Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Virol ; 89(13): 6608-18, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25878102

RESUMO

UNLABELLED: The elongation factor Tu GTP binding domain-containing protein 2 (EFTUD2) was identified as an anti-hepatitis C virus (HCV) host factor in our recent genome-wide small interfering RNA (siRNA) screen. In this study, we sought to further determine EFTUD2's role in HCV infection and investigate the interaction between EFTUD2 and other regulators involved in HCV innate immune (RIG-I, MDA5, TBK1, and IRF3) and JAK-STAT1 pathways. We found that HCV infection decreased the expression of EFTUD2 and the viral RNA sensors RIG-I and MDA5 in HCV-infected Huh7 and Huh7.5.1 cells and in liver tissue from in HCV-infected patients, suggesting that HCV infection downregulated EFTUD2 expression to circumvent the innate immune response. EFTUD2 inhibited HCV infection by inducing expression of the interferon (IFN)-stimulated genes (ISGs) in Huh7 cells. However, its impact on HCV infection was absent in both RIG-I knockdown Huh7 cells and RIG-I-defective Huh7.5.1 cells, indicating that the antiviral effect of EFTUD2 is dependent on RIG-I. Furthermore, EFTUD2 upregulated the expression of the RIG-I-like receptors (RLRs) RIG-I and MDA5 to enhance the innate immune response by gene splicing. Functional experiments revealed that EFTUD2-induced expression of ISGs was mediated through interaction of the EFTUD2 downstream regulators RIG-I, MDA5, TBK1, and IRF3. Interestingly, the EFTUD2-induced antiviral effect was independent of the classical IFN-induced JAK-STAT pathway. Our data demonstrate that EFTUD2 restricts HCV infection mainly through an RIG-I/MDA5-mediated, JAK-STAT-independent pathway, thereby revealing the participation of EFTUD2 as a novel innate immune regulator and suggesting a potentially targetable antiviral pathway. IMPORTANCE: Innate immunity is the first line defense against HCV and determines the outcome of HCV infection. Based on a recent high-throughput whole-genome siRNA library screen revealing a network of host factors mediating antiviral effects against HCV, we identified EFTUD2 as a novel innate immune regulator against HCV in the infectious HCV cell culture model and confirmed that its expression in HCV-infected liver tissue is inversely related to HCV infection. Furthermore, we determined that EFTUD2 exerts its antiviral activity mainly through governing its downstream regulators RIG-I and MDA5 by gene splicing to activate IRF3 and induce classical ISG expression independent of the JAT-STAT signaling pathway. This study broadens our understanding of the HCV innate immune response and provides a possible new antiviral strategy targeting this novel regulator of the innate response.


Assuntos
RNA Helicases DEAD-box/metabolismo , Hepacivirus/imunologia , Imunidade Inata , Fatores Imunológicos/metabolismo , Fatores de Alongamento de Peptídeos/metabolismo , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , Hepatócitos/imunologia , Hepatócitos/virologia , Humanos , Helicase IFIH1 Induzida por Interferon , Receptores Imunológicos
2.
J Hepatol ; 62(5): 1024-32, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25481564

RESUMO

BACKGROUND &/AIMS: The broadly used antiviral cytokine interferon-α (IFNα)'s mechanisms of action against HCV infection are not well understood. We previously identified SART1, a host protein involved in RNA splicing and pre-mRNA processing, as a regulator of IFN's antiviral effects. We hypothesized that SART1 regulates antiviral IFN effector genes (IEGs) through mRNA processing and splicing. METHODS: We performed siRNA knockdown in HuH7.5.1 cells and mRNA-sequencing with or without IFN treatment. Selected gene mRNA variants and their proteins, together with HCV replication, were monitored by qRT-PCR and Western blot in HCV OR6 replicon cells and the JFH1 HCV infectious model. RESULTS: We identified 419 genes with a greater than 2-fold expression difference between Neg siRNA and SART1 siRNA treated cells in the presence or absence of IFN. Bioinformatic analysis identified at least 10 functional pathways. SART1 knockdown reduced classical IFN stimulating genes (ISG) mRNA transcription including MX1 and OAS3. However, SART1 did not affect JAK-STAT pathway gene mRNA expression and IFN stimulated response element (ISRE) signaling. We identified alternative mRNA splicing events for several genes, including EIF4G3, GORASP2, ZFAND6, and RAB6A that contribute to their antiviral effects. EIF4G3 and GORASP2 were also confirmed to have anti-HCV effect. CONCLUSIONS: The spliceosome factor SART1 is not IFN-inducible but is an IEG. SART1 exerts its anti-HCV action through direct transcriptional regulation for some ISGs and alternative splicing for others, including EIF4G3, GORASP2. SART1 does not have an effect on IFN receptor or canonical signal transduction components. Thus, SART1 regulates ISGs using a novel, non-classical mechanism.


Assuntos
Antígenos de Neoplasias/genética , Hepacivirus/fisiologia , Hepatite C , Interferon-alfa , Splicing de RNA/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Spliceossomos/fisiologia , Antivirais/metabolismo , Antivirais/farmacologia , Técnicas de Silenciamento de Genes , Hepatite C/genética , Hepatite C/virologia , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Replicação Viral/fisiologia
3.
Hepatology ; 59(4): 1250-61, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23913866

RESUMO

UNLABELLED: Several genome-wide association studies (GWAS) have identified a genetic polymorphism associated with the gene locus for interleukin 28B (IL28B), a type III interferon (IFN), as a major predictor of clinical outcome in hepatitis C. Antiviral effects of the type III IFN family have previously been shown against several viruses, including hepatitis C virus (HCV), and resemble the function of type I IFN including utilization of the intracellular Janus kinase signal transducer and activator of transcription (JAK-STAT) pathway. Effects unique to IL28B that would distinguish it from IFN-α are not well defined. By analyzing the transcriptomes of primary human hepatocytes (PHH) treated with IFN-α or IL28B, we sought to identify functional differences between IFN-α and IL28B to better understand the roles of these cytokines in the innate immune response. Although our data did not reveal distinct gene signatures, we detected striking kinetic differences between IFN-α and IL28B stimulation for interferon stimulated genes (ISGs). While gene induction was rapid and peaked at 8 hours of stimulation with IFN-α in PHH, IL28B produced a slower, but more sustained increase in gene expression. We confirmed these findings in the human hepatoma cell line Huh7.5.1. Interestingly, in HCV-infected cells the rapid response after stimulation with IFN-α was blunted, and the induction pattern resembled that caused by IL28B. CONCLUSION: The kinetics of gene induction are fundamentally different for stimulations with either IFN-α or IL28B in hepatocytes, suggesting distinct roles of these cytokines within the immune response. Furthermore, the observed differences are substantially altered by infection with HCV.


Assuntos
Carcinoma Hepatocelular/epidemiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hepatite C/epidemiologia , Hepatócitos/metabolismo , Interferon-alfa/farmacologia , Interleucinas/farmacologia , Neoplasias Hepáticas/epidemiologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Comorbidade , Relação Dose-Resposta a Droga , Hepatite C/metabolismo , Hepatite C/patologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Interferons , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Fosforilação , Fator de Transcrição STAT1/metabolismo , Fatores de Tempo , Transcriptoma/efeitos dos fármacos
4.
J Hepatol ; 59(5): 942-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23831117

RESUMO

BACKGROUND & AIMS: Hepatitis C virus (HCV) is a major cause of chronic liver disease worldwide. The biological and therapeutic importance of host cellular cofactors for viral replication has been recently appreciated. Here we examined the roles of SNF1/AMP kinase-related kinase (SNARK) in HCV replication and pathogenesis. METHODS: The JFH1 infection system and the full-length HCV replicon OR6 cell line were used. Gene expression was knocked down by siRNAs. SNARK mutants were created by site-directed mutagenesis. Intracellular mRNA levels were measured by qRT-PCR. Endogenous and overexpressed proteins were detected by Western blot analysis and immunofluorescence. Transforming growth factor (TGF)-ß signaling was monitored by a luciferase reporter construct. Liver biopsy samples from HCV-infected patients were analyzed for SNARK expression. RESULTS: Knockdown of SNARK impaired viral replication, which was rescued by wild type SNARK but not by unphosphorylated or kinase-deficient mutants. Knockdown and overexpression studies demonstrated that SNARK promoted TGF-ß signaling in a manner dependent on both its phosphorylation and kinase activity. In turn, chronic HCV replication upregulated the expression of SNARK in patients. Further, the SNARK kinase inhibitor metformin suppressed both HCV replication and SNARK-mediated enhancement of TGF-ß signaling. CONCLUSIONS: Thus reciprocal regulation between HCV and SNARK promotes TGF-ß signaling, a major driver of hepatic fibrogenesis. These findings suggest that SNARK will be an attractive target for the design of novel host-directed antiviral and antifibrotic drugs.


Assuntos
Hepacivirus/fisiologia , Hepatite C/etiologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Replicação Viral/fisiologia , Biópsia , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatite C/fisiopatologia , Humanos , Fígado/patologia , Fígado/virologia , Metformina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/farmacologia
5.
J Virol ; 86(16): 8581-91, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22674974

RESUMO

Responses to alpha interferon (IFN-α)-based treatment are dependent on both host and viral factors and vary markedly among patients infected with different hepatitis C virus (HCV) genotypes (GTs). Patients infected with GT3 viruses consistently respond better to IFN treatment than do patients infected with GT1 viruses. The mechanisms underlying this difference are not well understood. In this study, we sought to determine the effects of HCV NS5A proteins from different genotypes on IFN signaling. We found that the overexpression of either GT1 or GT3 NS5A proteins significantly inhibited IFN-induced IFN-stimulated response element (ISRE) signaling, phosphorylated STAT1 (P-STAT1) levels, and IFN-stimulated gene (ISG) expression compared to controls. GT1 NS5A protein expression exhibited stronger inhibitory effects on IFN signaling than did GT3 NS5A protein expression. Furthermore, GT1 NS5A bound to STAT1 with a higher affinity than did GT3 NS5A. Domain mapping revealed that the C-terminal region of NS5A conferred these inhibitory effects on IFN signaling. The overexpression of HCV NS5A increased HCV replication levels in JFH1-infected cells through the further reduction of levels of P-STAT1, ISRE signaling, and downstream ISG responses. We demonstrated that the overexpression of GT1 NS5A proteins resulted in less IFN responsiveness than did the expression of GT3 NS5A proteins through stronger binding to STAT1. We confirmed that GT1 NS5A proteins exerted stronger IFN signaling inhibition than did GT3 NS5A proteins in an infectious recombinant JFH1 virus. The potent antiviral NS5A inhibitor BMS-790052 did not block NS5A-mediated IFN signaling suppression in an overexpression model, suggesting that NS5A's contributions to replication are independent of its subversive action on IFN. We propose a model in which the binding of the C-terminal region of NS5A to STAT1 leads to decreased levels of P-STAT1, ISRE signaling, and ISG transcription and, ultimately, to preferential GT1 resistance to IFN treatment.


Assuntos
Hepacivirus/patogenicidade , Interações Hospedeiro-Patógeno , Interferon Tipo I/imunologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo , Humanos , Modelos Biológicos , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas
6.
Theranostics ; 13(7): 2210-2225, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37153733

RESUMO

Background: Nonalcoholic steatohepatitis (NASH) is a leading cause of chronic liver diseases worldwide. There is a pressing clinical need to identify potential therapeutic targets for NASH treatment. Thioredoxin interacting protein (Txnip) is a stress responsive gene that has been implicated in the pathogenesis of NASH, but its exact role is not fully understood. Here, we investigated the liver- and gene-specific role of Txnip and its upstream/downstream signaling in the pathogenesis of NASH. Methods and Results: Using four independent NASH mouse models, we found that TXNIP protein abnormally accumulated in NASH mouse livers. A decrease in E3 ubiquitin ligase NEDD4L resulted in impaired TXNIP ubiquitination and its accumulation in the liver. TXNIP protein levels were positively correlated with that of CHOP, a major regulator of ER stress-mediated apoptosis, in NASH mouse liver. Moreover, gain- and loss-of-function studies showed that TXNIP increased protein not mRNA levels of Chop both in vitro and in vivo. Mechanistically, the C-terminus of TXNIP associated with the N-terminus of the α-helix domain of CHOP and decreased CHOP ubiquitination, thus increasing the stability of CHOP protein. Lastly, selective knockdown of Txnip by adenovirus-mediated shRNA (not targets Txnip antisense lncRNA) delivery in the livers of both young and aged NASH mice suppressed the expression of CHOP and its downstream apoptotic pathway, and ameliorated NASH by reducing hepatic apoptosis, inflammation, and fibrosis. Conclusions: Our study revealed a pathogenic role of hepatic TXNIP in NASH and identified a novel NEDD4L-TXNIP-CHOP axis in the pathogenesis of NASH.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fígado/metabolismo , Inflamação/metabolismo , Apoptose , Transdução de Sinais/genética , Camundongos Endogâmicos C57BL , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
7.
J Biol Chem ; 286(4): 2665-74, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21098019

RESUMO

HIV/HCV coinfection leads to accelerated hepatic fibrosis progression, with higher rates of cirrhosis, liver failure, and liver death than does HCV mono-infection. However, the profibrogenic role of HIV on hepatocytes and hepatic stellate cells (HSC) has not been fully clarified. We hypothesized that HIV, HCV induce liver fibrosis through altered regulation of the production of extracellular matrix and matrix metalloproteinases. We examined the fibrogenesis- and fibrolysis-related gene activity in LX2 HSC and Huh7.5.1 cells in the presence of inactivated CXCR4 and CCR5 HIV, as well as HCV JFH1 virus. The role of reactive oxygen species (ROS) upon fibrosis gene expression was assessed using the ROS inhibitor. Fibrosis-related transcripts including procollagen α1(I) (CoL1A), TIMP1, and MMP3 mRNA were measured by qPCR. TIMP1 and MMP3 protein expression were assessed by ELISA. We found that inactivated CXCR4 HIV and CCR5 HIV increased CoL1A, and TIMP1 expression in both HSC and Huh7.5.1 cells; the addition of JFH1 HCV further increased CoL1A and TIMP1 expression. CXCR4 HIV and CCR5 HIV induced ROS production in HSC and Huh7.5.1 cells which was further enhanced by JFH1 HCV. The ROS inhibitor DPI abrogated HIV-and HCV-induced CoL1A and TIMP1 expression. HIV and HCV-induced CoL1A and TIMP1 expression were also blocked by NFκB siRNA. Our data provide further evidence that HIV and HCV independently regulate hepatic fibrosis progression through the generation of ROS; this regulation occurs in an NFκB-dependent fashion. Strategies to limit the viral induction of oxidative stress are warranted to inhibit fibrogenesis.


Assuntos
Infecções por HIV , HIV/metabolismo , Hepacivirus/metabolismo , Hepatite C , Cirrose Hepática , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Hepatite C/complicações , Hepatite C/metabolismo , Hepatite C/patologia , Humanos , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/virologia , Estresse Oxidativo
8.
J Am Chem Soc ; 134(16): 6896-9, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22480142

RESUMO

Hepatitis C virus (HCV) infection has been clinically associated with serum lipid abnormalities, yet our understanding of the effects of HCV on host lipid metabolism and conversely the function of individual lipids in HCV replication remains incomplete. Using liquid chromatography-mass spectrometry metabolite profiling of the HCV JFH1 cell culture infection model, we identified a significant steady-state accumulation of desmosterol, an immediate precursor to cholesterol. Pharmacological inhibition or RNAi-mediated depletion of DHCR7 significantly reduced steady-state HCV protein expression and viral genomic RNA. Moreover, this effect was reversed when cultures were supplemented with exogenous desmosterol. Together, these observations suggest an intimate connection between HCV replication and desmosterol homeostasis and that the enzymes responsible for synthesis of desmosterol may be novel targets for antiviral design.


Assuntos
Antivirais/farmacologia , Desmosterol/farmacologia , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Metabolismo dos Lipídeos/efeitos dos fármacos , Antivirais/química , Antivirais/metabolismo , Células Cultivadas , Desmosterol/química , Desmosterol/metabolismo , Hepacivirus/metabolismo , Hepatite C/metabolismo , Humanos , Replicação Viral/efeitos dos fármacos
9.
J Hepatol ; 56(2): 326-33, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21888876

RESUMO

BACKGROUND & AIMS: The precise mechanisms by which IFN exerts its antiviral effect against HCV have not yet been elucidated. We sought to identify host genes that mediate the antiviral effect of IFN-α by conducting a whole-genome siRNA library screen. METHODS: High throughput screening was performed using an HCV genotype 1b replicon, pRep-Feo. Those pools with replicate robust Z scores ≥2.0 entered secondary validation in full-length OR6 replicon cells. Huh7.5.1 cells infected with JFH1 were then used to validate the rescue efficacy of selected genes for HCV replication under IFN-α treatment. RESULTS: We identified and confirmed 93 human genes involved in the IFN-α anti-HCV effect using a whole-genome siRNA library. Gene ontology analysis revealed that mRNA processing (23 genes, p=2.756e-22), translation initiation (nine genes, p=2.42e-6), and IFN signaling (five genes, p=1.00e-3) were the most enriched functional groups. Nine genes were components of U4/U6.U5 tri-snRNP. We confirmed that silencing squamous cell carcinoma antigen recognized by T cells (SART1), a specific factor of tri-snRNP, abrogates IFN-α's suppressive effects against HCV in both replicon cells and JFH1 infectious cells. We further found that SART1 was not IFN-α inducible, and its anti-HCV effector in the JFH1 infectious model was through regulation of interferon stimulated genes (ISGs) with or without IFN-α. CONCLUSIONS: We identified 93 genes that mediate the anti-HCV effect of IFN-α through genome-wide siRNA screening; 23 and nine genes were involved in mRNA processing and translation initiation, respectively. These findings reveal an unexpected role for mRNA processing in generation of the antiviral state, and suggest a new avenue for therapeutic development in HCV.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/genética , Interações Hospedeiro-Patógeno/genética , Interferon-alfa/farmacologia , Antígenos de Neoplasias/genética , Linhagem Celular , Biologia Computacional , Genoma Humano , Genômica , Hepacivirus/genética , Hepacivirus/fisiologia , Hepatite C Crônica/virologia , Ensaios de Triagem em Larga Escala , Humanos , Farmacogenética , RNA Interferente Pequeno/genética , Receptor de Interferon alfa e beta/genética , Replicon , Ribonucleoproteínas Nucleares Pequenas/genética
10.
J Infect Dis ; 204(4): 609-16, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21791663

RESUMO

BACKGROUND: Hepatitis C virus (HCV) chronically infects >170 million persons worldwide and is a leading cause of cirrhosis and hepatocellular carcinoma. The identification of more effective and better-tolerated agents for treating HCV is a high priority. We have reported elsewhere the discovery of the anti-HCV compound ceestatin using a high-throughput screen of a small molecule library. METHODS: To identify host or viral protein targets in an unbiased fashion, we performed affinity chromatography, using tandem liquid chromatography/mass spectrometry to identify specific potential targets. RESULTS. Ceestatin binds to 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) synthase and irreversibly inhibits HMG-CoA synthase in a dose-dependent manner. Ceestatin's anti-HCV effects are reversed by addition of HMG-CoA, mevalonic acid, or geranylgeraniol. Treatment with small interfering RNA against HMG-CoA synthase led to a substantial reduction in HCV replication, further validating HMG-CoA synthase as an enzyme essential for HCV replication. CONCLUSIONS: Ceestatin therefore exerts its anti-HCV effects through inhibition of HMG-CoA synthase. It may prove useful as an antiviral agent, as a probe to study HCV replication, and as a cholesterol-lowering agent. The logical stepwise process employed to discover the mechanism of action of ceestatin can serve as a general experimental strategy to uncover the targets on which novel uncharacterized anti-HCV compounds act.


Assuntos
Antivirais/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Hepacivirus/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Lactonas/farmacologia , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Cromatografia de Afinidade , Hepacivirus/fisiologia , Humanos , Espectrometria de Massas , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno
11.
J Hepatol ; 55(2): 289-98, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21147189

RESUMO

BACKGROUND & AIMS: The combination of pegylated interferon (IFN) α and ribavirin (RBV) is the standard therapy for patients with chronic HCV infection. However, it produces a sustained virologic response (SVR) in only half of the treated individuals and is associated with significant side effects. Recently, several single-nucleotide polymorphisms (SNPs) near the IL28B locus, also known as IFNλ3, were identified to be strong predictors of SVR in patients receiving PEG-IFN and RBV. We sought to determine whether IL28B was capable of inhibiting HCV replication and to determine the pathway by which IL28B exhibits anti-HCV activity. METHODS: Using the full-length HCV replicon OR6 and the infectious HCV clones JFH1 and Jc1, we assessed the anti-HCV effect of IL28B on HCV and characterized the key steps of the JAK-STAT pathway by real time PCR, luciferase assay, and Western blot. Finally, we evaluated the anti-HCV effect of IL28B in the presence of JAK-STAT pathway inhibitors such as blocking antibodies, a pharmacological inhibitor, and siRNAs. RESULTS: We found that IL28B inhibits HCV replication in a dose- and time-dependent manner. Like IFNα, IL28B induces the phosphorylation of STAT1 and STAT2, ISRE-driven transcription, and expression of known ISGs. The anti-HCV effects of IL28A, IL28B, and IL29 were abrogated by an IL10R2 blocking antibody, a pharmacological inhibitor of JAK1/TYK2, and by siRNA against IL28R1, STAT1, STAT2, and IRF9. CONCLUSIONS: Our data demonstrate that IL28A, IL28B, and IL29 signal through the JAK-STAT pathway to inhibit HCV. These data suggest possible applications of new approaches in HCV treatment.


Assuntos
Hepacivirus/efeitos dos fármacos , Hepacivirus/fisiologia , Interleucinas/farmacologia , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Antivirais/farmacologia , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/antagonistas & inibidores , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Interferons , Interleucinas/genética , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 1/metabolismo , Janus Quinases/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Polimorfismo de Nucleotídeo Único , RNA Interferente Pequeno/genética , Receptores de Interferon/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Transcrição STAT/antagonistas & inibidores , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/antagonistas & inibidores , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais/efeitos dos fármacos , TYK2 Quinase/antagonistas & inibidores , TYK2 Quinase/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
12.
J Hepatol ; 54(4): 612-20, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21146890

RESUMO

BACKGROUND & AIMS: HCV related liver disease is one of the most important complications in persons with HIV, with accelerated fibrosis progression in coinfected persons compared to those with HCV alone. We hypothesized that HCV-HIV coinfection increases HCV related hepatocyte apoptosis and that HCV and HIV influence TRAIL signaling in hepatocytes. METHODS: We analyzed the effect of HIV in JFH1-infected Huh7.5.1 cells. Apoptosis was measured by Caspase-Glo 3/7 assay and Western blotting for cleaved PARP. TRAIL, TRAIL receptor 1 (DR4), and 2 (DR5) mRNA and protein levels were assessed by real-time PCR and Western blot, respectively. We also investigated activation of caspase pathways using caspase inhibitors and assessed expression of Bid and cytochrome C. RESULTS: We found increased caspase 3/7 activity and cleaved PARP in JFH1 HCV-infected Huh7.5.1 cells in the presence of heat-inactivated HIV, compared to Huh7.5.1 cells infected with JFH1 or exposed to heat-inactivated HIV alone. Both DR4 and DR5 mRNA and protein expression were increased in JFH1-infected cells in the presence of inactivated HIV compared to Huh7.5.1 cells infected with JFH1 or exposed to heat-inactivated HIV alone. Pancaspase, caspase-8, and caspase-9 inhibition blocked apoptosis induced by HCV, inactivated HIV, and HCV plus inactivated HIV. A caspase-9 inhibitor blocked apoptosis induced by HCV, HIV, and HCV-HIV comparably to pancaspase and caspase-8 inhibitors. HCV induced the activation of Bid cleavage and cytochrome C release. The addition of HIV substantially augmented this induction. CONCLUSIONS: Our findings indicate that hepatocyte apoptosis is increased in the presence of HCV and HIV compared to HCV or HIV alone, and that this increase is mediated by DR4 and DR5 up-regulation. These results provide an additional mechanism for the accelerated liver disease progression observed in HCV-HIV co-infection.


Assuntos
Infecções por HIV/complicações , HIV-1/patogenicidade , Hepatite C Crônica/complicações , Hepatite C Crônica/patologia , Hepatócitos/patologia , Apoptose , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Sequência de Bases , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Citocromos c/metabolismo , Primers do DNA/genética , Infecções por HIV/genética , Infecções por HIV/metabolismo , Hepatite C Crônica/genética , Hepatite C Crônica/metabolismo , Hepatócitos/metabolismo , Humanos , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
13.
Gastroenterology ; 138(7): 2509-18, 2518.e1, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20230822

RESUMO

BACKGROUND & AIMS: The generation of oxidative stress and transforming growth factor beta1 (TGF-beta1) production play important roles in liver fibrogenesis. We have previously shown that hepatitis C virus (HCV) increases hepatocyte TGF-beta1 expression. However, the mechanisms by which this induction occurs have not been well studied. We explored the possibility that HCV infection regulates TGF-beta1 expression through the generation of reactive oxygen species (ROS), which act through > or =1 of the p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and nuclear factor kappaB (NFkappaB) signaling pathways to induce TGF-beta1 expression. METHODS: We used small molecule inhibitors and short interfering RNAs to knock down these pathways to study the mechanism by which HCV regulates TGF-beta1 production in the infectious JFH1 model. RESULTS: We demonstrated that HCV induces ROS and TGF-beta1 expression. We further found that JFH1 induces the phosphorylation of p38MAPK, JNK, ERK, and NFkappaB. We also found that HCV-mediated TGF-beta1 enhancement occurs through a ROS-induced and p38 MAPK, JNK, ERK1/2, NFkappaB-dependent pathway. CONCLUSIONS: These findings provide further evidence to support the hypothesis that HCV enhances hepatic fibrosis progression through the generation of ROS and induction of TGF-beta1. Strategies to limit the viral induction of oxidative stress appear to be warranted to inhibit fibrogenesis.


Assuntos
Hepacivirus/patogenicidade , NF-kappa B/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Estresse Oxidativo , RNA Interferente Pequeno/genética , Transdução de Sinais , Fator de Crescimento Transformador beta1/genética , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
14.
J Virol ; 84(12): 6060-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375166

RESUMO

We and others have observed that hepatic levels of suppressor of cytokine signaling 3 (SOCS3) are significantly higher in persons with chronic hepatitis C, particularly those who are nonresponders to interferon (IFN) treatment, than in healthy individuals. However, the relationship between SOCS3 and hepatitis C virus (HCV) replication remains unclear. Given its putative role, we hypothesized that SOCS3 is permissive for viral replication. We therefore used the OR6 cell line, which harbors a genotype 1b full-length HCV replicon, and the genotype 2a full-length HCV strain JFH1 infection system to analyze the effects of SOCS3 overexpression and short hairpin RNA (shRNA)-mediated knockdown on HCV replication. We further analyzed the role of mTOR in the effects of SOCS3 by treating selected cells with rapamycin. OR6 cells and JFH1-infected Huh7.5.1 cells expressed significantly less SOCS3 than control cells. Furthermore, inhibition of HCV replication with the HCV protease inhibitor BILN 2061 restored SOCS3 protein levels. SOCS3 overexpression in OR6 cells and JFH1-infected Huh7.5.1 cells resulted in significantly lower HCV replication than that in the control cells, despite SOCS3-related inhibition of STAT1 phosphorylation and type I IFN signaling. In contrast, JFH1-infected cells with stable SOCS3 knockdown expressed higher levels of HCV proteins and RNA than did control cells. SOCS3-targeting shRNA also knocked down mTOR and phospho-mTOR. The mTOR inhibitor rapamycin reversed the inhibitory effects of SOCS3. In independent investigations, SOCS3 unexpectedly suppressed HCV replication in an mTOR-dependent manner. These findings suggest that increased SOCS3 levels consistently observed in chronic IFN nonresponders may reflect a compensatory host antiviral response to persistent infection and that manipulation of SOCS3/mTOR may offer benefit against HCV infection.


Assuntos
Regulação para Baixo , Hepacivirus/fisiologia , Hepatite C Crônica/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Replicação Viral , Linhagem Celular , Hepacivirus/genética , Hepatite C Crônica/genética , Hepatite C Crônica/virologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Serina-Treonina Quinases/genética , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Serina-Treonina Quinases TOR
15.
Nat Chem Biol ; 5(4): 258-65, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19287398

RESUMO

Stepwise differentiation from embryonic stem cells (ESCs) to functional insulin-secreting beta cells will identify key steps in beta-cell development and may yet prove useful for transplantation therapy for diabetics. An essential step in this schema is the generation of pancreatic progenitors--cells that express Pdx1 and produce all the cell types of the pancreas. High-content chemical screening identified a small molecule, (-)-indolactam V, that induces differentiation of a substantial number of Pdx1-expressing cells from human ESCs. The Pdx1-expressing cells express other pancreatic markers and contribute to endocrine, exocrine and duct cells, in vitro and in vivo. Further analyses showed that (-)-indolactam V works specifically at one stage of pancreatic development, inducing pancreatic progenitors from definitive endoderm. This study describes a chemical screening platform to investigate human ESC differentiation and demonstrates the generation of a cell population that is a key milepost on the path to making beta cells.


Assuntos
Carcinógenos/farmacologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Indóis/farmacologia , Células Secretoras de Insulina/citologia , Lactamas/farmacologia , Animais , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células-Tronco Embrionárias/fisiologia , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Transativadores/metabolismo
16.
Nat Chem Biol ; 5(3): 154-6, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19151731

RESUMO

Small-molecule inhibition of extracellular proteins that activate membrane receptors has proven to be extremely challenging. Diversity-oriented synthesis and small-molecule microarrays enabled the discovery of robotnikinin, a small molecule that binds the extracellular Sonic hedgehog (Shh) protein and blocks Shh signaling in cell lines, human primary keratinocytes and a synthetic model of human skin. Shh pathway activity is rescued by small-molecule agonists of Smoothened, which functions immediately downstream of the Shh receptor Patched.


Assuntos
Proteínas Hedgehog/metabolismo , Lactamas/farmacologia , Lactonas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células 3T3 , Animais , Descoberta de Drogas , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Lactamas/metabolismo , Lactonas/metabolismo , Camundongos , Receptores Patched , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
17.
Gastroenterology ; 134(3): 803-11, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18325393

RESUMO

BACKGROUND & AIMS: Human immunodeficiency virus (HIV) coinfection increases hepatitis C virus (HCV)-related progression of hepatic fibrosis, increases HCV persistence, and decreases response rates to interferon-based anti-HCV therapy. It has remained unclear how HIV, a nonhepatotropic virus, accelerates the progression of liver disease by HCV. METHODS: We explored the possibility that circulating HIV and/or its proteins contribute to the pathogenesis of HCV through engagement of extracellular coreceptors on hepatocytes. RESULTS: In this study, we found that inactivated HIV or gp120 increases HCV replication and enhances HCV-regulated transforming growth factor (TGF)-beta1 expression in both a replicon and an infectious model of HCV. This proviral effect of HIV and gp120 on HCV replication is neutralized by antibodies to CCR5 or CXCR4. However, HIV and gp120 did not alter type I interferon-mediated signaling in these HCV models, indicating that HIV regulates HCV replication through an alternative mechanism. Interestingly, we found that human TGF-beta1 also enhanced HCV replication. The effect of HIV on HCV replication was blocked by a neutralizing antibody to TGF-beta1, indicating that its effects on HCV replication are TGF-beta1 dependent. CONCLUSIONS: These results suggest a novel mechanism by which HIV not only enhances HCV replication but also contributes to progression of hepatic fibrosis.


Assuntos
Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/metabolismo , Hepacivirus/metabolismo , Hepatócitos/virologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Anticorpos , Linhagem Celular Tumoral , Fibrose , Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/patogenicidade , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Interferon-alfa/metabolismo , RNA Viral/metabolismo , Receptores CCR5/imunologia , Receptores CCR5/metabolismo , Receptores CXCR4/imunologia , Receptores CXCR4/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta1/imunologia , Replicação Viral
18.
Bioorg Med Chem Lett ; 19(22): 6319-25, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19819139

RESUMO

We report the synthesis and biological activity of a library of aminoalcohol-derived macrocycles from which robotnikinin, a binder to and inhibitor of Sonic Hedgehog, was derived. Using an asymmetric alkylation to set a key stereocenter and an RCM reaction to close the macrocycle, we were able to synthesize compounds for testing. High-throughput screening via small-molecule microarray (SMM) technology led to the discovery of a compound capable of binding ShhN. Follow-up chemistry led to a library of macrocycles with enhanced biological activity relative to the original hit compounds. Differences in ring size and stereochemistry, leading to alterations in the mode of binding, may account for differences in the degree of biological activity. These compounds are the first ones reported that inhibit Shh signaling at the ShhN level.


Assuntos
Proteínas Hedgehog/síntese química , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade , Amino Álcoois/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Conformação Proteica , Proteínas Wnt
19.
World J Gastroenterol ; 22(45): 9954-9965, 2016 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-28018102

RESUMO

AIM: To characterize the role of apolipoprotein B100 (apoB100) in hepatitis C viral (HCV) infection. METHODS: In this study, we utilize a gene editing tool, transcription activator-like effector nucleases (TALENs), to generate human hepatoma cells with a stable genetic deletion of APOB to assess of apoB in HCV. Using infectious cell culture-competent HCV, viral pseudoparticles, replicon models, and lipidomic analysis we determined the contribution of apoB to each step of the viral lifecycle. We further studied the effect of mipomersen, an FDA-approved antisense inhibitor of apoB100, on HCV using in vitro cell-culture competent HCV and determined its impact on viral infectivity with the TCID50 method. RESULTS: We found that apoB100 is indispensable for HCV infection. Using the JFH-1 fully infectious cell-culture competent virus in Huh 7 hepatoma cells with TALEN-mediated gene deletion of apoB (APOB KO), we found a significant reduction in HCV RNA and protein levels following infection. Pseudoparticle and replicon models demonstrated that apoB did not play a role in HCV entry or replication. However, the virus produced by APOB KO cells had significantly diminished infectivity as measured by the TCID-50 method compared to wild-type virus. Lipidomic analysis demonstrated that these virions have a fundamentally altered lipidome, with complete depletion of cholesterol esters. We further demonstrate that inhibition of apoB using mipomersen, an FDA-approved anti-sense oligonucleotide, results in a potent anti-HCV effect and significantly reduces the infectivity of the virus. CONCLUSION: ApoB is required for the generation of fully infectious HCV virions, and inhibition of apoB with mipomersen blocks HCV. Targeting lipid metabolic pathways to impair viral infectivity represents a novel host targeted strategy to inhibit HCV.


Assuntos
Apolipoproteína B-100/genética , Hepatite C/genética , Hepatócitos/metabolismo , RNA Viral/metabolismo , Internalização do Vírus , Apolipoproteína B-100/antagonistas & inibidores , Linhagem Celular , Técnicas de Inativação de Genes , Hepacivirus , Hepatite C/virologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Técnicas In Vitro , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Oligonucleotídeos/farmacologia , Proteínas Virais/metabolismo , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
20.
Sci Rep ; 6: 22487, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26927933

RESUMO

HCV replication disrupts normal endoplasmic reticulum (ER) function and activates a signaling network called the unfolded protein response (UPR). UPR is directed by three ER transmembrane proteins including ATF6, IRE1, and PERK. HCV increases TGF-ß1 and oxidative stress, which play important roles in liver fibrogenesis. HCV has been shown to induce TGF-ß1 through the generation of reactive oxygen species (ROS) and p38 MAPK, JNK, ERK1/2, and NFκB-dependent pathways. However, the relationship between HCV-induced ER stress and UPR activation with TGF-ß1 production has not been fully characterized. In this study, we found that ROS and JNK inhibitors block HCV up-regulation of ER stress and UPR activation. ROS, JNK and IRE1 inhibitors blocked HCV-activated NFκB and TGF-ß1 expression. ROS, ER stress, NFκB, and TGF-ß1 signaling were blocked by JNK specific siRNA. Knockdown IRE1 inhibited JFH1-activated NFκB and TGF-ß1 activity. Knockdown of JNK and IRE1 blunted JFH1 HCV up-regulation of NFκB and TGF-ß1 activation. We conclude that HCV activates NFκB and TGF-ß1 through ROS production and induction of JNK and the IRE1 pathway. HCV infection induces ER stress and the UPR in a JNK-dependent manner. ER stress and UPR activation partially contribute to HCV-induced NF-κB activation and enhancement of TGF-ß1.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Endorribonucleases/metabolismo , Hepacivirus/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Fator 6 Ativador da Transcrição/genética , Linhagem Celular Tumoral , Endorribonucleases/antagonistas & inibidores , Endorribonucleases/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Estresse Oxidativo/fisiologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , eIF-2 Quinase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA