Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 670338, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745083

RESUMO

Proteins controlling mitochondrial fission have been recognized as essential regulators of mitochondrial functions, mitochondrial quality control and cell apoptosis. In the present study, we identified the critical B cell survival regulator TRAF3 as a novel binding partner of the key mitochondrial fission factor, MFF, in B lymphocytes. Elicited by our unexpected finding that the majority of cytoplasmic TRAF3 proteins were localized at the mitochondria in resting splenic B cells after ex vivo culture for 2 days, we found that TRAF3 specifically interacted with MFF as demonstrated by co-immunoprecipitation and GST pull-down assays. We further found that in the absence of stimulation, increased protein levels of mitochondrial TRAF3 were associated with altered mitochondrial morphology, decreased mitochondrial respiration, increased mitochondrial ROS production and membrane permeabilization, which eventually culminated in mitochondria-dependent apoptosis in resting B cells. Loss of TRAF3 had the opposite effects on the morphology and function of mitochondria as well as mitochondria-dependent apoptosis in resting B cells. Interestingly, co-expression of TRAF3 and MFF resulted in decreased phosphorylation and ubiquitination of MFF as well as decreased ubiquitination of TRAF3. Moreover, lentivirus-mediated overexpression of MFF restored mitochondria-dependent apoptosis in TRAF3-deficient malignant B cells. Taken together, our findings provide novel insights into the apoptosis-inducing mechanisms of TRAF3 in B cells: as a result of survival factor deprivation or under other types of stress, TRAF3 is mobilized to the mitochondria through its interaction with MFF, where it triggers mitochondria-dependent apoptosis. This new role of TRAF3 in controlling mitochondrial homeostasis might have key implications in TRAF3-mediated regulation of B cell transformation in different cellular contexts. Our findings also suggest that mitochondrial fission is an actionable therapeutic target in human B cell malignancies, including those with TRAF3 deletion or relevant mutations.


Assuntos
Linfócitos B/fisiologia , Dinâmica Mitocondrial/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Respiração Celular , Sobrevivência Celular , Dinaminas/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Fator 3 Associado a Receptor de TNF/análise
2.
J Biol Chem ; 297(3): 101097, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34418432

RESUMO

Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) plays context-specific roles in multiple receptor-mediated signaling pathways in different cell types. Mice lacking TRAF3 in T cells display defective T-cell-mediated immune responses to immunization and infection and demonstrate defective early signaling via the TCR complex. However, the role of TRAF3 in the function of GITR/TNFRSF18, an important costimulatory member of the TNFR superfamily, is unclear. Here we investigated the impact of T cell TRAF3 status on both GITR expression and activation of specific kinases in the GITR signaling pathway in T cells. Our results indicate that TRAF3 negatively regulates GITR functions in several ways. First, expression of GITR protein was elevated in TRAF3-deficient T cells, resulting from both transcriptional and posttranslational regulation that led to greater GITR transcript levels, as well as enhanced GITR protein stability. TRAF3 associated with T cell GITR in a manner dependent upon GITR ligation. TRAF3 also inhibited several events of the GITR mediated early signaling cascade, in a manner independent of recruitment of phosphatases, a mechanism by which TRAF3 inhibits signaling through several other cytokine receptors. These results add new information to our understanding of GITR signaling and function in T cells, which is relevant to the potential use of GITR to enhance immune therapies.


Assuntos
Receptores Coestimuladores e Inibidores de Linfócitos T/metabolismo , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Animais , Feminino , Proteína Relacionada a TNFR Induzida por Glucocorticoide/fisiologia , Interleucina-2/metabolismo , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Fator 3 Associado a Receptor de TNF/fisiologia
3.
Exp Dermatol ; 30(11): 1705-1710, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33999445

RESUMO

Recently described Hungarian and Anglo-Saxon pedigrees that are affected by CYLD cutaneous syndrome (syn: Brooke-Spiegler syndrome (BSS)) carry the same disease-causing mutation (c.2806C>T, p.Arg936X) of the cylindromatosis (CYLD) gene but exhibit striking phenotypic differences. Using whole exome sequencing, missense genetic variants of the TRAF3 and NBR1 genes were identified in the affected family members of the Hungarian pedigree that are not present in the Anglo-Saxon pedigree. This suggested that the affected proteins (TRAF3 and NBR1) are putative phenotype-modifying factors. An in vitro experimental system was set up to clarify how wild type and mutant TRAF3 and NBR1 modify the effect of CYLD on the NF-κB signal transduction pathway. Our study revealed that the combined expression of mutant CYLD(Arg936X) with TRAF3 and NBR1 caused increased NF-κB activity, regardless of the presence or absence of mutations in TRAF3 and NBR1. We concluded that increased expression levels of these proteins further strengthen the effect of the CYLD(Arg936X) mutation on NF-κB activity in HEK293 cells and may explain the phenotype-modifying effect of these genes in CYLD cutaneous syndrome. These results raise the potential that detecting the levels of TRAF3 and NBR1 might help explaining phenotypic differences and prognosis of CCS.


Assuntos
Enzima Desubiquitinante CYLD/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Mutação , NF-kappa B/fisiologia , Síndromes Neoplásicas Hereditárias/genética , Neoplasias Cutâneas/genética , Fator 3 Associado a Receptor de TNF/fisiologia , Humanos
4.
Front Immunol ; 11: 2110, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042123

RESUMO

Tumor necrosis factor receptor (TNFR)-associated factors (TRAFs) are vital signaling adaptor proteins for the innate immune response and are involved in many important pathways, such as the NF-κB- and interferon regulatory factor (IRF)-activated signaling pathways. In this study, the TRAF3 ortholog from the shrimp Litopenaeus vannamei (LvTRAF3) was cloned and characterized. LvTRAF3 has a transcript of 3,865 bp, with an open reading frame (ORF) of 1,002 bp and encodes a polypeptide of 333 amino acids, including a conserved TRAF-C domain. The expression of LvTRAF3 in the intestine and hemocyte was up-regulated in response to poly (I:C) challenge and white spot syndrome virus (WSSV) infection. RNAi knockdown of LvTRAF3 in vivo significantly increased WSSV gene transcription, viral loads, and mortality in WSSV-infected shrimp. Next, we found that LvTRAF3 was not able to induce the activation of the NF-κB pathway, which was crucial for synthesis of antimicrobial peptides (AMPs), which mediate antiviral immunity. Specifically, in dual-luciferase reporter assays, LvTRAF3 could not activate several types of promoters with NF-κB binding sites, including those from WSSV genes (wsv069, wsv056, and wsv403), Drosophila AMPs or shrimp AMPs. Accordingly, the mRNA levels of shrimp AMPs did not significantly change when TRAF3 was knocked down during WSSV infection. Instead, we found that LvTRAF3 signaled through the IRF-Vago antiviral cascade. LvTRAF3 functioned upstream of LvIRF to regulate the expression of LvVago4 and LvVago5 during WSSV infection in vivo. Taken together, these data provide experimental evidence of the participation of LvTRAF3 in the host defense to WSSV through the activation of the IRF-Vago pathway but not the NF-κB pathway.


Assuntos
Citocinas/fisiologia , Fatores Reguladores de Interferon/fisiologia , Penaeidae/imunologia , Transdução de Sinais/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Vírus da Síndrome da Mancha Branca 1/fisiologia , Sequência de Aminoácidos , Animais , Aquicultura , Sequência de Bases , Linhagem Celular , Hemócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Penaeidae/virologia , Filogenia , Interferência de RNA , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/farmacologia , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Fator 3 Associado a Receptor de TNF/antagonistas & inibidores , Fator 3 Associado a Receptor de TNF/biossíntese , Fator 3 Associado a Receptor de TNF/genética , Replicação Viral
5.
FASEB J ; 34(9): 12392-12405, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32779804

RESUMO

Type I interferons play a pivotal role in innate immune response to virus infection. The protein tyrosine phosphatase SHP-1 was reported to function as a negative regulator of inflammatory cytokine production by inhibiting activation of NF-κB and MAPKs during bacterial infection, however, the role of SHP-1 in regulating type I interferons remains unknown. Here, we demonstrated that knockout or knockdown of SHP-1 in macrophages promoted both HSV-1- and VSV-induced antiviral immune response. Conversely, overexpression of SHP-1 in L929 cells suppressed the HSV-1- and VSV-induced immune response; suppression was directly dependent on phosphatase activity. We identified a direct interaction between SHP-1 and TRAF3; the association between these two proteins resulted in diminished recruitment of CK1ε to TRAF3 and inhibited its K63-linked ubiquitination; SHP-1 inhibited K63-linked ubiquitination of TRAF3 by promoting dephosphorylation at Tyr116 and Tyr446. Taken together, our results identify SHP-1 as a negative regulator of antiviral immunity and suggest that SHP-1 may be a target for intervention in acute virus infection.


Assuntos
Proteína Tirosina Fosfatase não Receptora Tipo 6/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Viroses/imunologia , Animais , Células HEK293 , Humanos , Imunidade Inata , Camundongos , Células RAW 264.7 , Ubiquitinação
6.
J Am Soc Nephrol ; 31(9): 2026-2042, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32631974

RESUMO

BACKGROUND: CKD leads to vitamin D deficiency. Treatment with vitamin D receptor agonists (VDRAs) may have nephroprotective and anti-inflammatory actions, but their mechanisms of action are poorly understood. METHODS: Modulation of the noncanonical NF-κB2 pathway and its component TNF receptor-associated factor 3 (TRAF3) by the VDRA paricalcitol was studied in PBMCs from patients with ESKD, cytokine-stimulated cells, and preclinical kidney injury models. RESULTS: In PBMCs isolated from patients with ESKD, TRAF3 protein levels were lower than in healthy controls. This finding was associated with evidence of noncanonical NF-κB2 activation and a proinflammatory state. However, PBMCs from patients with ESKD treated with paricalcitol did not exhibit these features. Experiments in cultured cells confirmed the link between TRAF3 and NF-κB2/inflammation. Decreased TRAF3 ubiquitination in K48-linked chains and cIAP1-TRAF3 interaction mediated the mechanisms of paricalcitol action.TRAF3 overexpression by CRISPR/Cas9 technology mimicked VDRA's effects. In a preclinical model of kidney injury, paricalcitol inhibited renal NF-κB2 activation and decreased renal inflammation. In VDR knockout mice with renal injury, paricalcitol prevented TRAF3 downregulation and NF-κB2-dependent gene upregulation, suggesting a VDR-independent anti-inflammatory effect of paricalcitol. CONCLUSIONS: These data suggest the anti-inflammatory actions of paricalcitol depend on TRAF3 modulation and subsequent inhibition of the noncanonical NF-κB2 pathway, identifying a novel mechanism for VDRA's effects. Circulating TRAF3 levels could be a biomarker of renal damage associated with the inflammatory state.


Assuntos
Anti-Inflamatórios/farmacologia , Ergocalciferóis/farmacologia , Falência Renal Crônica/tratamento farmacológico , Receptores de Calcitriol/agonistas , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Células Cultivadas , Citocina TWEAK/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Receptores de Calcitriol/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator 3 Associado a Receptor de TNF/análise
7.
Biochimie ; 167: 217-227, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31654668

RESUMO

RANKL induces osteoclastogenesis via JNK1 signal that exerts an anti-apoptotic effect during osteoclastogenesis. However, the classic downstream c-Jun/AP-1 pathway is not included in anti-apoptosis of JNK1. Thus, the detail mechanism underlying JNK1-resisted apoptosis remains unknown during RANKL-induced osteoclastogenesis. RANKL-induced autophagy results in the degradation of the osteoclastogenesis-inhibitor TRAF3, and TRAF3 is thought as a regulator of apoptosis. Given the key effect of JNK1 in mediating autophagy, our study aims to investigate the significance of TRAF3 in bridging JNK1-mediated autophagy and apoptotic resistance during osteoclastogenesis. In this study, by using Bone Marrow-derived macrophages (BMMs) as osteoclast precursors (OCPs), we found that RANKL-induced TRAF3 degradation was significantly suppressed by JNK inhibitor (SP600125), which was restored by overexpression of Beclin1 (key autophagic protein). Nevertheless, TRAF3 silencing partially reversed the reduced osteoclastogenesis under SP600125 intervention. Besides, OCP apoptosis was positively regulated by TRAF3 overexpression, regardless of the application of autophagy inhibitor or SP600125. Remarkably, the enhanced apoptosis caused by the pharmacological inhibition of Beclin1 was reversed by TRAF3 silencing. Together, these results suggest that JNK1-mediated autophagy could promote RANKL-induced osteoclastogenesis via enhancing TRAF3 degradation. Importantly, JNK1 could prevent OCP apoptosis through autophagy-TRAF3 signaling, which provides more potential targets for clinical treatment of pathological bone loss.


Assuntos
Macrófagos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Apoptose , Autofagia , Proteína Beclina-1/metabolismo , Células Cultivadas , Macrófagos/citologia , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Osteoclastos/citologia , Ligante RANK/metabolismo , Transdução de Sinais
8.
Front Immunol ; 10: 1069, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31139191

RESUMO

The serine/threonine kinase phosphatase and tensin homolog (PTEN)-induced putative kinase 1(PINK1) controls mitochondrial quality and plays a vital role in the pathogenesis of early-onset Parkinson's disease. However, whether PINK1 has functions in innate antiviral immunity is largely unknown. Here, we report that viral infection down regulates PINK1 expression in macrophages. PINK1 knockdown results in decreased cytokine production and attenuated IRF3 and NF-κB activation upon viral infection. PINK1 promotes the retinoic-acid-inducible gene I (RIG-I)-like receptors (RLR)-triggered immune responses in a kinase domain-dependent manner. Furthermore, PINK1 associates with TRAF3 via the kinase domain and inhibits Parkin-mediated TRAF3 K48-linked proteasomal degradation. In addition, PINK1 interacts with Yes-associated protein 1 (YAP1) upon viral infection and impairs YAP1/IRF3 complex formation. Collectively, our results demonstrate that PINK1 positively regulates RIG-I triggered innate immune responses by inhibiting TRAF3 degradation and relieving YAP-mediated inhibition of the cellular antiviral response.


Assuntos
Proteína DEAD-box 58/fisiologia , Proteínas Mitocondriais/fisiologia , Proteínas Quinases/fisiologia , Viroses/imunologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Citocinas/biossíntese , Feminino , Células HEK293 , Humanos , Lactente , Fator Regulador 3 de Interferon/fisiologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Células RAW 264.7 , Receptores Imunológicos , Transdução de Sinais/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Fatores de Transcrição/fisiologia , Ubiquitinação , Proteínas de Sinalização YAP
9.
Biomol Concepts ; 8(3-4): 197-202, 2017 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-28753533

RESUMO

Tumour necrosis factor receptor-associated factor 3 (TRAF3) is a member of the TRAF adaptor protein family, which exerts different effects on the cell depending on the receptor to which it binds and the cell type in which it is expressed. TRAF3 is a major regulator of the innate immune response. To perform its functions properly, TRAF3 is transcriptionally and epigenetically regulated. At the transcriptional level, TRAF3 expression has been associated with neurological and cardiovascular diseases including stroke, among other pathologies. Epigenetic modifications of TRAF3 have been observed at the histone and DNA levels. It has been observed that acetylation of TRAF3, as well as other NF-κß target genes, is associated with cardiac hypertrophy. Furthermore, TRAF3 methylation has been associated with vascular recurrence after ischemic stroke in patients treated with clopidogrel. In this overview, we summarise the most interesting studies related to transcriptional and epigenetic regulation of TRAF3 focusing on those studies performed in neurological and cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/metabolismo , Doenças do Sistema Nervoso/metabolismo , Fator 3 Associado a Receptor de TNF/fisiologia , Acetilação , Epigênese Genética , Humanos , Metilação , NF-kappa B/metabolismo , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/metabolismo , Fator de Transcrição AP-1/metabolismo
10.
IUBMB Life ; 69(3): 170-178, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28185403

RESUMO

This study aims to investigate the effects of TNF receptors associated factor 3 (TRAF3) on the signaling pathway and expression of downstream products of nuclear factor kappa B (NF-κB) in the epithelial cells of renal ducts in individuals with polycystic kidney disease (PKD). We observe the TRAF3 genic overexpression of the epithelial cells, which form a tubular branch structure, in polycystic kidneys and to explore the protective effect of TRAF3 on the cystogenesis and progression of PKD. Western blotting analysis was conducted to examine the signaling changes of NF-κB in PKD the epithelial cells and TRAF3 transgenic PKD epithelial cells. Changes in the downstream apoptosis factor and cell proliferation in PKD epithelial cells and TRAF3 transgenic PKD epithelial cells were detected. A three-dimensional matrigel culture experiment was performed to examine abnormal tubulomorphogenesis in vitro. The overexpression of TRAF3 significantly inhibited the signaling pathway of NF-κB in the PKD epithelial cells, downregulated the expression of downstream factors Bcl-2 and Bcl-xl, and significantly decreased cystic epithelial cell proliferation. Additional branch structures were observed in the PKD epithelial cells with a three-dimensional culture compared to wildtype cells. TRAF3 may likely induce apoptosis and resistance to proliferation and may be a new target to inhibit the cyst formation in PKD by regulating the NF-κB signaling pathway Bcl-2 and Bcl-xl activity. © 2017 IUBMB Life, 69(3):170-178, 2017.


Assuntos
Doenças Renais Policísticas/metabolismo , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Cistos/metabolismo , Células Epiteliais/metabolismo , Rim/metabolismo , Rim/patologia , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Transdução de Sinais
11.
Proc Natl Acad Sci U S A ; 113(4): 1032-7, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26755589

RESUMO

The adaptor protein TNF receptor-associated factor 3 (TRAF3) regulates signaling through B-lymphocyte receptors, including CD40, BAFF receptor, and Toll-like receptors, and also plays a critical role inhibiting B-cell homoeostatic survival. Consistent with these findings, loss-of-function human TRAF3 mutations are common in B-cell cancers, particularly multiple myeloma and B-cell lymphoma. B cells of B-cell-specific TRAF3(-/-) mice (B-Traf3(-/-)) display remarkably enhanced survival compared with littermate control (WT) B cells. The mechanism for this abnormal homeostatic survival is poorly understood, a key knowledge gap in selecting optimal treatments for human B-cell cancers with TRAF3 deficiency. We show here for the first time to our knowledge that TRAF3 is a resident nuclear protein that associates with the transcriptional regulator cAMP response element binding protein (CREB) in both mouse and human B cells. The TRAF-C domain of TRAF3 was necessary and sufficient to localize TRAF3 to the nucleus via a functional nuclear localization signal. CREB protein was elevated in TRAF3(-/-) B cells, without change in mRNA, but with a decrease in CREB ubiquitination. CREB-mediated transcriptional activity was increased in TRAF3-deficient B cells. Consistent with these findings, Mcl-1, an antiapoptotic target of CREB-mediated transcription, was increased in the absence of TRAF3 and enhanced Mcl-1 was suppressed with CREB inhibition. TRAF3-deficient B cells were also preferentially sensitive to survival inhibition with pharmacologic CREB inhibitor. Our results identify a new mechanism by which nuclear TRAF3 regulates B-cell survival via inhibition of CREB stability, information highly relevant to the role of TRAF3 in B-cell malignancies.


Assuntos
Linfócitos B/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Proteínas Nucleares/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Adolescente , Adulto , Animais , Linhagem Celular , Sobrevivência Celular , Humanos , Linfoma de Células B/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Sinais de Localização Nuclear
12.
Cancer Res ; 76(5): 1135-45, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26759244

RESUMO

Chronic infection with the bacterial Helicobacter pylori is a major cause of gastric and duodenal ulcer disease, gastric mucosal atrophy, and cancer. H. pylori-induced expression of the intestinal epithelial-specific transcription factor caudal-related homeobox 2 (Cdx2) contributes to intestinal metaplasia, a precursor event to gastric cancer. Given a role for the bacterial pattern recognition molecule nucleotide-binding oligomerization domain 1 (NOD1) in the innate immune response to bacterial infection, we investigated mechanisms used by NOD1 to regulate H. pylori infection and its propensity towards the development of intestinal metaplasia. We found that Cdx2 was induced by H. pylori infection in both normal and neoplastic gastric epithelial cells in a manner that was inversely related to NOD1 signaling. Mechanistic investigations revealed that Cdx2 induction relied upon activation of NF-κB but was suppressed by NOD1-mediated activation of TRAF3, a negative regulator of NF-κB. In vivo, prolonged infection of NOD1-deficient mice with H. pylori led to increased Cdx2 expression and intestinal metaplasia. Furthermore, gastric epithelial cells from these mice exhibited increased nuclear expression of the NF-κB p65 subunit and decreased expression of TRAF3. Overall, our findings illuminated a role for NOD1 signaling in attenuating H. pylori-induced Cdx2 expression in gastric epithelial cells, suggesting a rationale to augment NOD1 signaling in H. pylori-infected patients to limit their risks of accumulating precancerous gastric lesions.


Assuntos
Mucosa Gástrica/patologia , Infecções por Helicobacter/complicações , Helicobacter pylori , Proteínas de Homeodomínio/fisiologia , Proteína Adaptadora de Sinalização NOD1/fisiologia , Fatores de Transcrição/fisiologia , Animais , Fator de Transcrição CDX2 , Linhagem Celular Tumoral , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Imunidade Inata , Masculino , Metaplasia , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Regiões Promotoras Genéticas , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/fisiologia , Fatores de Transcrição/genética
13.
J Hepatol ; 64(1): 146-59, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26334576

RESUMO

BACKGROUND & AIMS: The hallmarks of hepatic ischemia/reperfusion (I/R) injury, a common clinical problem that occurs during liver surgical procedures, include severe cell death and inflammatory responses that contribute to early graft failure and a higher incidence of organ rejection. Unfortunately, effective therapeutic strategies are limited. Tumor necrosis factor receptor (TNFR)-associated factor (TRAF) 3 transduces apoptosis and/or inflammation-related signaling pathways to regulate cell survival and cytokine production. However, the role of TRAF3 in hepatic I/R-induced liver damage remains unknown. METHODS: Hepatocyte- or myeloid cell-specific TRAF3 knockdown or transgenic mice were subjected to an I/R model in vivo, and in vitro experiments were performed by treating primary hepatocytes from these mice with hypoxia/reoxygenation stimulation. The function of TRAF3 in I/R-induced liver damage and the potential underlying mechanisms were investigated through various phenotypic analyses and biological approaches. RESULTS: Hepatocyte-specific, but not myeloid cell-specific, TRAF3 deficiency reduced cell death, inflammatory cell infiltration, and cytokine production in both in vivo and in vitro hepatic I/R models, whereas hepatic TRAF3 overexpression resulted in the opposite effects. Mechanistically, TRAF3 directly binds to TAK1, which enhances the activation of the downstream NF-κB and JNK pathways. Importantly, inhibition of TAK1 almost completely reversed the TRAF3 overexpression-mediated exacerbation of I/R injury. CONCLUSIONS: TRAF3 is a novel hepatic I/R mediator that promotes liver damage and inflammation via TAK1-dependent activation of the JNK and NF-κB pathways. Inhibition of hepatic TRAF3 may represent a promising approach to protect the liver against I/R injury-related diseases.


Assuntos
Fígado/irrigação sanguínea , Traumatismo por Reperfusão/etiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Feminino , Humanos , MAP Quinase Quinase Quinases/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/fisiologia , Traumatismo por Reperfusão/prevenção & controle
14.
Hypertension ; 66(2): 356-67, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26034202

RESUMO

Cardiac hypertrophy, a common early symptom of heart failure, is regulated by numerous signaling pathways. Here, we identified tumor necrosis factor receptor-associated factor 3 (TRAF3), an adaptor protein in tumor necrosis factor-related signaling cascades, as a key regulator of cardiac hypertrophy in response to pressure overload. TRAF3 expression was upregulated in hypertrophied mice hearts and failing human hearts. Four weeks after aortic banding, cardiac-specific conditional TRAF3-knockout mice exhibited significantly reduced cardiac hypertrophy, fibrosis, and dysfunction. Conversely, transgenic mice overexpressing TRAF3 in the heart developed exaggerated cardiac hypertrophy in response to pressure overload. TRAF3 also promoted an angiotensin II- or phenylephrine-induced hypertrophic response in isolated cardiomyocytes. Mechanistically, TRAF3 directly bound to TANK-binding kinase 1 (TBK1), causing increased TBK1 phosphorylation in response to hypertrophic stimuli. This interaction between TRAF3 and TBK1 further activated AKT signaling, which ultimately promoted the development of cardiac hypertrophy. Our findings not only reveal a key role of TRAF3 in regulating the hypertrophic response but also uncover TRAF3-TBK1-AKT as a novel signaling pathway in the development of cardiac hypertrophy and heart failure. This pathway may represent a potential therapeutic target for this pathological process.


Assuntos
Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Transdução de Sinais/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Regulação para Cima/fisiologia , Angiotensina II/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Hipertrofia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fenilefrina/farmacologia , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Fator 3 Associado a Receptor de TNF/deficiência , Fator 3 Associado a Receptor de TNF/genética
15.
Am J Physiol Endocrinol Metab ; 308(6): E460-9, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25628422

RESUMO

Myeloid cells, particularly macrophages, mediate metabolic inflammation, thus promoting insulin resistance and metabolic disease progression in obesity. Numerous cytokines, toxic metabolites, damage-associated molecular patterns, and pathogen-associated molecular patterns are involved in activating macrophages via their cognate receptors in obesity. TRAF3 (TNF receptor-associated factor 3) is a common signaling molecule for these ligands/receptors and negatively regulates the proinflammatory NF-κB and MAPK pathways, but its metabolic activity is unknown. We here show that myeloid cell TRAF3 is required for metabolic inflammation and metabolic disease progression in obesity. Myeloid cell-specific deletion of TRAF3 significantly attenuated insulin resistance, hyperglycemia, hyperinsulinemia, glucose intolerance, and hepatic steatosis in mice with either genetic (ob/ob) or high-fat diet (HFD)-induced obesity. Myeloid cell-specific deletion of TRAF3 had the opposite effects on metabolic inflammation between obese and lean mice. It decreased the expression of proinflammatory cytokines in the liver and adipose tissue of obese mice and largely prevented HFD-induced inflammation in these metabolic tissues; by contrast, in lean mice, it increased the expression of proinflammatory cytokines in the liver and adipose tissue. These data suggest that, in obesity progression, myeloid TRAF3 functionally switches its activity from anti-inflammatory to proinflammatory modes, thereby coupling overnutrition to metabolic inflammation, insulin resistance, and metabolic disease.


Assuntos
Fígado Gorduroso/genética , Inflamação/genética , Resistência à Insulina/genética , Obesidade/complicações , Obesidade/genética , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Células Cultivadas , Fígado Gorduroso/imunologia , Fígado Gorduroso/metabolismo , Deleção de Genes , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Células Mieloides/metabolismo , Obesidade/imunologia , Obesidade/metabolismo
16.
Mol Immunol ; 54(1): 40-7, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23220069

RESUMO

Our earlier studies indicated that activation of the lymphotoxin beta receptor (LTßR) by T cell derived LTα(1)ß(2) regulates inflammatory cytokine expression. While characterizing the cellular and molecular mechanisms responsible for the down regulation of the inflammatory reaction after LTßR stimulation we were able to identify the specific induction of TRIM30α expression as a result of LTßR signalling in mouse macrophages. Furthermore, we could demonstrate that LTßR activation in these cells results in the down regulation of pro-inflammatory cytokine (e.g. TNF and IL-6) and mediator expression upon TLR4 and TLR9 re-stimulation, demonstrating that LTßR activation on mouse macrophages dampens pro-inflammatory cytokine and mediator expression. Thus, LTßR signalling renders macrophages hypo-responsive to subsequent stimulation with TLR ligands. The observation of an LTßR-mediated TLR-tolerance in the human monocyte cell line THP-1 suggests that similar signalling mechanisms seem to exist in human cells. Signalling pathway analysis clearly demonstrated that LTßR-induced TRIM30α expression is mediated by an IκBα-dependent signalling pathway. Furthermore, the LTßR-induced TRIM30α expression seems to be TRAF3 dependent. Our data suggest that LTßR activation on mouse macrophages is involved in the control of pro-inflammatory cytokine and mediator expression by activation of a signalling pathway that controls exacerbating inflammatory cytokine production.


Assuntos
Citocinas/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Receptor beta de Linfotoxina/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Células Cultivadas , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/genética , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptor beta de Linfotoxina/agonistas , Receptor beta de Linfotoxina/antagonistas & inibidores , Receptor beta de Linfotoxina/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/fisiologia , Camundongos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/metabolismo
17.
IUBMB Life ; 64(9): 748-56, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22715070

RESUMO

Calcineurin (CN) is the only serine/threonine specific protein phosphatase regulated by Ca(2+) /calmodulin (CaM), which is composed of catalytic A subunit (CNA) and regulatory B subunit (CNB). Tumor necrosis factor (TNF) receptor associated factor 3 (TRAF3) is an essential component in the Toll like receptors and TNF receptors (TNFRs) pathways. The TRAF domain of TRAF3 interacts with a large range of proteins, which share consensus sequences known as TRAF interacting motifs (TIMs). By sequence alignment, we identified two potential TIMs in CNB. However, the relation between TRAF3 and CN has not been reported before. To explore this, we highly expressed the former insoluble TRAF domain of TRAF3 in soluble form by using CaM fusion system for the first time. We demonstrated that the TRAF domain of TRAF3 interacted with CNB. On further investigation, over-expression of TRAF3 inhibited endogenous CN's activity, which decreased NFAT reporter activity and IL-2 production. Knock-down of TRAF3 partially enhanced CN's activity. The possible mechanism was that TRAF3 functioned as ubiquitin E3 ligase for CN and promoted its degradation.


Assuntos
Calcineurina/metabolismo , Fatores de Transcrição NFATC/metabolismo , Proteólise , Fator 3 Associado a Receptor de TNF/fisiologia , Sinalização do Cálcio , Células HEK293 , Humanos , Interleucina-2/metabolismo , Células Jurkat , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Fator 3 Associado a Receptor de TNF/química , Fator 3 Associado a Receptor de TNF/metabolismo
19.
J Leukoc Biol ; 90(6): 1149-57, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21971520

RESUMO

The key role of TRAF6 in TLR signaling pathways is well known. More recent evidence has implicated TRAF3 as another TRAF family member important to certain TLR responses of myeloid cells. Previous studies demonstrate that TRAF3 functions are highly context-dependent, displaying receptor and cell-type specificity. We thus examined the TLR responses of TRAF3(-/-)mouse B lymphocytes to test the hypothesis that TRAF3 plays distinct roles in such responses, depending on cell type. TRAF3(-/-) DC are known to have a defect in type 1 IFN production and here, showed diminished production of TNF and IL-10 and unaltered IL-6. In marked contrast, TRAF3(-/-) B cells made elevated amounts of TNF and IL-6 protein, as well as IL-10 and IP-10 mRNA, in response to TLR ligands. Also, in contrast to TRAF3(-/-) DC, the type 1 IFN pathway was elevated in TRAF3(-/-) B cells. Increased early responses of TRAF3(-/-) B cells to TLR signals were independent of cell survival or proliferation but associated with elevated canonical NF-κB activation. Additionally, TRAF3(-/-) B cells displayed enhanced TLR-mediated expression of AID and Ig isotype switching. Thus, TRAF3 plays varied and cell type-specific, biological roles in TLR responses.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Fator 3 Associado a Receptor de TNF/deficiência , Fator 3 Associado a Receptor de TNF/fisiologia , Receptores Toll-Like/biossíntese , Animais , Subpopulações de Linfócitos B/citologia , Células Cultivadas , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais/genética , Transdução de Sinais/imunologia
20.
Nat Rev Immunol ; 11(7): 457-68, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21660053

RESUMO

Tumour necrosis factor receptor (TNFR)-associated factor (TRAF) proteins are essential components of signalling pathways activated by TNFR or Toll-like receptor (TLR) family members. Acting alone or in combination, the seven known TRAFs control many biological processes, including cytokine production and cell survival. The function of one TRAF in particular, TRAF3, remained elusive for many years. Recent work has revealed that TRAF3 is a highly versatile regulator that positively controls type I interferon production, but negatively regulates mitogen-activated protein kinase activation and alternative nuclear factor-κB signalling. In this Review, we discuss our current understanding of the role of TRAF3 in TNFR and TLR signalling pathways, and its role in disease.


Assuntos
Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Receptores Toll-Like/metabolismo , Animais , Humanos , Interferon Tipo I/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Fator 3 Associado a Receptor de TNF/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...