Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 8(1): 1789, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29176620

RESUMO

Cellular microRNAs (miRNAs) have been shown to regulate hepatitis C virus (HCV) replication, yet a systematic interrogation of the repertoire of miRNAs impacting HCV life cycle is lacking. Here we apply integrative functional genomics strategies to elucidate global HCV-miRNA interactions. Through genome-wide miRNA mimic and hairpin inhibitor phenotypic screens, and miRNA-mRNA transcriptomics analyses, we identify three proviral and nine antiviral miRNAs that interact with HCV. These miRNAs are functionally linked to particular steps of HCV life cycle and related viral host dependencies. Further mechanistic studies demonstrate that miR-25, let-7, and miR-130 families repress essential HCV co-factors, thus restricting viral infection at multiple stages. HCV subverts the antiviral actions of these miRNAs by dampening their expression in cell culture models and HCV-infected human livers. This comprehensive HCV-miRNA interaction map provides fundamental insights into HCV-mediated pathogenesis and unveils molecular pathways linking RNA biology to viral infections.


Assuntos
Hepacivirus/fisiologia , Hepatite C/genética , Interações Hospedeiro-Patógeno/genética , MicroRNAs/metabolismo , Replicação Viral/genética , Adulto , Biópsia , Linhagem Celular Tumoral , Perfilação da Expressão Gênica/métodos , Genômica/métodos , Hepatite C/tratamento farmacológico , Hepatite C/patologia , Hepatite C/virologia , Humanos , Isoquinolinas/uso terapêutico , Fígado/patologia , RNA Mensageiro/metabolismo , Sulfonamidas/uso terapêutico , Adulto Jovem
2.
Gastroenterology ; 152(1): 257-270.e7, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27693511

RESUMO

BACKGROUND & AIMS: The signaling molecule and transcriptional regulator SMAD6, which inhibits the transforming growth factor ß signaling pathway, is required for infection of hepatocytes by hepatitis C virus (HCV). We investigated the mechanisms by which SMAD6 and another inhibitory SMAD (SMAD7) promote HCV infection in human hepatoma cells and hepatocytes. METHODS: We infected Huh7 and Huh7.5.1 cells and primary human hepatocytes with Japanese fulminant hepatitis-1 (JFH1) HCV cell culture system (HCVcc). We then measured HCV binding, intracellular levels of HCV RNA, and expression of target genes. We examined HCV entry in HepG2/microRNA (miR) 122/CD81 cells, which support entry and replication of HCV, were transfected these cells with small interfering RNAs targeting inhibitory SMADs to analyze gene expression profiles. Uptake of labeled low-density lipoprotein (LDL) and cholesterol was measured. Cell surface proteins were quantified by flow cytometry. We obtained liver biopsy samples from 69 patients with chronic HCV infection and 19 uninfected individuals (controls) and measured levels of syndecan 1 (SDC1), SMAD7, and SMAD6 messenger RNAs (mRNAs). RESULTS: Small interfering RNA knockdown of SMAD6 blocked the binding and infection of hepatoma cell lines and primary human hepatocytes by HCV, whereas SMAD6 overexpression increased HCV infection. We found levels of mRNAs encoding heparan sulfate proteoglycans (HSPGs), particularly SDC1 mRNA, and cell surface levels of heparan sulfate to be reduced in cells after SMAD6 knockdown. SMAD6 knockdown also reduced transcription of genes encoding lipoprotein and cholesterol uptake receptors, including the LDL receptor (LDLR), the very LDLR, and the scavenger receptor class B member 1 in hepatocytes; knockdown of SMAD6 also inhibited cell uptake of cholesterol and lipoprotein. Overexpression of SMAD6 increased the expression of these genes. Similar effects were observed with knockdown and overexpression of SMAD7. In addition, HCV infection of cells increased the expression of SMAD6, which required the activity of nuclear factor-κB, but not transforming growth factor ß. Liver tissues from patients with chronic HCV infection had significantly higher levels of SMAD6, SMAD7, and HSPG mRNAs than controls. CONCLUSIONS: In studies of hepatoma cell lines and primary human hepatocytes, we found that infection with HCV leads to activation of nuclear factor-κB, resulting in increased expression of SMAD6 and SMAD7. Up-regulation of SMAD6 and SMAD7 induces the expression of HSPGs, such as SDC1, as well as LDLR, very LDLR, and the scavenger receptor class B member 1, which promote HCV entry and propagation, as well as cellular uptake of cholesterol and lipoprotein.


Assuntos
Hepacivirus/fisiologia , Proteoglicanas de Heparan Sulfato/genética , Hepatócitos , RNA Mensageiro/análise , RNA Viral/análise , Proteína Smad6/genética , Proteína Smad7/genética , Internalização do Vírus , Proteína Morfogenética Óssea 6/metabolismo , Proteína Morfogenética Óssea 7/metabolismo , Colesterol/metabolismo , Técnicas de Silenciamento de Genes , Células Hep G2 , Hepatite C Crônica/genética , Humanos , Lipoproteínas/metabolismo , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Receptores de LDL/genética , Transdução de Sinais , Proteína Smad6/metabolismo , Proteína Smad7/metabolismo , Sindecana-1/genética , Sindecana-1/metabolismo , Transfecção , Regulação para Cima
3.
Proc Natl Acad Sci U S A ; 113(27): 7620-5, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27298373

RESUMO

Hepatitis C virus (HCV) enters the host cell through interactions with a cascade of cellular factors. Although significant progress has been made in understanding HCV entry, the precise mechanisms by which HCV exploits the receptor complex and host machinery to enter the cell remain unclear. This intricate process of viral entry likely depends on additional yet-to-be-defined cellular molecules. Recently, by applying integrative functional genomics approaches, we identified and interrogated distinct sets of host dependencies in the complete HCV life cycle. Viral entry assays using HCV pseudoparticles (HCVpps) of various genotypes uncovered multiple previously unappreciated host factors, including E-cadherin, that mediate HCV entry. E-cadherin silencing significantly inhibited HCV infection in Huh7.5.1 cells, HepG2/miR122/CD81 cells, and primary human hepatocytes at a postbinding entry step. Knockdown of E-cadherin, however, had no effect on HCV RNA replication or internal ribosomal entry site (IRES)-mediated translation. In addition, an E-cadherin monoclonal antibody effectively blocked HCV entry and infection in hepatocytes. Mechanistic studies demonstrated that E-cadherin is closely associated with claudin-1 (CLDN1) and occludin (OCLN) on the cell membrane. Depletion of E-cadherin drastically diminished the cell-surface distribution of these two tight junction proteins in various hepatic cell lines, indicating that E-cadherin plays an important regulatory role in CLDN1/OCLN localization on the cell surface. Furthermore, loss of E-cadherin expression in hepatocytes is associated with HCV-induced epithelial-to-mesenchymal transition (EMT), providing an important link between HCV infection and liver cancer. Our data indicate that a dynamic interplay among E-cadherin, tight junctions, and EMT exists and mediates an important function in HCV entry.


Assuntos
Caderinas/metabolismo , Transição Epitelial-Mesenquimal , Hepacivirus/fisiologia , Hepatite C/virologia , Internalização do Vírus , Linhagem Celular Tumoral , Claudina-1/metabolismo , Regulação da Expressão Gênica , Humanos , Ocludina/metabolismo
4.
Physiol Rep ; 3(1)2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25626872

RESUMO

The functions of smooth muscle sodium calcium exchanger (NCX) in the vasculature are controversial and poorly understood. To determine the possible roles of NCX in the vascular phenotype and function, we developed a novel mouse model (SM-NCX1 KO) in which the smooth muscle-specific NCX type-1 (NCX1) was conditionally knocked out using tamoxifen-inducible Cre-loxP recombination technique. SM-NCX1 KO mice exhibit significantly lower blood pressure and attenuated angiotensin II (Ang II)-salt-induced hypertension (measured by radio telemetry and intra-arterial catheterization). Isolated, pressurized mesenteric small resistance arteries from SM-NCX1 KO mice, compared to control arteries, were characterized by the following: (1) ~90% reduced NCX1 protein expression; (2) impaired functional responses to (i) acute NCX inhibition by SEA0400 or SN-6, (ii) NCX activation by low [Na(+)]o, and (iii) Na(+) pump inhibition by ouabain; (3) attenuated myogenic reactivity; and (4) attenuated vasoconstrictor response to phenylephrine but not Ang II. These results provided direct evidence that arterial NCX1 normally mediates net Ca(2+) influx that helps maintain basal vascular tone in small resistance arteries and blood pressure under physiological conditions. Importantly, NCX1 contributes to blood pressure elevation in Ang II-salt hypertension, possibly by regulating α-adrenergic receptor activation.

5.
PLoS Pathog ; 10(5): e1004163, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24852294

RESUMO

Recent functional genomics studies including genome-wide small interfering RNA (siRNA) screens demonstrated that hepatitis C virus (HCV) exploits an extensive network of host factors for productive infection and propagation. How these co-opted host functions interact with various steps of HCV replication cycle and exert pro- or antiviral effects on HCV infection remains largely undefined. Here we present an unbiased and systematic strategy to functionally interrogate HCV host dependencies uncovered from our previous infectious HCV (HCVcc) siRNA screen. Applying functional genomics approaches and various in vitro HCV model systems, including HCV pseudoparticles (HCVpp), single-cycle infectious particles (HCVsc), subgenomic replicons, and HCV cell culture systems (HCVcc), we identified and characterized novel host factors or pathways required for each individual step of the HCV replication cycle. Particularly, we uncovered multiple HCV entry factors, including E-cadherin, choline kinase α, NADPH oxidase CYBA, Rho GTPase RAC1 and SMAD family member 6. We also demonstrated that guanine nucleotide binding protein GNB2L1, E2 ubiquitin-conjugating enzyme UBE2J1, and 39 other host factors are required for HCV RNA replication, while the deubiquitinating enzyme USP11 and multiple other cellular genes are specifically involved in HCV IRES-mediated translation. Families of antiviral factors that target HCV replication or translation were also identified. In addition, various virologic assays validated that 66 host factors are involved in HCV assembly or secretion. These genes included insulin-degrading enzyme (IDE), a proviral factor, and N-Myc down regulated Gene 1 (NDRG1), an antiviral factor. Bioinformatics meta-analyses of our results integrated with literature mining of previously published HCV host factors allows the construction of an extensive roadmap of cellular networks and pathways involved in the complete HCV replication cycle. This comprehensive study of HCV host dependencies yields novel insights into viral infection, pathogenesis and potential therapeutic targets.


Assuntos
Genômica/métodos , Hepacivirus/fisiologia , Hepatite C/genética , Hepatite C/virologia , Interações Hospedeiro-Patógeno/genética , Replicação Viral/genética , Células Cultivadas/enzimologia , Genes Virais , Humanos , RNA Interferente Pequeno/farmacologia , Receptores Virais/genética , Integração de Sistemas , Montagem de Vírus/genética , Internalização do Vírus , Eliminação de Partículas Virais/genética
6.
Nat Med ; 19(6): 722-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23708292

RESUMO

Hepatitis C virus (HCV) interacts extensively with host factors to not only establish productive infection but also trigger unique pathological processes. Our recent genome-wide siRNA screen demonstrated that IκB kinase-α (IKK-α) is a crucial host factor for HCV. Here we describe a new nuclear factor κB (NF-κB)-independent and kinase-mediated nuclear function of IKK-α in HCV assembly. HCV, through its 3' untranslated region, interacts with DEAD box polypeptide 3, X-linked (DDX3X) to activate IKK-α, which translocates to the nucleus and induces a CBP/p300-mediated transcriptional program involving sterol regulatory element-binding proteins (SREBPs). This innate pathway induces lipogenic genes and enhances core-associated lipid droplet formation to facilitate viral assembly. Chemical inhibitors of IKK-α suppress HCV infection and IKK-α-induced lipogenesis, offering a proof-of-concept approach for new HCV therapeutic development. Our results show that HCV uses a novel mechanism to exploit intrinsic innate responses and hijack lipid metabolism, which may contribute to high chronicity rates and the pathological hallmark of steatosis in HCV infection.


Assuntos
Hepatite C/metabolismo , Quinase I-kappa B/fisiologia , Lipogênese , Montagem de Vírus , Regiões 3' não Traduzidas , Transporte Ativo do Núcleo Celular , RNA Helicases DEAD-box/fisiologia , Hepatite C/virologia , Humanos , NF-kappa B/fisiologia , Fosforilação , Transdução de Sinais , Proteínas de Ligação a Elemento Regulador de Esterol/fisiologia , Fatores de Transcrição de p300-CBP/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...