activation of the noncanonical pathway through the interaction of lymphotoxin (LT)-alpha(1)beta(2) and LT-betaR is essential for the development of secondary lymphoid organs including lymph nodes (LN) and Peyer's patches (PP). Although TNFR-associated factor (TRAF) 2 and TRAF5 were identified as signal transducers for the LT-betaR, roles for TRAF2 and TRAF5 in the development of secondary lymphoid organs remain obscure. In this study, we show that PP but not mesenteric LN development is severely impaired in traf2(-/-) and traf2(-/-)traf5(-/-) mice. Development of VCAM-1(+) and ICAM-1(+) mesenchymal cells and expression of CXCL13, a crucial chemokine for the development of PP, are severely impaired in PP anlagen in the intestines of traf2(-/-) mice. Surprisingly, TNF-alpha stimulation potently up-regulates cxcl13 mRNA expression in wild-type murine embryonic fibroblasts, which is impaired in traf2(-/-) and relA(-/-) murine embryonic fibroblasts. Moreover, RelA is recruited to the promoter of cxcl13 gene upon TNF-alpha stimulation and PP development is impaired in TNFR type 1 (tnfr1)(-/-) mice. These results underscore a crucial role for the TNFR1-TRAF2-RelA-dependent canonical pathway in the development of PP through up-regulation of cxcl13 mRNA.


Assuntos
Quimiocinas CXC/imunologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Nódulos Linfáticos Agregados/embriologia , Fator 2 Associado a Receptor de TNF/imunologia , Fator 5 Associado a Receptor de TNF/imunologia , Fator de Transcrição RelA/imunologia , Animais , Quimiocina CXCL13 , Quimiocinas CXC/biossíntese , Fibroblastos/citologia , Fibroblastos/imunologia , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/imunologia , Intestino Delgado/citologia , Intestino Delgado/embriologia , Intestino Delgado/imunologia , Linfonodos/embriologia , Linfonodos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Receptor beta de Linfotoxina/imunologia , Mesoderma/citologia , Mesoderma/imunologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 5 Associado a Receptor de TNF/deficiência , Fator 5 Associado a Receptor de TNF/metabolismo , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Regulação para Cima/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
11.
Eur J Immunol ; 36(9): 2535-43, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16897814

RESUMO

The activation molecule CD40 and the death receptor CD95/Fas play important roles in regulating B cells so that effective antimicrobial immunity occurs without autoimmunity. CD40 signaling increases CD95 expression, sensitizing cells to apoptosis, but sustained CD40 signals rescue B cells from CD95 killing. Here we describe a mechanism of early CD40-mediated rescue from CD95-induced apoptosis in B cells. Maximal rescue was achieved when CD40 signals were given within 1-2 h of initiating CD95 apoptosis. CD40 signaling did not block association of Fas-associated death domain-containing protein with CD95, but decreased CD95-induced activation of caspases 3 and 8. Rapid CD40 rescue did not require NF-kappaB activation and was independent of de novo protein synthesis, but was dependent upon active PI3 K. Signaling via a CD40 mutant that does not bind TNFR-associated factor (TRAF)1, TRAF2, and TRAF3 rescued B cells from CD95-induced apoptosis. TRAF1/2/3-independent rescue was confirmed in B cell lines made deficient in these TRAF molecules by gene targeting. In contrast, CD40 rescue was completely abrogated in TRAF6-deficient B cells, which showed reduced activation of Akt in response to CD40 engagement. These results reveal a new rapid mechanism to balance B cell activation and apoptosis.


Assuntos
Apoptose/imunologia , Linfócitos B/imunologia , Antígenos CD40/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Receptor fas/imunologia , Animais , Linfócitos B/metabolismo , Antígenos CD40/metabolismo , Caspases/imunologia , Linhagem Celular , Ativação Enzimática/imunologia , Immunoblotting , Imunoprecipitação , Ativação Linfocitária/imunologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator 1 Associado a Receptor de TNF/deficiência , Fator 1 Associado a Receptor de TNF/imunologia , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/imunologia , Fator 3 Associado a Receptor de TNF/deficiência , Fator 3 Associado a Receptor de TNF/imunologia , Fator 6 Associado a Receptor de TNF/deficiência , Fator 6 Associado a Receptor de TNF/imunologia , Receptor fas/metabolismo
12.
J Immunol ; 176(9): 5388-400, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16622006

RESUMO

TNFR-associated factor 1 (TRAF1) is unique among the TRAF family, lacking most zinc-binding features, and showing marked up-regulation following activation signals. However, the biological roles that TRAF1 plays in immune cell signaling have been elusive, with many reports assigning contradictory roles to TRAF1. The overlapping binding site for TRAFs 1, 2, and 3 on many TNFR superfamily molecules, together with the early lethality of mice deficient in TRAFs 2 and 3, has complicated the quest for a clear understanding of the functions of TRAF1. Using a new method for gene targeting by homologous recombination in somatic cells, we produced and studied signaling by CD40 and its viral oncogenic mimic, latent membrane protein 1 (LMP1) in mouse B cell lines lacking TRAF1, TRAF2, or both TRAFs. Results indicate that TRAFs 1 and 2 cooperate in CD40-mediated activation of the B cell lines, with a dual deficiency leading to a markedly greater loss of function than that of either TRAF alone. In the absence of TRAF1, an increased amount of TRAF2 was recruited to lipid rafts, and subsequently, more robust degradation of TRAF2 and TRAF3 was induced in response to CD40 signaling. In contrast, LMP1 did not require either TRAFs 1 or 2 to induce activation. Taken together, our findings indicate that TRAF1 and TRAF2 cooperate in CD40 but not LMP1 signaling and suggest that cellular levels of TRAF1 may play an important role in modulating the degradation of TRAF2 and TRAF3 in response to signals from the TNFR superfamily.


Assuntos
Antígenos CD40/metabolismo , Transdução de Sinais , Fator 1 Associado a Receptor de TNF/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linhagem Celular , Ativação Enzimática , Regulação da Expressão Gênica , Imunoglobulinas/biossíntese , Imunoglobulinas/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Fator 1 Associado a Receptor de TNF/deficiência , Fator 1 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/genética
13.
J Biol Chem ; 281(5): 2631-8, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16299380

RESUMO

The endoplasmic reticulum represents the quality control site of the cell for folding and assembly of cargo proteins. A variety of conditions can alter the ability of the endoplasmic reticulum (ER) to properly fold proteins, thus resulting in ER stress. Cells respond to ER stress by activating different signal transduction pathways leading to increased transcription of chaperone genes, decreased protein synthesis, and eventually to apoptosis. In the present paper we analyzed the role that the adaptor protein tumor necrosis factor-receptor associated factor 2 (TRAF2) plays in regulating cellular responses to apoptotic stimuli from the endoplasmic reticulum. Mouse embryonic fibroblasts derived from TRAF2-/- mice were more susceptible to apoptosis induced by ER stress than the wild type counterpart. This increased susceptibility to ER stress-induced apoptosis was because of an increased accumulation of reactive oxygen species following ER stress, and was abolished by the use of antioxidant. In addition, we demonstrated that the NF-kappaB pathway protects cells from ER stress-induced apoptosis, controlling ROS accumulation. Our results underscore the involvement of TRAF2 in regulating ER stress responses and the role of NF-kappaB in protecting cells from ER stress-induced apoptosis.


Assuntos
Apoptose , Retículo Endoplasmático/metabolismo , Estresse Oxidativo , Fator 2 Associado a Receptor de TNF/fisiologia , Animais , Antioxidantes/farmacologia , Fibroblastos/citologia , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator 2 Associado a Receptor de TNF/deficiência , eIF-2 Quinase
14.
Neuroscience ; 131(2): 283-92, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15708473

RESUMO

The major histocompatibility complex (MHC) glycoproteins, MHC1 and MHC2, play a key role in the presentation of antigen and the development of the immune response. In the current study we examined the regulation of the MHC2 in the mouse brain after facial axotomy. The normal facial motor nucleus showed very few slender and elongated MHC2+ cells. Transection of the facial nerve led to a gradual but strong upregulation in the number of MHC2+ cells, beginning at day 2 and reaching a maximum 14 days after axotomy, correlated with the induction of mRNA for tumor necrosis factor (TNF) alpha, interleukin (IL) 1beta and interferon-gamma (IFNgamma) and a peak in neuronal cell death. In almost all cases, MHC2 immunoreactivity was restricted to perivascular macrophages that colocalized with vascular basement membrane laminin and macrophage IBA1-immunoreactivity, with no immunoreactivity on phagocytic microglia, astrocytes or invading T-cells. Heterologous transplantation and systemic injection of endotoxin or IFNgamma did not affect this perivascular MHC2 immunoreactivity, and transgenic deletion of the IL1 receptor type I, or TNF receptor type 1, also had no effect. However, the deletion of IFNgamma receptor subunit 1 caused a significant increase, and that of TNF receptor type 2 a strong reduction in the number of MHC2+ macrophages, pointing to a counter-regulatory role of IFNgamma and TNFalpha in the immune surveillance of the injured nervous system.


Assuntos
Traumatismos do Nervo Facial/metabolismo , Genes MHC da Classe II/fisiologia , Macrófagos/metabolismo , Receptores de Interferon/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Animais , Axotomia/métodos , Nervo Facial/metabolismo , Traumatismos do Nervo Facial/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/genética , Fator 1 Associado a Receptor de TNF/deficiência , Fator 1 Associado a Receptor de TNF/genética , Fator 1 Associado a Receptor de TNF/fisiologia , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/fisiologia , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA