Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 10(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35091453

RESUMO

BACKGROUND: While stimulator of interferon genes (STING) activation in innate immune cells of the tumor microenvironment can result in CD8 T cell-dependent antitumor immunity, whether STING signaling affects CD4 T-cell responses remains elusive. METHODS: Here, we tested whether STING activation modulated the effector functions of CD4 T cells in vivo by analyzing tumor-infiltrating CD4 T cells and evaluating the contribution of the CD4 T cell-derived cytokines in the antitumor activity of the STING ligand 2'3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) in two mouse tumor models. We performed ex vivo experiments to assess the impact of STING activation on CD4 T-cell differentiation and investigate the underlying molecular mechanisms. Finally, we tested whether STING activation enhances TH9 cell antitumor activity against mouse melanoma upon adoptive transfer. RESULTS: We found that activation of STING signaling cell-intrinsically enhances the differentiation and antitumor functions of TH1 and TH9 cells by increasing their respective production of interferon gamma (IFN-γ) and interleukin-9. IRF3 and type I interferon receptors (IFNARs) are required for the STING-driven enhancement of TH1 cell differentiation. However, STING activation favors TH9 cell differentiation independently of the IFNARs/IRF3 pathway but through mammalian target of rapamycin (mTOR) signaling, underscoring that STING activation differentially affects the fate of distinct CD4 T-cell subsets. The therapeutic effect of STING activation relies on TH1 and TH9-derived cytokines, and STING activation enhances the antitumor activity of TH9 cells upon adoptive transfer. CONCLUSION: Our results reveal the STING signaling pathway as a therapeutic target to boost CD4 T-cell effector functions and antitumor immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interleucina-9/fisiologia , Proteínas de Membrana/fisiologia , Células Th1/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular , Feminino , Fator Regulador 3 de Interferon/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Nucleotídeos Cíclicos/farmacologia , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Células Th1/citologia
2.
Int J Mol Sci ; 22(16)2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34445616

RESUMO

Neovascularization restores blood flow recovery after ischemia in peripheral arterial disease. The main two components of neovascularization are angiogenesis and arteriogenesis. Both of these processes contribute to functional improvements of blood flow after occlusion. However, discriminating between the specific contribution of each process is difficult. A frequently used model for investigating neovascularization is the murine hind limb ischemia model (HLI). With this model, it is difficult to determine the role of angiogenesis, because usually the timing for the sacrifice of the mice is chosen to be optimal for the analysis of arteriogenesis. More importantly, the occurring angiogenesis in the distal calf muscles is probably affected by the proximally occurring arteriogenesis. Therefore, to understand and subsequently intervene in the process of angiogenesis, a model is needed which investigates angiogenesis without the influence of arteriogenesis. In this study we evaluated the in vivo Matrigel plug assay in genetic deficient mice to investigate angiogenesis. Mice deficient for interferon regulatory factor (IRF)3, IRF7, RadioProtective 105 (RP105), Chemokine CC receptor CCR7, and p300/CBP-associated factor (PCAF) underwent the in vivo Matrigel model. Histological analysis of the Matrigel plugs showed an increased angiogenesis in mice deficient of IRF3, IRF7, and RP105, and a decreased angiogenesis in PCAF deficient mice. Our results also suggest an involvement of CCR7 in angiogenesis. Comparing our results with results of the HLI model found in the literature suggests that the in vivo Matrigel plug assay is superior in evaluating the angiogenic response after ischemia.


Assuntos
Antígenos CD/fisiologia , Membro Posterior/irrigação sanguínea , Fator Regulador 3 de Interferon/fisiologia , Fator Regulador 7 de Interferon/fisiologia , Isquemia/patologia , Neovascularização Patológica/patologia , Fatores de Transcrição de p300-CBP/fisiologia , Animais , Colágeno , Combinação de Medicamentos , Membro Posterior/patologia , Isquemia/metabolismo , Laminina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/metabolismo , Proteoglicanas , Recuperação de Função Fisiológica
3.
Mol Cell Endocrinol ; 518: 110890, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32781250

RESUMO

Lipotoxic injury of pancreatic ß cells is an important pathological feature in type 2 diabetes mellitus (T2DM). Stimulator of interferon genes (STING) can recognize its own DNA leaked into the cytoplasm from damaged mitochondria or nuclei of the host cell, thus activating its downstream factor interferon regulatory factor 3 (IRF3), causing inflammation and apoptosis. The STING-IRF3 signaling pathway is closely related to glycolipid metabolism, but its relationship with the lipotoxicity of pancreatic ß cells has rarely been reported. Here, we investigated the role of the STING-IRF3 signaling pathway in lipotoxicity-induced inflammation, apoptosis, and dysfunction of pancreatic ß cells. We examined the activation of STING and IRF3 in islets of db/db mice and identified the role of the STING-IRF3 signaling pathway in palmitic acid (PA)-induced lipotoxic injury of INS-1, a rat insulinoma cell line. STING and phosphorylated IRF3 including downstream interferon-ß were upregulated in islets of db/db mice and PA-induced INS-1 cells. Gene silencing of STING or IRF3 ameliorated PA-induced INS-1 cell inflammation and apoptosis, and reversed impaired insulin synthesis. Additionally, PA induced downregulation of the phosphoinositide 3-kinase-AKT signaling pathway, and impaired high glucose-stimulated insulin secretion was reversed after knockdown of STING or IRF3. Our results suggest that activation of the STING-IRF3 pathway triggers inflammation and apoptosis of pancreatic ß cells, leading to ß-cell damage and dysfunction. Hence, inhibition of this signaling pathway may represent a novel approach for ß-cell protection in T2DM.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Células Secretoras de Insulina/efeitos dos fármacos , Fator Regulador 3 de Interferon/fisiologia , Proteínas de Membrana/fisiologia , Ácido Palmítico/toxicidade , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Ácido Palmítico/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Front Immunol ; 11: 606, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373114

RESUMO

MyD88 is a conserved intracellular adaptor, which plays an important role in the innate immune system. MyD88 transmits signals for downstream of toll-like and IL-1 receptors to activate NF-κB signaling pathway, which is tightly controlled in the immune response to maintain immune intensity and immune homeostasis at different stages. NF-κB signaling pathway has been extensively studied in mammals, but regulatory molecular mechanism is still unclear in teleost fish. We determined that IRF3 and IRF8 can regulate MyD88-mediated NF-κB signaling pathway in fish. Interestingly, MyD88 is precisely regulated by IRF3 and IRF8 through the same mechanism but in completely opposite ways. IRF3 promotes MyD88-mediated NF-κB signaling pathway, whereas IRF8 inhibits the signaling pathway. MyD88 is regulated via ubiquitin-proteasome degradation, whereas IRF3 or IRF8 inhibited or promoted MyD88 degradation in this pathway. Specifically, in the early stage of lipopolysaccharide (LPS) stimulation or Vibrio infection, up-regulation of IRF3 and down-regulation of IRF8 eventually increased MyD88 expression to activate the NF-κB signaling pathway to trigger immune response. In the late stage of stimulation, down-regulated IRF3 and up-regulated IRF8 synergistically regulate the expression of MyD88 to a normal level, thus maintaining the immune balance of homeostasis and preventing serious damage from persistent over-immunization. This study presents information on Myd88-NF-κB signaling pathway in teleost fish and provides new insights into its regulatory mechanism in fish immune system.


Assuntos
Fator Regulador 3 de Interferon/fisiologia , Fatores Reguladores de Interferon/fisiologia , Fator 88 de Diferenciação Mieloide/fisiologia , NF-kappa B/fisiologia , Perciformes/imunologia , Animais , Células Cultivadas , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/análise , Complexo de Endopeptidases do Proteassoma/fisiologia , Transdução de Sinais/fisiologia , Ubiquitinação , Vibrioses/imunologia
5.
Eur J Immunol ; 50(8): 1142-1153, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32135578

RESUMO

Exposure to diesel exhaust particles (DEPs) is associated with acute inflammatory responses in the lung and exacerbation of respiratory diseases. However, the mechanism by which DEPs trigger the inflammatory responses remains unclear. Here, we demonstrated that the IFN response factors IRF3 and IRF7 played pivotal roles in DEP-induced pulmonary inflammation. DEPs could not directly induce inflammatory cytokine expression in mouse cells, whereas DEPs triggered autophagy both in vitro and in vivo. The DEP-induced autophagy was augmented in the absence of IRF3 and IRF7, but not in the absence of IFNAR. The expression of Raptor was induced by IRF3 and IRF7 in response to DEPs treatment. Furthermore, administration of the mechanistic target of rapamycin (mTOR) inhibitor alleviated the inflammatory responses in the lung during DEP exposure. Our findings define an IFNAR-independent role of increased autophagy in the absence of IRF3 and IRF7 during pulmonary DEP exposure, and provide the basis to develop new therapeutic approaches to counteract the adverse effects of DEPs and possibly other ambient particulate matters.


Assuntos
Autofagia/fisiologia , Fator Regulador 3 de Interferon/fisiologia , Fator Regulador 7 de Interferon/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Pneumonia/etiologia , Emissões de Veículos/toxicidade , Animais , Citocinas/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta/fisiologia , Sirolimo/farmacologia
6.
Int J Radiat Biol ; 96(5): 642-650, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31914348

RESUMO

Purpose: Pulmonary inflammation is an adverse consequence of radiation therapy in breast cancer. The aim of this study was to elucidate biological pathways leading to this pathology.Materials and methods: Lung endothelial cells were isolated 24 h after thorax-irradiation (sham or 10 Gy X-ray) from female C57Bl/6 mice and cultivated for 6 days.Results: Quantitative proteomic analysis of lung endothelial cells was done using data independent acquisition (DIA) mass spectrometry. The data were analyzed using Ingenuity Pathway Analysis and STRINGdb. In total, 4220 proteins were identified using DIA of which 60 were dysregulated in the irradiated samples (fold change ≥2.00 or ≤0.50; q-value <0.05). Several (12/40) upregulated proteins formed a cluster of inflammatory proteins with STAT1 and IRF3 as predicted upstream regulators. The several-fold increased expression of STAT1 and STAT-associated ISG15 was confirmed by immunoblotting. The expression of antioxidant proteins SOD1 and PRXD5 was downregulated suggesting radiation-induced oxidative stress. Similarly, the phosphorylated (active) forms of STING and IRF3, both members of the cGAS/STING pathway, were downregulated.Conclusions: These data suggest the involvement of JAK/STAT and cGas/STING pathways in the genesis of radiation-induced lung inflammation. These pathways may be used as novel targets for the prevention of radiation-induced lung damage.


Assuntos
Células Endoteliais/efeitos da radiação , Inflamação/etiologia , Pulmão/efeitos da radiação , Espectrometria de Massas/métodos , Fator de Transcrição STAT1/fisiologia , Animais , Feminino , Fator Regulador 3 de Interferon/fisiologia , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteômica , Transdução de Sinais
7.
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
8.
Cell Rep ; 27(9): 2649-2664.e5, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31141689

RESUMO

Lung inflammation induced by silica impairs host control of tuberculosis, yet the underlying mechanism remains unclear. Here, we show that silica-driven exacerbation of M. tuberculosis infection associates with raised type 2 immunity. Silica increases pulmonary Th2 cell and M2 macrophage responses, while reducing type 1 immunity after M. tuberculosis infection. Silica induces lung damage that prompts extracellular self-DNA release and activates STING. This STING priming potentiates M. tuberculosis DNA sensing by and activation of cGAS/STING, which triggers enhanced type I interferon (IFNI) response and type 2 immunity. cGAS-, STING-, and IFNAR-deficient mice are resistant to silica-induced exacerbation of M. tuberculosis infection. Thus, silica-induced self-DNA primes the host response to M. tuberculosis-derived nucleic acids, which increases type 2 immunity while reducing type 1 immunity, crucial for controlling M. tuberculosis infection. These data show how cGAS/STING pathway activation, at the crossroads of sterile inflammation and infection, may affect the host response to pathogens such as M. tuberculosis.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Proteínas de Membrana/fisiologia , Mycobacterium tuberculosis/imunologia , Pneumonia/complicações , Dióxido de Silício/toxicidade , Tuberculose/etiologia , Animais , Células Dendríticas , Fator Regulador 3 de Interferon/fisiologia , Interferon Tipo I/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nucleotidiltransferases/fisiologia , Pneumonia/induzido quimicamente , Receptor de Interferon alfa e beta/fisiologia , Transdução de Sinais , Tuberculose/metabolismo , Tuberculose/patologia
9.
Front Immunol ; 10: 474, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30936875

RESUMO

Interferon-inducible protein (IFI204) (p204, the murine homolog of human IFI16) is known as a cytosolic DNA sensor to recognize DNA viruses and intracellular bacteria. However, little is known about its role during extracellular bacterial infection. Here we show that IFI204 is required for host defense against the infection of Staphylococcus aureus, an extracellular bacterial pathogen. IFI204 deficiency results in decreased survival, increased bacterial loads, severe organs damage, and decreased recruitment of neutrophils and macrophages. Production of several inflammatory cytokines/chemokines including IFN-ß and KC is markedly decreased, as well as the related STING-IRF3 and NF-κB pathways are impaired. However, exogenous administration of recombinant KC or IFN-ß is unable to rescue the susceptibility of IFI204-deficient mice, suggesting that other mechanisms rather than KC and IFN-ß account for IFI204-mediated host defense. IFI204 deficiency leads to a defect in extracellular bacterial killing in macrophages and neutrophils, although bacterial engulf, and intracellular killing activity are normal. Moreover, the defect of bactericidal activity is mediated by decreased extracellular trap formation in the absence of IFI204. Adoptively transferred WT bone marrow cells significantly protect WT and IFI204-deficient recipients against Staphylococcus infection compared with transferred IFI204-deficient bone marrow cells. Hence, this study suggests that IFI204 is essential for the host defense against Staphylococcus infection.


Assuntos
DNA Bacteriano/imunologia , Macrófagos/imunologia , Neutrófilos/microbiologia , Proteínas Nucleares/imunologia , Fosfoproteínas/imunologia , Infecções Estafilocócicas/imunologia , Animais , Bacteriemia/imunologia , Bacteriemia/microbiologia , Carga Bacteriana , Transplante de Medula Óssea , Quimiocina CXCL1/biossíntese , Quimiocina CXCL1/imunologia , Quimiocina CXCL1/farmacologia , Citocinas/biossíntese , Feminino , Fator Regulador 3 de Interferon/fisiologia , Interferon beta/biossíntese , Interferon beta/imunologia , Interferon beta/farmacologia , Macrófagos/microbiologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Masculino , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Neutrófilos/imunologia , Proteínas Nucleares/deficiência , Fosfoproteínas/deficiência , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Quimera por Radiação , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Infecções Estafilocócicas/microbiologia
10.
J Hepatol ; 70(5): 974-984, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30710579

RESUMO

BACKGROUND & AIMS: Interferon regulatory factor 3 (IRF3) is a transcription factor mediating antiviral responses, yet recent evidence indicates that IRF3 also has critical non-transcriptional functions, including activating RIG-I-like receptors-induced IRF-3-mediated pathway of apoptosis (RIPA) and restricting activity of NF-κB. Using a novel murine model expressing only non-transcriptional IRF3 activity (Irf3S1/S1), we tested the hypothesis that non-transcriptional functions of IRF3 modulate innate immune responses in the Gao-binge (acute-on-chronic) model of alcohol-related liver disease. METHODS: IRF3 and IRF3-mediated signals were analysed in liver samples from 5 patients transplanted for alcoholic hepatitis and 5 healthy controls. C57BL/6, Irf3-/- and Irf3S1/S1 mice were exposed to Gao-binge ethanol-induced liver injury. IRF3-mediated RIPA was investigated in cultured macrophages. RESULTS: Phospho-IRF3 and IRF3-mediated signals were elevated in livers of patients with alcoholic hepatitis. In C57BL/6 mice, Gao-binge ethanol exposure activated IRF3 signaling and resulted in hepatocellular injury. Indicators of liver injury were differentially impacted by Irf3 genotype. Irf3-/-, but not Irf3S1/S1, mice were protected from steatosis, elevated alanine/aspartate aminotransferase levels and inflammatory cytokine expression. In contrast, neutrophil accumulation and endoplasmic reticulum stress were independent of genotype. Protection from Gao-binge injury in Irf3-/- mice was associated with an increased ratio of Ly6Clow (restorative) to Ly6Chigh (inflammatory) cells compared to C57BL/6 and Irf3S1/S1 mice. Reduced ratios of Ly6Clow/Ly6Chigh in C57BL/6 and Irf3S1/S1 mice were associated with increased apoptosis in the Ly6Clow population in response to Gao-binge. Activation of primary macrophage cultures with Poly (I:C) induced translocation of IRF3 to the mitochondria, where it associated with Bax and activated caspases 3 and 9, processes indicative of activation of the RIPA pathway. CONCLUSIONS: Taken together, these data identify that the non-transcriptional function of IRF3 plays an important role in modulating the innate immune environment in response to Gao-binge ethanol exposure, via regulation of immune cell apoptosis. LAY SUMMARY: Activation of the innate immune system contributes to inflammation in the progression of alcohol-related liver disease, as well as to the resolution of injury. Here we show that the protein IRF3 modulates the innate immune environment of the liver in a mouse model of alcoholic hepatitis. It does this by increasing the apoptotic cell death of immune cells that promote the resolution of injury.


Assuntos
Hepatite Alcoólica/imunologia , Fator Regulador 3 de Interferon/fisiologia , Fígado/imunologia , Animais , Apoptose , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático , Hepatite Alcoólica/etiologia , Humanos , Imunidade Inata , Fígado/patologia , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/fisiologia , Neutrófilos/fisiologia , Transcrição Gênica
11.
Int Immunopharmacol ; 64: 1-9, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30142469

RESUMO

Toll-like receptors (TLRs) play a crucial role in the induction of innate immune response against bacterial and viral infections. TLRs induce downstream signaling via MyD88- and TRIF-dependent pathways. Cardamonin is a naturally occurring chalcone from Alpinia species exhibiting anti-inflammatory effects. However, the principal molecular mechanisms remain unclear. The objective of this study was to investigate the role of cardamonin in TLR signaling pathways. Cardamonin inhibited NF-κB activation as well as COX-2 expression induced by TLR agonists. Cardamonin inhibited the activation of IRF3 and the expression of interferon-inducible protein-10 (IP-10) induced by TLR3 or TLR4 agonists. Cardamonin also inhibited ligand-independent NF-κB activation overexpressed by MyD88, IKKß, or p65 and IRF3 activation overexpressed by TRIF, TBK1, or IRF3. However, cardamonin had no effect on TBK1 kinase activity in vitro. These results suggest that cardamonin modulates both the MyD88- and TRIF-dependent pathways of TLRs and represents a potentially new anti-inflammatory candidate.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Chalconas/farmacologia , Fator 88 de Diferenciação Mieloide/fisiologia , Transdução de Sinais/efeitos dos fármacos , Receptores Toll-Like/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Animais , Fator Regulador 3 de Interferon/fisiologia , Camundongos , NF-kappa B/antagonistas & inibidores , Células RAW 264.7
12.
Metabolism ; 81: 13-24, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29106945

RESUMO

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a common result of obesity and metabolic syndrome. Hepatocyte injury and metabolic disorders are hallmarks of NAFLD. Stimulator of interferon genes (STING) and its downstream factor interferon regulatory factor 3 (IRF3) trigger inflammatory reaction in response to the presence of cytosolic DNA. STING has recently been shown to play an important role in early alcoholic liver disease. However, little is known about the role of STING-IRF3 pathway in hepatocyte injury. Here, we aimed to examine the effect of STING-IRF3 pathway on hepatocyte metabolism, inflammation and apoptosis. METHODS: We examined the activation of the STING-IRF3 pathway, a high-fat diet (HFD)-induced obese mouse model, and determined the role of this pathway in a free fatty acid (FFA)-induced hepatocyte inflammatory response, injury, and dysfunction in L-O2 human liver cells. RESULTS: STING and IRF3 were upregulated in livers of HFD-fed mice and in FFA-induced L-O2 cells. Knocking down either STING or IRF3 led to a significant reduction in FFA-induced hepatic inflammation and apoptosis, as evidenced by modulation of the nuclear factor κB (NF-κB) signaling pathway, inflammatory cytokines, and apoptotic signaling. Additionally, STING/IRF3 knockdown enhanced glycogen storage and alleviated lipid accumulation, which were found to be associated with increased expression of hepatic enzymes in glycolysis and lipid catabolism, and attenuated expression of hepatic enzymes in gluconeogenesis and lipid synthesis. CONCLUSIONS: Our results suggest that the STING-IRF3 pathway promotes hepatocyte injury and dysfunction by inducing inflammation and apoptosis and by disturbing glucose and lipid metabolism. This pathway may be a novel therapeutic target for preventing NAFLD development and progression.


Assuntos
Apoptose , Hepatite/etiologia , Hepatócitos/patologia , Fator Regulador 3 de Interferon/fisiologia , Proteínas de Membrana/fisiologia , Doenças Metabólicas/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Células Cultivadas , Dieta Hiperlipídica , Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos , Masculino , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/metabolismo , Transdução de Sinais
13.
Nat Med ; 23(12): 1481-1487, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29106401

RESUMO

Interferon regulatory factor 3 (IRF3) and type I interferons (IFNs) protect against infections and cancer, but excessive IRF3 activation and type I IFN production cause autoinflammatory conditions such as Aicardi-Goutières syndrome and STING-associated vasculopathy of infancy (SAVI). Myocardial infarction (MI) elicits inflammation, but the dominant molecular drivers of MI-associated inflammation remain unclear. Here we show that ischemic cell death and uptake of cell debris by macrophages in the heart fuel a fatal response to MI by activating IRF3 and type I IFN production. In mice, single-cell RNA-seq analysis of 4,215 leukocytes isolated from infarcted and non-infarcted hearts showed that MI provokes activation of an IRF3-interferon axis in a distinct population of interferon-inducible cells (IFNICs) that were classified as cardiac macrophages. Mice genetically deficient in cyclic GMP-AMP synthase (cGAS), its adaptor STING, IRF3, or the type I IFN receptor IFNAR exhibited impaired interferon-stimulated gene (ISG) expression and, in the case of mice deficient in IRF3 or IFNAR, improved survival after MI as compared to controls. Interruption of IRF3-dependent signaling resulted in decreased cardiac expression of inflammatory cytokines and chemokines and decreased inflammatory cell infiltration of the heart, as well as in attenuated ventricular dilation and improved cardiac function. Similarly, treatment of mice with an IFNAR-neutralizing antibody after MI ablated the interferon response and improved left ventricular dysfunction and survival. These results identify IRF3 and the type I IFN response as a potential therapeutic target for post-MI cardioprotection.


Assuntos
Fator Regulador 3 de Interferon/fisiologia , Interferon Tipo I/fisiologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/mortalidade , Animais , Células Cultivadas , Citocinas/metabolismo , Inflamação/genética , Inflamação/metabolismo , Fator Regulador 3 de Interferon/genética , Interferon Tipo I/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/patologia , Receptor de Interferon alfa e beta/metabolismo , Receptor de Interferon alfa e beta/fisiologia , Índice de Gravidade de Doença
14.
J Exp Med ; 214(4): 991-1010, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28254948

RESUMO

Stimulator of IFN genes (STING) is a central adaptor protein that mediates the innate immune responses to DNA virus infection. Although ubiquitination is essential for STING function, how the ubiquitination/deubiquitination system is regulated by virus infection to control STING activity remains unknown. In this study, we found that USP21 is an important deubiquitinating enzyme for STING and that it negatively regulates the DNA virus-induced production of type I interferons by hydrolyzing K27/63-linked polyubiquitin chain on STING. HSV-1 infection recruited USP21 to STING at late stage by p38-mediated phosphorylation of USP21 at Ser538. Inhibition of p38 MAPK enhanced the production of IFNs in response to virus infection and protected mice from lethal HSV-1 infection. Thus, our study reveals a critical role of p38-mediated USP21 phosphorylation in regulating STING-mediated antiviral functions and identifies p38-USP21 axis as an important pathway that DNA virus adopts to avoid innate immunity responses.


Assuntos
Vírus de DNA/imunologia , Proteínas de Membrana/fisiologia , Ubiquitina Tiolesterase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Chlorocebus aethiops , Células HEK293 , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/fisiologia , Interferon Tipo I/fisiologia , Camundongos , Fosforilação , Células Vero
15.
Handb Exp Pharmacol ; 240: 3-20, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27417432

RESUMO

Noncanonical functions of several nuclear transcription factors in the mitochondria have been gaining exceptional traction over the years. These transcription factors include nuclear hormone receptors like estrogen, glucocorticoid, and thyroid hormone receptors: p53, IRF3, STAT3, STAT5, CREB, NF-kB, and MEF-2D. Mitochondria-localized nuclear transcription factors regulate mitochondrial processes like apoptosis, respiration and mitochondrial transcription albeit being nuclear in origin and having nuclear functions. Hence, the cell permits these multi-stationed transcription factors to orchestrate and fine-tune cellular metabolism at various levels of operation. Despite their ubiquitous distribution in different subcompartments of mitochondria, their targeting mechanism is poorly understood. Here, we review the current status of mitochondria-localized transcription factors and discuss the possible targeting mechanism besides the functional interplay between these factors.


Assuntos
Mitocôndrias/metabolismo , Fatores de Transcrição/fisiologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Humanos , Fator Regulador 3 de Interferon/fisiologia , NF-kappa B/fisiologia , Transporte Proteico , Receptores de Estrogênio/análise , Receptores de Glucocorticoides/análise , Receptores dos Hormônios Tireóideos/análise , Fatores de Transcrição STAT/fisiologia
16.
J Biol Chem ; 291(52): 26794-26805, 2016 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-27810900

RESUMO

Fibrosis, driven by inflammation, marks the transition from benign to progressive stages of chronic liver diseases. Although inflammation promotes fibrogenesis, it is not known whether other events, such as hepatocyte death, are required for the development of fibrosis. Interferon regulatory factor 3 (IRF3) regulates hepatocyte apoptosis and production of type I IFNs. In the liver, IRF3 is activated via Toll-like receptor 4 (TLR4) signaling or the endoplasmic reticulum (ER) adapter, stimulator of interferon genes (STING). We hypothesized that IRF3-mediated hepatocyte death is an independent determinant of chemically induced liver fibrogenesis. To test this, we performed acute or chronic CCl4 administration to WT and IRF3-, Toll/Interleukin-1R (TIR) domain-containing adapter-inducing interferon-ß (TRIF)-, TRIF-related adaptor molecule (TRAM)-, and STING-deficient mice. We report that acute CCl4 administration to WT mice resulted in early ER stress, activation of IRF3, and type I IFNs, followed by hepatocyte apoptosis and liver injury, accompanied by liver fibrosis upon repeated administration of CCl4 Deficiency of IRF3 or STING prevented hepatocyte death and fibrosis both in acute or chronic CCl4 In contrast, mice deficient in type I IFN receptors or in TLR4 signaling adaptors, TRAM or TRIF, upstream of IRF3, were not protected from hepatocyte death and/or fibrosis, suggesting that the pro-apoptotic role of IRF3 is independent of TLR signaling in fibrosis. Hepatocyte death is required for liver fibrosis with causal involvement of STING and IRF3. Thus, our results identify that IRF3, by its association with STING in the presence of ER stress, couples hepatocyte apoptosis with liver fibrosis and indicate that innate immune signaling regulates outcomes of liver fibrosis via modulation of hepatocyte death in the liver.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Estresse do Retículo Endoplasmático , Hepatócitos/patologia , Fator Regulador 3 de Interferon/fisiologia , Cirrose Hepática/etiologia , Proteínas de Membrana/fisiologia , Receptor de Interferon alfa e beta/fisiologia , Animais , Tetracloreto de Carbono/toxicidade , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Feminino , Hepatócitos/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
17.
J Leukoc Biol ; 98(1): 119-28, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25896227

RESUMO

The innate immune response is largely initiated by pathogen-responsive activation of the transcription factor IRF3. Among other target genes, IRF3 controls the expression of IFN-ß, which triggers the activation of the transcription factor ISGF3 via the IFNAR. IRF3 and ISGF3 have been reported to control many of the same target genes and together, control the antimicrobial innate-immune program; however, their respective contributions and specificities remain unclear. Here, we used genomic technologies to characterize their specificity in terms of their physical DNA-binding and genetic function. With the use of ChiP-seq and transcriptomic measurements in WT versus ifnar(-/-) versus ifnar(-/-)irf3(-/-) macrophages responding to intracellular dsRNA, we confirmed the known ISGF3 DNA-binding motif and further specified a distinct IRF3 consensus sequence. The functional specificity of IRF3 is particularly pronounced in cytokine/chemokine regulation; yet, even in the control of IFN-ß, that specificity is not absolute. By mathematically modeling IFN-ß production within an abstracted tissue layer, we find that IRF3 versus ISGF3 specificity may be critical to limiting IFN-ß production and ISGF3 activation, temporally and spatially, but that partial overlap in their specificity is tolerable and may enhance the effectiveness of the innate-immune response.


Assuntos
Imunidade Inata/fisiologia , Fator Regulador 3 de Interferon/fisiologia , Fator Gênico 3 Estimulado por Interferon/fisiologia , RNA de Cadeia Dupla/imunologia , Transcrição Gênica/fisiologia , Animais , Imunoprecipitação da Cromatina , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Proc Natl Acad Sci U S A ; 112(7): E710-7, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25646421

RESUMO

Cytosolic DNA-sensing pathways that signal via Stimulator of interferon genes (STING) mediate immunity to pathogens and also promote autoimmune pathology in DNaseII- and DNaseIII-deficient mice. In contrast, we report here that STING potently suppresses inflammation in a model of systemic lupus erythematosus (SLE). Lymphoid hypertrophy, autoantibody production, serum cytokine levels, and other indicators of immune activation were markedly increased in STING-deficient autoimmune-prone mice compared with STING-sufficient littermates. As a result, STING-deficient autoimmune-prone mice had significantly shorter lifespans than controls. Importantly, Toll-like receptor (TLR)-dependent systemic inflammation during 2,6,10,14-tetramethylpentadecane (TMPD)-mediated peritonitis was similarly aggravated in STING-deficient mice. Mechanistically, STING-deficient macrophages failed to express negative regulators of immune activation and thus were hyperresponsive to TLR ligands, producing abnormally high levels of proinflammatory cytokines. This hyperreactivity corresponds to dramatically elevated numbers of inflammatory macrophages and granulocytes in vivo. Collectively these findings reveal an unexpected negative regulatory role for STING, having important implications for STING-directed therapies.


Assuntos
Autoimunidade/fisiologia , Proteínas de Membrana/fisiologia , Animais , Autoanticorpos/biossíntese , Células Dendríticas/imunologia , Regulação da Expressão Gênica/fisiologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/fisiologia , Interferons/fisiologia , Ativação Linfocitária , Proteínas de Membrana/genética , Camundongos
19.
Mol Cell ; 56(6): 719-20, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25526529

RESUMO

In this issue, Xu et al. (2014) show that innate antiviral RIG-I-like receptors (RLR) signaling represses TGF-ß-induced growth inhibition, epithelial-mesenchyme transition (EMT), and regulatory T cell (Treg) differentiation via IRF3-mediated Smads function.


Assuntos
Fator Regulador 3 de Interferon/fisiologia , Vírus Sendai/imunologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Humanos
20.
Mol Cell ; 56(6): 723-37, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25526531

RESUMO

TGF-ß signaling is essential in many processes, including immune surveillance, and its dysregulation controls various diseases, including cancer, fibrosis, and inflammation. Studying the innate host defense, which functions in most cell types, we found that RLR signaling represses TGF-ß responses. This regulation is mediated by activated IRF3, using a dual mechanism of IRF3-directed suppression. Activated IRF3 interacts with Smad3, thus inhibiting TGF-ß-induced Smad3 activation and, in the nucleus, disrupts functional Smad3 transcription complexes by competing with coregulators. Consequently, IRF3 activation by innate antiviral signaling represses TGF-ß-induced growth inhibition, gene regulation and epithelial-mesenchymal transition, and the generation of Treg effector lymphocytes from naive CD4(+) lymphocytes. Conversely, silencing IRF3 expression enhances epithelial-mesenchymal transition, TGF-ß-induced Treg cell differentiation upon virus infection, and Treg cell generation in vivo. We present a mechanism of regulation of TGF-ß signaling by the antiviral defense, with evidence for its role in immune tolerance and cancer cell behavior.


Assuntos
Fator Regulador 3 de Interferon/fisiologia , Vírus Sendai/imunologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Diferenciação Celular , Transição Epitelial-Mesenquimal , Células HEK293 , Células Hep G2 , Humanos , Imunidade Inata , Camundongos Endogâmicos C57BL , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Transcrição Gênica , Ativação Transcricional/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...