Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antiviral Res ; 220: 105744, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37944823

RESUMEN

Working with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is restricted to biosafety level III (BSL-3) laboratory. The study used a trans-complementation system consisting of virus-like particles (VLPs) and DNA-launched replicons to generate SARS-CoV-2 single-round infectious particles (SRIPs) with variant-specific spike (S) proteins. S gene of Wuhan-Hu-1 strain (SWH1) or Omicron BA.1 variant (SBA.1), along with the envelope (E) and membrane (M) genes, were cloned into a tricistronic vector, co-expressed in the cells to produce variant-specific S-VLPs. Additionally, the replicon of the WH1-like strain without S, E, M and accessory genes, was engineered under the control by a CMV promoter to produce self-replicating RNAs within VLP-producing cells, led to create SWH1- and SBA.1-based SARS-CoV-2 SRIPs. The SBA.1-based SRIP showed lower virus yield, replication, N protein expression, fusogenicity, and infectivity compared to SWH1-based SRIPs. SBA.1-based SRIP also exhibited intermediate resistance to neutralizing antibodies produced by SWH1-based vaccines, but were effective at infecting cells with low ACE2 expression. Importantly, both S-based SRIPs responded similarly to remdesivir and GC376, with EC50 values ranging from 0.17 to 1.46 µM, respectively. The study demonstrated that this trans-complementation system is a reliable and efficient tool for generating SARS-CoV-2 SRIPs with variant-specific S proteins. SARS-CoV-2 SRIPs, mimicking authentic live viruses, facilitate comprehensive analysis of variant-specific virological characteristics, including antibody neutralization, and drug sensitivity in non-BSL-3 laboratories.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Neutralizantes , Anticuerpos Antivirales
2.
J Virol ; 91(20)2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28768860

RESUMEN

Influenza A virus (IAV) replication relies on an intricate interaction between virus and host cells. How the cellular proteins are usurped for IAV replication remains largely obscure. The aim of this study was to search for novel and potential cellular factors that participate in IAV replication. ZBTB25, a transcription repressor of a variety of cellular genes, was identified by an RNA interference (RNAi) genomic library screen. Depletion of ZBTB25 significantly reduced IAV production. Conversely, overexpression of ZBTB25 enhanced it. ZBTB25 interacted with the viral RNA-dependent RNA polymerase (RdRp) protein and modulated its transcription activity. In addition, ZBTB25 also functioned as a viral RNA (vRNA)-binding protein, binding preferentially to the U-rich sequence within the 5' untranslated region (UTR) of vRNA. Both protein-protein and protein-RNA interactions involving ZBTB25 facilitated viral RNA transcription and replication. In addition, ZBTB25 suppressed interferon production, further enhancing viral replication. ZBTB25-associated functions required an intact zinc finger domain and posttranslational SUMO-1 modification of ZBTB25. Furthermore, treatment with disulfiram (a zinc ejector) of ZBTB25-overexpressing cells showed significantly reduced IAV production as a result of reduced RNA synthesis. Our findings indicate that IAV usurps ZBTB25 for IAV RNA synthesis and serves as a novel and potential therapeutic antiviral target.IMPORTANCE IAV-induced seasonal influenza causes severe illness and death in high-risk populations. However, IAV has developed resistance to current antiviral drugs due to its high mutation rate. Therefore, development of drugs targeting cellular factors required for IAV replication is an attractive alternative for IAV therapy. Here, we discovered a cellular protein, ZBTB25, that enhances viral RdRp activity by binding to both viral RdRp and viral RNA to stimulate viral RNA synthesis. A unique feature of ZBTB25 in the regulation of viral replication is its dual transcription functions, namely, promoting viral RNA transcription through binding to the U-rich region of vRNA and suppressing cellular interferon production. ZBTB25 contains a zinc finger domain that is required for RNA-inhibitory activity by chelating zinc ions. Disulfiram treatment disrupts the zinc finger functions, effectively repressing IAV replication. Based on our findings, we demonstrate that ZBTB25 regulates IAV RNA transcription and replication and serves as a promising antiviral target for IAV treatment.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Interacciones Huésped-Patógeno , Virus de la Influenza A/genética , Proteínas Nucleares/metabolismo , Transcripción Genética , Zinc/metabolismo , Células A549 , Antivirales/farmacología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Disulfiram/farmacología , Células HEK293 , Humanos , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/enzimología , Virus de la Influenza A/fisiología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Unión Proteica , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Proteína SUMO-1/metabolismo , Replicación Viral/efectos de los fármacos
3.
mBio ; 8(3)2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28536288

RESUMEN

Influenza A virus (IAV) RNA segments are individually packaged with viral nucleoprotein (NP) and RNA polymerases to form a viral ribonucleoprotein (vRNP) complex. We previously reported that NP is a monoubiquitinated protein which can be deubiquitinated by a cellular ubiquitin protease, USP11. In this study, we identified an E3 ubiquitin ligase, CNOT4 (Ccr4-Not transcription complex subunit 4), which can ubiquitinate NP. We found that the levels of viral RNA, protein, viral particles, and RNA polymerase activity in CNOT4 knockdown cells were lower than those in the control cells upon IAV infection. Conversely, overexpression of CNOT4 rescued viral RNP activity. In addition, CNOT4 interacted with the NP in the cell. An in vitro ubiquitination assay also showed that NP could be ubiquitinated by in vitro-translated CNOT4, but ubiquitination did not affect the protein stability of NP. Significantly, CNOT4 increased NP ubiquitination, whereas USP11 decreased it. Mass spectrometry analysis of ubiquitinated NP revealed multiple ubiquitination sites on the various lysine residues of NP. Three of these, K184, K227, and K273, are located on the RNA-binding groove of NP. Mutations of these sites to arginine reduced viral RNA replication. These results indicate that CNOT4 is a ubiquitin ligase of NP, and ubiquitination of NP plays a positive role in viral RNA replication.IMPORTANCE Influenza virus, particularly influenza A virus, causes severe and frequent outbreaks among human and avian species. Finding potential target sites for antiviral agents is of utmost importance from the public health point of view. We previously found that viral nucleoprotein (NP) is ubiquitinated, and ubiquitination enhances viral RNA replication. In this study, we found a cellular ubiquitin ligase, CNOT4, capable of ubiquitinating NP. The ubiquitination sites are scattered on the surface of the NP molecule, which is critical for RNA replication. CNOT4 and a ubiquitin protease, USP11, together regulate the extent of NP ubiquitination and thereby the efficiency of RNA replication. This study thus identifies a potential antiviral target site and reveals a novel posttranslational mechanism for regulating viral replication. This represents a novel finding in the literature of influenza virus research.


Asunto(s)
Interacciones Huésped-Patógeno , ARN Viral/biosíntesis , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación , Proteínas del Núcleo Viral/metabolismo , Replicación Viral , Animales , Línea Celular , Perros , Humanos , Espectrometría de Masas , Proteínas de la Nucleocápside , Mapeo de Interacción de Proteínas , Tioléster Hidrolasas/metabolismo
4.
J Virol ; 89(22): 11245-55, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311876

RESUMEN

UNLABELLED: Influenza A virus (IAV) undergoes RNA transcription by a unique capped-mRNA-dependent transcription, which is carried out by the viral RNA-dependent RNA polymerase (RdRp), consisting of the viral PA, PB1, and PB2 proteins. However, how the viral RdRp utilizes cellular factors for virus transcription is not clear. Previously, we conducted a genome-wide pooled short hairpin RNA (shRNA) screen to identify host factors important for influenza A virus replication. Ribosomal RNA processing 1 homolog B (RRP1B) was identified as one of the candidates. RRP1B is a nucleolar protein involved in ribosomal biogenesis. Upon IAV infection, part of RRP1B was translocated from the nucleolus to the nucleoplasm, where viral RNA synthesis likely takes place. The depletion of RRP1B significantly reduced IAV mRNA transcription in a minireplicon assay and in virus-infected cells. Furthermore, we showed that RRP1B interacted with PB1 and PB2 of the RdRp and formed a coimmunoprecipitable complex with RdRp. The depletion of RRP1B reduced the amount of capped mRNA in the RdRp complex. Taken together, these findings indicate that RRP1B is a host factor essential for IAV transcription and provide a target for new antivirals. IMPORTANCE: Influenza virus is an important human pathogen that causes significant morbidity and mortality and threatens the human population with epidemics and pandemics every year. Due to the high mutation rate of the virus, antiviral drugs targeting viral proteins might ultimately lose their effectiveness. An alternative strategy that explores the genetic stability of host factors indispensable for influenza virus replication would thus be desirable. Here, we characterized the rRNA processing 1 homolog B (RRP1B) protein as an important cellular factor for influenza A virus transcription. We showed that silencing RRP1B hampered viral RNA-dependent RNA polymerase (RdRp) activity, which is responsible for virus transcription and replication. Furthermore, we reported that RRP1B is crucial for RdRp binding to cellular capped mRNA, which is a critical step of virus transcription. Our study not only provides a deeper understanding of influenza virus-host interplay, but also suggests a potential target for antiviral drug development.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas Cromosómicas no Histona/fisiología , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , ARN Mensajero/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Transcripción Genética/genética , Transporte Activo de Núcleo Celular/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular , Proteínas Cromosómicas no Histona/genética , Perros , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Operón Lac/genética , Células de Riñón Canino Madin Darby , Interferencia de ARN , ARN Interferente Pequeño , ARN Viral , Replicación Viral/genética
5.
J Clin Bioinforma ; 5: 2, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25745555

RESUMEN

Genome-wide RNA interference (RNAi) screening is an emerging and powerful technique for genetic screens, which can be divided into arrayed RNAi screen and pooled RNAi screen/selection based on different screening strategies. To date, several genome-wide RNAi screens have been successfully performed to identify host factors essential for influenza virus replication. However, the host factors identified by different research groups are not always consistent. Taking influenza virus screens as an example, we found that a number of screening parameters may directly or indirectly influence the primary hits identified by the screens. This review highlights the differences among the published genome-wide screening approaches and offers recommendations for performing a good pooled shRNA screen/selection.

6.
J Virol ; 89(7): 3671-82, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25589657

RESUMEN

UNLABELLED: Influenza A virus (IAV) depends on cellular factors to complete its replication cycle; thus, investigation of the factors utilized by IAV may facilitate antiviral drug development. To this end, a cellular transcriptional repressor, DR1, was identified from a genome-wide RNA interference (RNAi) screen. Knockdown (KD) of DR1 resulted in reductions of viral RNA and protein production, demonstrating that DR1 acts as a positive host factor in IAV replication. Genome-wide transcriptomic analysis showed that there was a strong induction of interferon-stimulated gene (ISG) expression after prolonged DR1 KD. We found that beta interferon (IFN-ß) was induced by DR1 KD, thereby activating the JAK-STAT pathway to turn on ISG expression, which led to a strong inhibition of IAV replication. This result suggests that DR1 in normal cells suppresses IFN induction, probably to prevent undesired cytokine production, but that this suppression may create a milieu that favors IAV replication once cells are infected. Furthermore, biochemical assays of viral RNA replication showed that DR1 KD suppressed viral RNA replication. We also showed that DR1 associated with all three subunits of the viral RNA-dependent RNA polymerase (RdRp) complex, indicating that DR1 may interact with individual components of the viral RdRp complex to enhance viral RNA replication. Thus, DR1 may be considered a novel host susceptibility gene for IAV replication via a dual mechanism, not only suppressing the host defense to indirectly favor IAV replication but also directly facilitating viral RNA replication. IMPORTANCE: Investigations of virus-host interactions involved in influenza A virus (IAV) replication are important for understanding viral pathogenesis and host defenses, which may manipulate influenza virus infection or prevent the emergence of drug resistance caused by a high error rate during viral RNA replication. For this purpose, a cellular transcriptional repressor, DR1, was identified from a genome-wide RNAi screen as a positive regulator in IAV replication. In the current studies, we showed that DR1 suppressed the gene expression of a large set of host innate immunity genes, which indirectly facilitated IAV replication in the event of IAV infection. Besides this scenario, DR1 also directly enhanced the viral RdRp activity, likely through associating with individual components of the viral RdRp complex. Thus, DR1 represents a novel host susceptibility gene for IAV replication via multiple functions, not only suppressing the host defense but also enhancing viral RNA replication. DR1 may be a potential target for drug development against influenza virus infection.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Inmunidad Innata , Virus de la Influenza A/fisiología , Fosfoproteínas/metabolismo , ARN Viral/biosíntesis , Factores de Transcripción/metabolismo , Replicación Viral , Animales , Línea Celular , Perros , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Interferón beta/inmunología
7.
PLoS One ; 9(6): e99022, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24905011

RESUMEN

Nonstructural protein 5A (NS5A) of hepatitis C virus (HCV) serves dual functions in viral RNA replication and virus assembly. Here, we demonstrate that HCV replication complex along with NS5A and Core protein was transported to the lipid droplet (LD) through microtubules, and NS5A-Core complexes were then transported from LD through early-to-late endosomes to the plasma membrane via microtubules. Further studies by cofractionation analysis and immunoelectron microscopy of the released particles showed that NS5A-Core complexes, but not NS4B, were present in the low-density fractions, but not in the high-density fractions, of the HCV RNA-containing virions and associated with the internal virion core. Furthermore, exosomal markers CD63 and CD81 were also detected in the low-density fractions, but not in the high-density fractions. Overall, our results suggest that HCV NS5A is associated with the core of the low-density virus particles which exit the cell through a preexisting endosome/exosome pathway and may contribute to HCV natural infection.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/metabolismo , Gotas Lipídicas/virología , Proteínas del Núcleo Viral/metabolismo , Proteínas no Estructurales Virales/metabolismo , Línea Celular , Exosomas/metabolismo , Exosomas/patología , Exosomas/virología , Hepatitis C/patología , Interacciones Huésped-Patógeno , Humanos , Gotas Lipídicas/metabolismo , Gotas Lipídicas/patología , Microtúbulos/metabolismo , Microtúbulos/patología , Microtúbulos/virología , Transporte de Proteínas
8.
Proc Natl Acad Sci U S A ; 110(43): 17516-21, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24101521

RESUMEN

Influenza viruses, like other viruses, rely on host factors to support their life cycle as viral proteins usually "hijack," or collaborate with, cellular proteins to execute their functions. Identification and understanding of these factors can increase the knowledge of molecular mechanisms manipulated by the viruses and facilitate development of antiviral drugs. To this end, we developed a unique genome-wide pooled shRNA screen to search for cellular factors important for influenza A virus (IAV) replication. We identified an E3 ubiquitin ligase, Itch, as an essential factor for an early step in the viral life cycle. In Itch knockdown cells, the incorporation of viral ribonucleoprotein complex into endosomes was normal, but its subsequent release from endosomes and transport to the nucleus was retarded. In addition, upon virus infection, Itch was phosphorylated and recruited to the endosomes, where virus particles were located. Furthermore, Itch interacted with viral M1 protein and ubiquitinated M1 protein. Collectively, our findings unravel a critical role of Itch in mediating IAV release from the endosome and offer insights into the mechanism for IAV uncoating during virus entry. These findings also highlight the feasibility of pooled RNAi screening for exploring the cellular cofactors of lytic viruses.


Asunto(s)
Endosomas/metabolismo , Virus de la Influenza A/crecimiento & desarrollo , Interferencia de ARN , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Internalización del Virus , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Endosomas/virología , Genoma Humano/genética , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/metabolismo , Virus de la Influenza A/fisiología , Microscopía Fluorescente , Fosforilación , Unión Proteica , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas de la Matriz Viral/metabolismo , Replicación Viral
9.
PLoS Pathog ; 8(6): e1002766, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22792060

RESUMEN

For Hepatitis C virus (HCV), initiation of translation is cap-independently mediated by its internal ribosome entry site (IRES). Unlike other IRES-containing viruses that shut off host cap-dependent translation, translation of HCV coexists with that of the host. How HCV IRES-mediated translation is regulated in the infected cells remains unclear. Here, we show that the intracellular level of 40S ribosomal subunit plays a key role in facilitating HCV translation over host translation. In a loss-of-function screen, we identified small subunit ribosomal protein 6 (RPS6) as an indispensable host factor for HCV propagation. Knockdown of RPS6 selectively repressed HCV IRES-mediated translation, but not general translation. Such preferential suppression of HCV translation correlated well with the reduction of the abundance of 40S ribosomal subunit following knockdown of RPS6 or other RPS genes. In contrast, reduction of the amount of ribosomal proteins of the 60S subunit did not produce similar effects. Among the components of general translation machineries, only knockdowns of RPS genes caused inhibitory effects on HCV translation, pointing out the unique role of 40S subunit abundance in HCV translation. This work demonstrates an unconventional notion that the translation initiation of HCV and host possess different susceptibility toward reduction of 40S ribosomal subunit, and provides a model of selective modulation of IRES-mediated translation through manipulating the level of 40S subunit.


Asunto(s)
Hepacivirus/fisiología , Iniciación de la Cadena Peptídica Traduccional/fisiología , Proteína S6 Ribosómica/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Replicación Viral/fisiología , Línea Celular , Técnicas de Silenciamiento del Gen , Hepatitis C/genética , Hepatitis C/metabolismo , Humanos , Proteína S6 Ribosómica/genética , Subunidades Ribosómicas Grandes de Eucariotas/genética , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/genética
10.
J Virol ; 86(8): 4139-50, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301157

RESUMEN

The hepatitis C virus (HCV) RNA replicates in hepatic cells by forming a replication complex on the lipid raft (detergent-resistant membrane [DRM]). Replication complex formation requires various viral nonstructural (NS) proteins as well as host cellular proteins. In our previous study (C. K. Lai, K. S. Jeng, K. Machida, and M. M. Lai, J. Virol. 82:8838-8848, 2008), we found that a cellular protein, annexin A2 (Anxa2), interacts with NS3/NS4A. Since NS3/NS4A is a membranous protein and Anxa2 is known as a lipid raft-associated scaffold protein, we postulate that Anxa2 helps in the formation of the HCV replication complex on the lipid raft. Further studies showed that Anxa2 was localized at the HCV-induced membranous web and interacted with NS4B, NS5A, and NS5B and colocalized with them in the perinuclear region. The silencing of Anxa2 decreased the formation of membranous web-like structures and viral RNA replication. Subcellular fractionation and bimolecular fluorescence complementation analysis revealed that Anxa2 was partially associated with HCV at the lipid raft enriched with phosphatidylinositol-4-phosphate (PI4P) and caveolin-2. Further, the overexpression of Anxa2 in HCV-nonsusceptible HEK293 cells caused the enrichment of HCV NS proteins in the DRM fraction and increased the colony-forming ability of the HCV replicon. Since Anxa2 is known to induce the formation of the lipid raft microdomain, we propose that Anxa2 recruits HCV NS proteins and enriches them on the lipid raft to form the HCV replication complex.


Asunto(s)
Anexina A2/metabolismo , Hepacivirus/metabolismo , Microdominios de Membrana/metabolismo , Replicación Viral , Anexina A2/genética , Caveolina 2/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/virología , Hepacivirus/genética , Humanos , Microdominios de Membrana/ultraestructura , Fosfatos de Fosfatidilinositol/metabolismo , Unión Proteica , Transporte de Proteínas , ARN Viral/biosíntesis , ARN Viral/genética , Proteínas no Estructurales Virales/metabolismo
11.
PLoS One ; 7(1): e30360, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22276183

RESUMEN

Several studies have demonstrated that cytokine-mediated noncytopathic suppression of hepatitis B virus (HBV) replication may provide an alternative therapeutic strategy for the treatment of chronic hepatitis B infection. In our previous study, we showed that transforming growth factor-beta1 (TGF-ß1) could effectively suppress HBV replication at physiological concentrations. Here, we provide more evidence that TGF-ß1 specifically diminishes HBV core promoter activity, which subsequently results in a reduction in the level of viral pregenomic RNA (pgRNA), core protein (HBc), nucleocapsid, and consequently suppresses HBV replication. The hepatocyte nuclear factor 4alpha (HNF-4α) binding element(s) within the HBV core promoter region was characterized to be responsive for the inhibitory effect of TGF-ß1 on HBV regulation. Furthermore, we found that TGF-ß1 treatment significantly repressed HNF-4α expression at both mRNA and protein levels. We demonstrated that RNAi-mediated depletion of HNF-4α was sufficient to reduce HBc synthesis as TGF-ß1 did. Prevention of HNF-4α degradation by treating with proteasome inhibitor MG132 also prevented the inhibitory effect of TGF-ß1. Finally, we confirmed that HBV replication could be rescued by ectopic expression of HNF-4α in TGF-ß1-treated cells. Our data clarify the mechanism by which TGF-ß1 suppresses HBV replication, primarily through modulating the expression of HNF-4α gene.


Asunto(s)
Virus de la Hepatitis B/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Replicación Viral/efectos de los fármacos , Northern Blotting , Western Blotting , Línea Celular , Inhibidores de Cisteína Proteinasa/farmacología , Ensayo de Cambio de Movilidad Electroforética , Células Hep G2 , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Humanos , Leupeptinas/farmacología , Nucleocápside/genética , Nucleocápside/metabolismo , Regiones Promotoras Genéticas/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo
12.
J Virol ; 86(3): 1739-49, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22130530

RESUMEN

Hepatitis C virus (HCV) reorganizes intracellular membranes to establish sites of replication. How viral and cellular proteins target, bind, and rearrange specific membranes into the replication factory remains a mystery. We used a lentivirus-based RNA interference (RNAi) screening approach to identify the potential cellular factors that are involved in HCV replication. A protein with membrane-deforming activity, proline-serine-threonine phosphatase-interacting protein 2 (PSTPIP2), was identified as a potential factor. Knockdown of PSTPIP2 in HCV subgenomic replicon-harboring and HCV-infected cells was associated with the reduction of HCV protein and RNA expression. PSTPIP2 was localized predominantly in detergent-resistant membranes (DRMs), which contain the RNA replication complex. PSTPIP2 knockdown caused a significant reduction of the formation of HCV- and NS4B-induced membranous webs. A PSTPIP2 mutant defective in inducing membrane curvature failed to support HCV replication, confirming that the membrane-deforming ability of PSTPIP2 is essential for HCV replication. Taking these results together, we suggest that PSTPIP2 facilitates membrane alterations and is a key player in the formation of the membranous web, which is the site of the HCV replication complex.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Hepacivirus/fisiología , Replicación Viral , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Hepacivirus/enzimología , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
13.
J Virol ; 85(20): 10561-71, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21835792

RESUMEN

Autophagy has been shown to facilitate replication or production of hepatitis C virus (HCV); nevertheless, how HCV induces autophagy remains unclear. Here, we demonstrate that HCV nonstructural protein 4B (NS4B) alone can induce autophagy signaling; amino acid residues 1 to 190 of NS4B are sufficient for this induction. Further studies showed that the phosphorylation levels of S6K and 4E-BP1 were not altered, suggesting that the mTOR/S6 kinase pathway and mTOR/4E-BP1 pathway did not contribute to NS4B- or HCV-induced autophagy. Inhibition of Rab5 function by silencing Rab5 or overexpressing dominant-negative Rab5 mutant (S34N) resulted in significant reduction of NS4B- or HCV-induced autophagic vesicle formation. Moreover, the autophagy induction was impaired by inhibition of class III phosphoinositide 3-kinase (PI 3-kinase) Vps34 function. Finally, the coimmunoprecipitation assay indicated that NS4B formed a complex with Rab5 and Vps34, supporting the notion that Rab5 and Vps34 are involved in NS4B-induced autophagy. Taken together, these results not only reveal a novel role of NS4B in autophagy but also offer a clue to the mechanism of HCV-induced autophagy.


Asunto(s)
Autofagia , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno , Proteínas no Estructurales Virales/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Línea Celular , Silenciador del Gen , Humanos , Inmunoprecipitación , Proteínas Mutantes/metabolismo , Unión Proteica , Proteínas de Unión al GTP rab5/antagonistas & inhibidores , Proteínas de Unión al GTP rab5/genética
14.
J Virol ; 85(16): 7954-64, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21632751

RESUMEN

Sequences in the 5' untranslated region (5'UTR) of hepatitis C virus (HCV) RNA is important for modulating both translation and RNA replication. The translation of the HCV genome depends on an internal ribosome entry site (IRES) located within the 341-nucleotide 5'UTR, while RNA replication requires a smaller region. A question arises whether the replication and translation functions require different regions of the 5'UTR and different sets of RNA-binding proteins. Here, we showed that the 5'-most 157 nucleotides of HCV RNA is the minimum 5'UTR for RNA replication, and it partially overlaps with the IRES. Stem-loops 1 and 2 of the 5'UTR are essential for RNA replication, whereas stem-loop 1 is not required for translation. We also found that poly(C)-binding protein 2 (PCBP2) bound to the replication region of the 5'UTR and associated with detergent-resistant membrane fractions, which are the sites of the HCV replication complex. The knockdown of PCBP2 by short hairpin RNA decreased the amounts of HCV RNA and nonstructural proteins. Antibody-mediated blocking of PCBP2 reduced HCV RNA replication in vitro, indicating that PCBP2 is directly involved in HCV RNA replication. Furthermore, PCBP2 knockdown reduced IRES-dependent translation preferentially from a dual reporter plasmid, suggesting that PCBP2 also regulated IRES activity. These findings indicate that PCBP2 participates in both HCV RNA replication and translation. Moreover, PCBP2 interacts with HCV 5'- and 3'UTR RNA fragments to form an RNA-protein complex and induces the circularization of HCV RNA, as revealed by electron microscopy. This study thus demonstrates the mechanism of the participation of PCBP2 in HCV translation and replication and provides physical evidence for HCV RNA circularization through 5'- and 3'UTR interaction.


Asunto(s)
Regiones no Traducidas 5' , Hepacivirus/genética , Hepacivirus/fisiología , ARN Viral/biosíntesis , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Replicación Viral , Anticuerpos Monoclonales , Secuencia de Bases , Línea Celular Tumoral , Electroporación , Genes Reporteros , Genoma Viral , Hepacivirus/crecimiento & desarrollo , Humanos , Secuencias Invertidas Repetidas/genética , Microscopía Electrónica , Plásmidos , Biosíntesis de Proteínas , Interferencia de ARN , ARN Circular , ARN Interferente Pequeño , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Replicación Viral/genética
15.
J Virol ; 85(13): 6618-28, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21507966

RESUMEN

SUMOylation is an important posttranslational modification for regulation of cellular functions and viral replication. Here, we report that protein SUMOylation regulates the replication of influenza A virus at the steps of viral maturation and assembly. Knocking down the SUMO-conjugating enzyme Ubc9 resulted in the reduction of virus production. Dissection of the virus life cycle revealed that SUMOylation is involved in the processes of virus maturation and assembly. The viral matrix protein M1 is SUMOylated at K242. A virus carrying the SUMO-defective M1 produced a lower titer of virus, while its viral proteins and viral RNA (vRNA) accumulated in the cells. Furthermore, the mechanistic studies showed that the SUMOylation of M1 is required for the interaction between M1 and viral RNP (vRNP) to form the M1-vRNP complex. The lack of M1 SUMOylation prevented the nuclear export of vRNP and subsequent viral morphogenesis. Taken together, our findings elucidate that the maturation and assembly of influenza A virus is controlled by the SUMO modification of M1 protein. Therefore, we suggest that M1 can serve as a target for developing a new generation of drugs for flu therapy.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Influenza A/fisiología , Sumoilación , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus , Replicación Viral , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Perros , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Virus de la Influenza A/ultraestructura , Microscopía Electrónica de Transmisión , ARN Viral/genética , ARN Viral/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas Virales/metabolismo
16.
J Biomed Sci ; 18: 96, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22208719

RESUMEN

BACKGROUND: The covalently closed-circular DNA (cccDNA) of hepatitis B virus (HBV) is associated with viral persistence in HBV-infected hepatocytes. However, the regulation of cccDNA and its transcription in the host cells at different growth stages is not well understood. METHODS: We took advantages of a stably HBV-producing cell line, 1.3ES2, and examine the dynamic changes of HBV cccDNA, viral transcripts, and viral replication intermediates in different cellular growth stages. RESULTS: In this study, we showed that cccDNA increased suddenly in the initial proliferation phase of cell growth, probably attributable to its nuclear replenishment by intracellular nucleocapsids. The amount of cccDNA then decreased dramatically in the cells during their exponential proliferation similar to the loss of extrachromosomal plasmid DNA during cell division, after which it accumulated gradually while the host cells grew to confluency. We found that cccDNA was reduced in dividing cells and could be removed when proliferating cells were subjected to long term of lamivudine (3TC) treatment. The amounts of viral replicative intermediates were rapidly reduced in these proliferating cells and were significantly increased after cells reaching confluency. The expression levels of viral transcripts were increased in parallel with the elevated expression of hepatic transcription factors (HNF4α, CEBPα, PPARα, etc.) during cell growth confluency. The HBV transcripts were transcribed from both integrated viral genome and cccDNA, however the transcriptional abilities of cccDNA was less efficient then that from integrated viral genome in all cell growth stages. We also noted increases in the accumulation of intracellular viral particles and the secretion of mature virions as the cells reached confluency and ceased to grow. CONCLUSIONS: Based on the dynamics of HBV replication, we propose that HBV replication is modulated differently in the different stages of cell growth, and can be divided into three phases (initial proliferation phase, exponential proliferation phase and growth confluency phase) according to the cell growth curve. The regulation of cccDNA in different cell growth phase and its importance regarding HBV replication are discussed.


Asunto(s)
ADN Circular/metabolismo , ADN Viral/metabolismo , Virus de la Hepatitis B/genética , Transcripción Genética , Proliferación Celular , Replicación del ADN , Virus de la Hepatitis B/metabolismo , Plásmidos , Replicación Viral/genética
17.
EMBO J ; 29(22): 3879-90, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-20924359

RESUMEN

Influenza A virus RNA replication requires an intricate regulatory network involving viral and cellular proteins. In this study, we examined the roles of cellular ubiquitinating/deubiquitinating enzymes (DUBs). We observed that downregulation of a cellular deubiquitinating enzyme USP11 resulted in enhanced virus production, suggesting that USP11 could inhibit influenza virus replication. Conversely, overexpression of USP11 specifically inhibited viral genomic RNA replication, and this inhibition required the deubiquitinase activity. Furthermore, we showed that USP11 interacted with PB2, PA, and NP of viral RNA replication complex, and that NP is a monoubiquitinated protein and can be deubiquitinated by USP11 in vivo. Finally, we identified K184 as the ubiquitination site on NP and this residue is crucial for virus RNA replication. We propose that ubiquitination/deubiquitination of NP can be manipulated for antiviral therapeutic purposes.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Humana/virología , Nucleoproteínas/metabolismo , ARN Viral/genética , Ubiquitinación , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Línea Celular , Regulación Viral de la Expresión Génica , Genoma Viral , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Datos de Secuencia Molecular , ARN Polimerasa Dependiente del ARN/metabolismo , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo , Proteínas Virales/genética , Replicación Viral
18.
J Virol ; 84(21): 11590-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20739534

RESUMEN

Hepatitis C virus (HCV) assembly is known to occur in juxtaposition to lipid droplets, but the mechanisms of nascent virion transport and release remain poorly understood. Here we demonstrate that HCV core protein targets to early and late endosomes but not to mitochondria or peroxisomes. Further, by employing inhibitors of early and late endosome motility in HCV-infected cells, we demonstrate that the movement of core protein to the early and late endosomes and virus production require an endosome-based secretory pathway. We also observed that this way is independent of that of the internalization of endocytosed virus particles during virus entry.


Asunto(s)
Endosomas/metabolismo , Hepacivirus/fisiología , Proteínas del Núcleo Viral/metabolismo , Liberación del Virus , Transporte de Proteínas , Virión/metabolismo , Internalización del Virus
19.
J Virol ; 84(16): 7983-93, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534861

RESUMEN

Hepatitis C virus (HCV) replication involves many viral and host factors. Here, we employed a lentivirus-based RNA interference (RNAi) screening approach to search for possible cellular factors. By using a kinase-phosphatase RNAi library and an HCV replicon reporter system, we identified a serine-threonine kinase, Polo-like kinase 1 (Plk1), as a potential host factor regulating HCV replication. Knockdown of Plk1 reduced both HCV RNA replication and nonstructural (NS) protein production in both HCV replicon cells and HCV-infected cells while it did not significantly affect host cellular growth or cell cycle. Overexpression of Plk1 in the knockdown cells rescued HCV replication. Interestingly, the ratio between the hyperphosphorylated form (p58) and the basal phosphorylated form (p56) of NS5A was lower in the Plk1 knockdown cells and Plk1 kinase inhibitor-treated cells than in the control groups. Further studies showed that Plk1 could be immunoprecipitated together with NS5A. Both proteins partially colocalized in the perinuclear region. Furthermore, Plk1 could phosphorylate NS5A to both the p58 and p56 forms in an in vitro assay system; the phosphorylation efficiency was comparable to that of the reported casein kinase. Taken together, this study shows that Plk1 is an NS5A phosphokinase and thereby indirectly regulates HCV RNA replication. Because of the differential effects of Plk1 on HCV replication and host cell growth, Plk1 could potentially serve as a target for anti-HCV therapy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Hepacivirus/fisiología , Interacciones Huésped-Patógeno , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular , Técnicas de Silenciamiento del Gen/métodos , Vectores Genéticos , Humanos , Inmunoprecipitación , Lentivirus/genética , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Viral/biosíntesis , Proteínas Virales/biosíntesis , Quinasa Tipo Polo 1
20.
J Virol ; 84(2): 918-27, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19889771

RESUMEN

Hepatitis delta antigen (HDAg) is a nuclear protein that is intimately involved in hepatitis delta virus (HDV) RNA replication. HDAg consists of two protein species, the small form (S-HDAg) and the large form (L-HDAg). Previous studies have shown that posttranslational modifications of S-HDAg, such as phosphorylation, acetylation, and methylation, can modulate HDV RNA replication. In this study, we show that S-HDAg is a small ubiquitin-like modifier 1 (SUMO1) target protein. Mapping data showed that multiple lysine residues are SUMO1 acceptors within S-HDAg. Using a genetic fusion strategy, we found that conjugation of SUMO1 to S-HDAg selectively enhanced HDV genomic RNA and mRNA synthesis but not antigenomic RNA synthesis. This result supports our previous proposition that the cellular machinery involved in the synthesis of HDV antigenomic RNA is different from that for genomic RNA synthesis and mRNA transcription, requiring different modified forms of S-HDAg. Sumoylation represents a new type of modification for HDAg.


Asunto(s)
Genoma Viral , Virus de la Hepatitis Delta/metabolismo , Antígenos de Hepatitis delta/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteína SUMO-1/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Virus de la Hepatitis Delta/genética , Antígenos de Hepatitis delta/genética , Humanos , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , ARN Viral/genética , Proteína SUMO-1/genética , Transfección , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...