51
|
Li Q, Zhang YY, Chiu S, Hu Z, Lan KH, Cha H, Sodroski C, Zhang F, Hsu CS, Thomas E, Liang TJ. Integrative functional genomics of hepatitis C virus infection identifies host dependencies in complete viral replication cycle. PLoS Pathog 2014; 10:e1004163. [PMID: 24852294 PMCID: PMC4095987 DOI: 10.1371/journal.ppat.1004163] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/21/2014] [Indexed: 12/20/2022] Open
Abstract
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. Hepatitis C virus (HCV) is a positive strand RNA virus that belongs to the Flaviridae family. Chronic HCV infection is a leading cause of end-stage liver disease, which is associated with significant morbidity and mortality in the world. Our recent genome-wide siRNA screen has revealed that HCV depends extensively on host factors for efficient infection and propagation. Here we systematically and functionally catalogued these host dependencies to various stages of the HCV replication cycle. Applying systems virology and functional genomics approaches with various in vitro HCV model systems, we further defined multiple previously unrecognized host factors or pathways that are involved in either HCV entry, IRES-mediated translation, RNA replication, or assembly/secretion. By bioinformatics meta-analyses and literature mining of existing publications and databases, we constructed an extensive roadmap of the cellular networks and pathways requisite for the complete HCV replication cycle. Our study yields novel insights into viral infection, pathogenesis and potential therapeutic targets. Furthermore, this study serves as a valuable reference source for subsequent work on host pathways and virus-host interactions in general.
Collapse
Affiliation(s)
- Qisheng Li
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yong-Yuan Zhang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stephan Chiu
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zongyi Hu
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Keng-Hsin Lan
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Helen Cha
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Catherine Sodroski
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fang Zhang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ching-Sheng Hsu
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Emmanuel Thomas
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - T. Jake Liang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
52
|
Hagen N, Bayer K, Rösch K, Schindler M. The intraviral protein interaction network of hepatitis C virus. Mol Cell Proteomics 2014; 13:1676-89. [PMID: 24797426 DOI: 10.1074/mcp.m113.036301] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hepatitis C virus (HCV) is a global health problem and one of the main reasons for chronic liver diseases such as cirrhosis and hepatocellular carcinoma. The HCV genome is translated into a polyprotein which is proteolytically processed into 10 viral proteins. The interactome of the HCV proteins with the host cell has been worked out; however, it remains unclear how viral proteins interact with each other. We aimed to generate the interaction network of these 10 HCV proteins using a flow-cytometry-based FRET assay established in our laboratory (Banning, C., Votteler, J., Hoffmann, D., Koppensteiner, H., Warmer, M., Reimer, R., Kirchhoff, F., Schubert, U., Hauber, J., and Schindler, M. (2010) A flow cytometry-based FRET assay to identify and analyse protein-protein interactions in living cells. PLoS One 5, e9344). HCV proteins were constructed as fusions with the chromophores CFP and YFP. All HCV fusions were expressed and localized to specific subcellular compartments, indicating that they were functional. FACS-FRET measurements identified a total of 20 interactions; 13 of these were previously described and have now been confirmed in living cells via our method. Among the seven novel protein binding pairs, HCV p7 plays a pivotal role. It binds to the HCV capsid protein Core and the two glycoproteins E1 and E2. These interplays were further demonstrated in the relevant context of Huh7.5 liver cells expressing infectious HCV. Our work demonstrates the feasibility of rapidly generating small interaction networks via FACS-FRET and defines the network of intra-HCV protein interactions. Furthermore, our data support an important role of p7 in HCV assembly.
Collapse
Affiliation(s)
- Nicole Hagen
- From the ‡Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Karen Bayer
- ¶Institute of Virology, Helmholtz Zentrum Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Kathrin Rösch
- From the ‡Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Michael Schindler
- From the ‡Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany; ¶Institute of Virology, Helmholtz Zentrum Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; ‖Institute of Medical Virology and Epidemiology of Viral Diseases, University Clinic Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
53
|
Stearoyl-CoA desaturase inhibition blocks formation of hepatitis C virus-induced specialized membranes. Sci Rep 2014; 4:4549. [PMID: 25008545 PMCID: PMC4091094 DOI: 10.1038/srep04549] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/13/2014] [Indexed: 01/11/2023] Open
Abstract
Hepatitis C virus (HCV) replication is dependent on the formation of specialized membrane structures; however, the host factor requirements for the formation of these HCV complexes remain unclear. Herein, we demonstrate that inhibition of stearoyl-CoA desaturase 1 (SCD-1) halts the biosynthesis of unsaturated fatty acids, such as oleic acid, and negatively modulates HCV replication. Unsaturated fatty acids play key roles in membrane curvature and fluidity. Mechanistically, we demonstrate that SCD-1 inhibition disrupts the integrity of membranous HCV replication complexes and renders HCV RNA susceptible to nuclease-mediated degradation. Our work establishes a novel function for unsaturated fatty acids in HCV replication.
Collapse
|
54
|
Ivanisenko NV, Mishchenko EL, Akberdin IR, Demenkov PS, Likhoshvai VA, Kozlov KN, Todorov DI, Gursky VV, Samsonova MG, Samsonov AM, Clausznitzer D, Kaderali L, Kolchanov NA, Ivanisenko VA. A new stochastic model for subgenomic hepatitis C virus replication considers drug resistant mutants. PLoS One 2014; 9:e91502. [PMID: 24643004 PMCID: PMC3958367 DOI: 10.1371/journal.pone.0091502] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 02/12/2014] [Indexed: 12/17/2022] Open
Abstract
As an RNA virus, hepatitis C virus (HCV) is able to rapidly acquire drug resistance, and for this reason the design of effective anti-HCV drugs is a real challenge. The HCV subgenomic replicon-containing cells are widely used for experimental studies of the HCV genome replication mechanisms, for drug testing in vitro and in studies of HCV drug resistance. The NS3/4A protease is essential for virus replication and, therefore, it is one of the most attractive targets for developing specific antiviral agents against HCV. We have developed a stochastic model of subgenomic HCV replicon replication, in which the emergence and selection of drug resistant mutant viral RNAs in replicon cells is taken into account. Incorporation into the model of key NS3 protease mutations leading to resistance to BILN-2061 (A156T, D168V, R155Q), VX-950 (A156S, A156T, T54A) and SCH 503034 (A156T, A156S, T54A) inhibitors allows us to describe the long term dynamics of the viral RNA suppression for various inhibitor concentrations. We theoretically showed that the observable difference between the viral RNA kinetics for different inhibitor concentrations can be explained by differences in the replication rate and inhibitor sensitivity of the mutant RNAs. The pre-existing mutants of the NS3 protease contribute more significantly to appearance of new resistant mutants during treatment with inhibitors than wild-type replicon. The model can be used to interpret the results of anti-HCV drug testing on replicon systems, as well as to estimate the efficacy of potential drugs and predict optimal schemes of their usage.
Collapse
Affiliation(s)
- Nikita V. Ivanisenko
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Elena L. Mishchenko
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Ilya R. Akberdin
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Pavel S. Demenkov
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Vitaly A. Likhoshvai
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Konstantin N. Kozlov
- Department of Computational Biology, St. Petersburg State Polytechnical University, St. Petersburg, Russia
| | - Dmitry I. Todorov
- Department of Computational Biology, St. Petersburg State Polytechnical University, St. Petersburg, Russia
- Chebyshev Laboratory, St. Petersburg State University, St. Petersburg, Russia
| | - Vitaly V. Gursky
- Department of Computational Biology, St. Petersburg State Polytechnical University, St. Petersburg, Russia
- Theoretical Department, Ioffe Physical-Technical Institute of the Russian Academy of Sciences, St.Petersburg, Russia
| | - Maria G. Samsonova
- Department of Computational Biology, St. Petersburg State Polytechnical University, St. Petersburg, Russia
| | - Alexander M. Samsonov
- Department of Computational Biology, St. Petersburg State Polytechnical University, St. Petersburg, Russia
- Theoretical Department, Ioffe Physical-Technical Institute of the Russian Academy of Sciences, St.Petersburg, Russia
| | - Diana Clausznitzer
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
| | - Lars Kaderali
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
| | - Nikolay A. Kolchanov
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Vladimir A. Ivanisenko
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
- PB-soft Llc, Novosibirsk, Russia
- * E-mail:
| |
Collapse
|
55
|
Ruggieri V, Mazzoccoli C, Pazienza V, Andriulli A, Capitanio N, Piccoli C. Hepatitis C virus, mitochondria and auto/mitophagy: Exploiting a host defense mechanism. World J Gastroenterol 2014; 20:2624-2633. [PMID: 24627598 PMCID: PMC3949271 DOI: 10.3748/wjg.v20.i10.2624] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/06/2013] [Accepted: 12/13/2013] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is the major reason for liver transplantation and the main cause of liver-related morbidity and mortality in a great number of countries. As for the other viruses, this pathogen interferes in more than one process and in more than one way with host cell biology. A mounting body of evidence points, in particular, toward the drastic alterations of mitochondrial physiology and functions that virus is able to induce, albeit the mechanisms have partly remained elusive. Role of the mitochondria in immunity and in quality control systems, as autophagy, as well as the strategies that HCV has evolved to evade and even to manipulate mitochondrial surveillance for its benefit, highlights the importance of deepening the mechanisms that modulate this virus-mitochondrion interaction, not only to intensify our knowledge of the HCV infection pathogenesis but also to design efficient antiviral strategies.
Collapse
|
56
|
Ivanisenko NV, Mishchenko EL, Akberdin IR, Demenkov PS, Likhoshvai VA, Kozlov KN, Todorov DI, Samsonova MG, Samsonov AM, Kolchanov NA, Ivanisenko VA. Replication of the subgenomic hepatitis C virus replicon in the presence of the NS3 protease inhibitors: a stochastic model. Biophysics (Nagoya-shi) 2014. [DOI: 10.1134/s0006350913050059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
57
|
Dynamic imaging of the hepatitis C virus NS5A protein during a productive infection. J Virol 2014; 88:3636-52. [PMID: 24429364 DOI: 10.1128/jvi.02490-13] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Hepatitis C virus (HCV) NS5A is essential for viral genome replication within cytoplasmic replication complexes and virus assembly at the lipid droplet (LD) surface, although its definitive functions are poorly understood. We developed approaches to investigate NS5A dynamics during a productive infection. We report here that NS5A motility and efficient HCV RNA replication require the microtubule network and the cytoplasmic motor dynein and demonstrate that both motile and relatively static NS5A-positive foci are enriched with host factors VAP-A and Rab5A. Pulse-chase imaging revealed that newly synthesized NS5A foci are small and distinct from aged foci, while further studies using a unique dual fluorescently tagged infectious HCV chimera showed a relatively stable association of NS5A foci with core-capped LDs. These results reveal new details about the dynamics and maturation of NS5A and the nature of potential sites of convergence of HCV replication and assembly pathways. IMPORTANCE Hepatitis C virus (HCV) is a major cause of serious liver disease worldwide. An improved understanding of the HCV replication cycle will enable development of novel and improved antiviral strategies. Here we have developed complementary fluorescent labeling and imaging approaches to investigate the localization, traffic and interactions of the HCV NS5A protein in living, virus-producing cells. These studies reveal new details as to the traffic, composition and biogenesis of NS5A foci and the nature of their association with putative sites of virus assembly.
Collapse
|
58
|
TIP47 is associated with the hepatitis C virus and its interaction with Rab9 is required for release of viral particles. Eur J Cell Biol 2013; 92:374-82. [PMID: 24480419 DOI: 10.1016/j.ejcb.2013.12.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/11/2013] [Accepted: 12/13/2013] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) morphogenesis and release are closely linked to lipid metabolism. It has been described recently by our group that TIP47 plays an essential role for the targeting of the NS5A-complexed RNA genome from the replicon complex to the lipid droplet. Moreover, apolipoprotein (apo) E was found to be associated with the viral particle. In light of the fact, that TIP47 harbors an apoE like domain and has a high affinity to lipoproteins, the interaction of TIP47 with the viral particle and the potential relevance for the release of the viral particle were investigated. Coimmunoprecipitations and electron microscopy analysis using immunogold labeling revealed that TIP47 binds to the viral particle and stays associated with the released HCV particle. Silencing of the TIP47 binding partner Rab9 by lentiviral transduction abolishes the viral replication. However, destruction of TIP47-Rab9 interactions by deletion/mutation of the Rab9 binding does not abolish the genome replication domain but prevents the release of HCV particles. The binding of these TIP47 mutants to the viral particle is not affected by destruction of the Rab9 binding domain. Moreover, we found that these TIP47 mutants lacking the binding site for Rab9 misdirect the de novo synthesized viral particles to the autophagosomal/lysosomal compartment where the particles are degraded. From this we conclude that the Rab9-complexed TIP47 plays an essential role for the proper release of hepatitis C viral particles.
Collapse
|
59
|
Lohmann V, Bartenschlager R. On the History of Hepatitis C Virus Cell Culture Systems. J Med Chem 2013; 57:1627-42. [DOI: 10.1021/jm401401n] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Volker Lohmann
- Department of Infectious
Diseases, Molecular Virology, Heidelberg University, Heidelberg, 69120, Germany
| | - Ralf Bartenschlager
- Department of Infectious
Diseases, Molecular Virology, Heidelberg University, Heidelberg, 69120, Germany
| |
Collapse
|
60
|
Ali N, Allam H, Bader T, May R, Basalingappa KM, Berry WL, Chandrakesan P, Qu D, Weygant N, Bronze MS, Umar S, Janknecht R, Sureban SM, Huycke M, Houchen CW. Fluvastatin interferes with hepatitis C virus replication via microtubule bundling and a doublecortin-like kinase-mediated mechanism. PLoS One 2013; 8:e80304. [PMID: 24260365 PMCID: PMC3833963 DOI: 10.1371/journal.pone.0080304] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 10/11/2013] [Indexed: 12/18/2022] Open
Abstract
Hepatitis C virus (HCV)-induced alterations in lipid metabolism and cellular protein expression contribute to viral pathogenesis. The mechanism of pleiotropic actions of cholesterol-lowering drugs, statins, against HCV and multiple cancers are not well understood. We investigated effects of fluvastatin (FLV) on microtubule-associated and cancer stem cell marker (CSC), doublecortin-like kinase 1 (DCLK1) during HCV-induced hepatocarcinogenesis. HCV replication models, cancer cell lines and normal human hepatocytes were used to investigate the antiviral and antitumor effects of statins. FLV treatment resulted in induction of microtubule bundling, cell-cycle arrest and alterations in cellular DCLK1 distribution in HCV-expressing hepatoma cells. These events adversely affected the survival of liver-derived tumor cells without affecting normal human hepatocytes. FLV downregulated HCV replication in cell culture where the ATP pool and cell viability were not compromised. Pravastatin did not exhibit these effects on HCV replication, microtubules and cancer cells. The levels of miR-122 that regulates liver homeostasis and provides HCV genomic stability remained at steady state whereas DCLK1 mRNA levels were considerably reduced during FLV treatment. We further demonstrated that HCV replication was increased with DCLK1 overexpression. In conclusion, unique effects of FLV on microtubules and their binding partner DCLK1 are likely to contribute to its anti-HCV and antitumor activities in addition to its known inhibitory effects on 3-hydroxy-3-methylglutary-CoA reductase (HMGCR).
Collapse
Affiliation(s)
- Naushad Ali
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Department of Veterans Affairs Medical Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- * E-mail: (NA); (CWH)
| | - Heba Allam
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Department of Microbiology and Immunology, National Liver Institute, Menoufiya University, Menoufiya, Egypt
| | - Ted Bader
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Department of Veterans Affairs Medical Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Randal May
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Department of Veterans Affairs Medical Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Kanthesh M. Basalingappa
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - William L. Berry
- Department of Cell Biology, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Parthasarathy Chandrakesan
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Dongfeng Qu
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Nathaniel Weygant
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Michael S. Bronze
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Shahid Umar
- Department of Molecular and Integrative Physiology, and Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Sripathi M. Sureban
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Department of Veterans Affairs Medical Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Mark Huycke
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Department of Veterans Affairs Medical Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Courtney W. Houchen
- Department of Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- Department of Veterans Affairs Medical Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
- * E-mail: (NA); (CWH)
| |
Collapse
|
61
|
McCartney EM, Helbig KJ, Narayana SK, Eyre NS, Aloia AL, Beard MR. Signal transducer and activator of transcription 3 is a proviral host factor for hepatitis C virus. Hepatology 2013; 58:1558-68. [PMID: 23703790 DOI: 10.1002/hep.26496] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 04/05/2013] [Accepted: 04/22/2013] [Indexed: 12/31/2022]
Abstract
UNLABELLED Host factors play an important role in all facets of the hepatitis C virus (HCV) life cycle and one such host factor is signal transducer and activator of transcription 3 (STAT3). The HCV core protein has been shown to directly interact with and activate STAT3, while oxidative stress generated during HCV replication in a replicon-based model also induced STAT3 activation. However, despite these findings the precise role of STAT3 in the HCV life cycle remains unknown. We have established that STAT3 is actively phosphorylated in the presence of replicating HCV. Furthermore, expression of a constitutively active form of STAT3 leads to marked increases in HCV replication, whereas, conversely, chemical inhibition and small interfering RNA (siRNA) knockdown of STAT3 leads to significant decreases in HCV RNA levels. This strongly implicates STAT3 as a proviral host factor. As STAT3 is a transcription factor, up-regulation of a distinct set of STAT3-dependent genes may create an environment that is favorable for HCV replication. However, STAT3 has recently been demonstrated to positively regulate microtubule (MT) dynamics, by way of a direct sequestration of the MT depolymerizing protein Stathmin 1 (STMN1), and we provide evidence that STAT3 may exert its effect on the HCV life cycle by way of positive regulation of MT dynamics. CONCLUSION We have demonstrated that STAT3 plays a role in the life cycle of HCV and have clarified the role of STAT3 as a proviral host factor.
Collapse
Affiliation(s)
- Erin M McCartney
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, and Centre for Cancer Biology, SA Pathology, Adelaide, South Australia
| | | | | | | | | | | |
Collapse
|
62
|
Helbig KJ, Beard MR. The role of viperin in the innate antiviral response. J Mol Biol 2013; 426:1210-9. [PMID: 24157441 DOI: 10.1016/j.jmb.2013.10.019] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/15/2013] [Accepted: 10/16/2013] [Indexed: 12/18/2022]
Abstract
Viral infection of the cell is able to initiate a signaling cascade of events that ultimately attempts to limit viral replication and prevent escalating infection through expression of host antiviral proteins. Recent work has highlighted the importance of the host antiviral protein viperin in this process, with its ability to limit a large variety of viral infections as well as play a role in the production of type I interferon and the modulation of a number of transcription factor binding sites. Viperin appears to have the ability to modulate varying conditions within the cell and to interfere with proviral host proteins in its attempts to create an unfavorable environment for viral replication. The study of the mechanistic actions of viperin has come a long way in recent years, describing important functional domains of the protein for its antiviral and immune modulator actions as well as demonstrating its role as a member of the radical SAM enzyme family. However, despite the rapid expansion of knowledge regarding the functions of this highly conserved and ancient antiviral protein, there still remains large gaps in our understanding of the precise mechanisms at play for viperin to exert such a wide variety of roles within the cell.
Collapse
Affiliation(s)
- Karla J Helbig
- School of Molecular and Biomedical Science and the Institute for Molecular Pathology, The University of Adelaide, South Australia 5005, Australia; Centre for Cancer Biology, SA Pathology, Adelaide, South Australia 5001, Australia
| | - Michael R Beard
- School of Molecular and Biomedical Science and the Institute for Molecular Pathology, The University of Adelaide, South Australia 5005, Australia; Centre for Cancer Biology, SA Pathology, Adelaide, South Australia 5001, Australia.
| |
Collapse
|
63
|
Functional characterization of bovine viral diarrhea virus nonstructural protein 5A by reverse genetic analysis and live cell imaging. J Virol 2013; 88:82-98. [PMID: 24131714 DOI: 10.1128/jvi.01957-13] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nonstructural protein 5A (NS5A) of bovine viral diarrhea virus (BVDV) is a hydrophilic phosphoprotein with RNA binding activity and a critical component of the viral replicase. In silico analysis suggests that NS5A encompasses three domains interconnected by two low-complexity sequences (LCSs). While domain I harbors two functional determinants, an N-terminal amphipathic helix important for membrane association, and a Zn-binding site essential for RNA replication, the structure and function of the C-terminal half of NS5A are still ill defined. In this study, we introduced a panel of 10 amino acid deletions covering the C-terminal half of NS5A. In the context of a highly efficient monocistronic replicon, deletions in LCS I and the N-terminal part of domain II, as well as in domain III, were tolerated with regard to RNA replication. When introduced into a bicistronic replicon, only deletions in LCS I and the N-terminal part of domain II were tolerated. In the context of the viral full-length genome, these mutations allowed residual virion morphogenesis. Based on these data, a functional monocistronic BVDV replicon coding for an NS5A variant with an insertion of the fluorescent protein mCherry was constructed. Live cell imaging demonstrated that a fraction of NS5A-mCherry localizes to the surface of lipid droplets. Taken together, this study provides novel insights into the functions of BVDV NS5A. Moreover, we established the first pestiviral replicon expressing fluorescent NS5A-mCherry to directly visualize functional viral replication complexes by live cell imaging.
Collapse
|
64
|
Farhat R, Goueslain L, Wychowski C, Belouzard S, Fénéant L, Jackson CL, Dubuisson J, Rouillé Y. Hepatitis C virus replication and Golgi function in brefeldin a-resistant hepatoma-derived cells. PLoS One 2013; 8:e74491. [PMID: 24058576 PMCID: PMC3776844 DOI: 10.1371/journal.pone.0074491] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/01/2013] [Indexed: 12/15/2022] Open
Abstract
Recent reports indicate that the replication of hepatitis C virus (HCV) depends on the GBF1-Arf1-COP-I pathway. We generated Huh-7-derived cell lines resistant to brefeldin A (BFA), which is an inhibitor of this pathway. The resistant cell lines could be sorted into two phenotypes regarding BFA-induced toxicity, inhibition of albumin secretion, and inhibition of HCV infection. Two cell lines were more than 100 times more resistant to BFA than the parental Huh-7 cells in these 3 assays. This resistant phenotype was correlated with the presence of a point mutation in the Sec7 domain of GBF1, which is known to impair the binding of BFA. Surprisingly, the morphology of the cis-Golgi of these cells remained sensitive to BFA at concentrations of the drug that allowed albumin secretion, indicating a dichotomy between the phenotypes of secretion and Golgi morphology. Cells of the second group were about 10 times more resistant than parental Huh-7 cells to the BFA-induced toxicity. The EC50 for albumin secretion was only 1.5–1.8 fold higher in these cells than in Huh-7 cells. However their level of secretion in the presence of inhibitory doses of BFA was 5 to 15 times higher. Despite this partially effective secretory pathway in the presence of BFA, the HCV infection was almost as sensitive to BFA as in Huh-7 cells. This suggests that the function of GBF1 in HCV replication does not simply reflect its role of regulator of the secretory pathway of the host cell. Thus, our results confirm the involvement of GBF1 in HCV replication, and suggest that GBF1 might fulfill another function, in addition to the regulation of the secretory pathway, during HCV replication.
Collapse
Affiliation(s)
- Rayan Farhat
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| | - Lucie Goueslain
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Czeslaw Wychowski
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| | - Sandrine Belouzard
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| | - Lucie Fénéant
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| | - Catherine L. Jackson
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Jean Dubuisson
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| | - Yves Rouillé
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
- * E-mail:
| |
Collapse
|
65
|
Cell-cell contact-mediated hepatitis C virus (HCV) transfer, productive infection, and replication and their requirement for HCV receptors. J Virol 2013; 87:8545-58. [PMID: 23720720 DOI: 10.1128/jvi.01062-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C virus (HCV) infection is believed to begin with interactions between cell-free HCV and cell receptors that include CD81, scavenger receptor B1 (SR-B1), claudin-1 (CLDN1), and occludin (OCLN). In this study, we have demonstrated that HCV spreading from infected hepatocytes to uninfected hepatocytes leads to the transfer of HCV and the formation of infection foci and is cell density dependent. This cell-cell contact-mediated (CCCM) HCV transfer occurs readily and requires all these known HCV receptors and an intact actin cytoskeleton. With a fluorescently labeled replication-competent HCV system, the CCCM transfer process was further dissected by live-cell imaging into four steps: donor cell-target cell contact, formation of viral puncta-target cell conjugation, transfer of viral puncta, and posttransfer. Importantly, the CCCM HCV transfer leads to productive infection of target cells. Taken together, these results show that CCCM HCV transfer constitutes an important and effective route for HCV infection and dissemination. These findings will aid in the development of new and novel strategies for preventing and treating HCV infection.
Collapse
|
66
|
Three-dimensional architecture of tick-borne encephalitis virus replication sites and trafficking of the replicated RNA. J Virol 2013; 87:6469-81. [PMID: 23552408 DOI: 10.1128/jvi.03456-12] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Flavivirus replication is accompanied by the rearrangement of cellular membranes that may facilitate viral genome replication and protect viral components from host cell responses. The topological organization of viral replication sites and the fate of replicated viral RNA are not fully understood. We exploited electron microscopy to map the organization of tick-borne encephalitis virus (TBEV) replication compartments in infected cells and in cells transfected with a replicon. Under both conditions, 80-nm vesicles were seen within the lumen of the endoplasmic reticulum (ER) that in infected cells also contained virions. By electron tomography, the vesicles appeared as invaginations of the ER membrane, displaying a pore that could enable release of newly synthesized viral RNA into the cytoplasm. To track the fate of TBEV RNA, we took advantage of our recently developed method of viral RNA fluorescent tagging for live-cell imaging combined with bleaching techniques. TBEV RNA was found outside virus-induced vesicles either associated to ER membranes or free to move within a defined area of juxtaposed ER cisternae. From our results, we propose a biologically relevant model of the possible topological organization of flavivirus replication compartments composed of replication vesicles and a confined extravesicular space where replicated viral RNA is retained. Hence, TBEV modifies the ER membrane architecture to provide a protected environment for viral replication and for the maintenance of newly replicated RNA available for subsequent steps of the virus life cycle.
Collapse
|
67
|
Inoue T, Tsai B. How viruses use the endoplasmic reticulum for entry, replication, and assembly. Cold Spring Harb Perspect Biol 2013; 5:a013250. [PMID: 23284050 DOI: 10.1101/cshperspect.a013250] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
To cause infection, a virus enters a host cell, replicates, and assembles, with the resulting new viral progeny typically released into the extracellular environment to initiate a new infection round. Virus entry, replication, and assembly are dynamic and coordinated processes that require precise interactions with host components, often within and surrounding a defined subcellular compartment. Accumulating evidence pinpoints the endoplasmic reticulum (ER) as a crucial organelle supporting viral entry, replication, and assembly. This review focuses on the molecular mechanism by which different viruses co-opt the ER to accomplish these crucial infection steps. Certain bacterial toxins also hijack the ER for entry. An interdisciplinary approach, using rigorous biochemical and cell biological assays coupled with advanced microscopy strategies, will push to the next level our understanding of the virus-ER interaction during infection.
Collapse
Affiliation(s)
- Takamasa Inoue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| | | |
Collapse
|
68
|
Competing and noncompeting activities of miR-122 and the 5' exonuclease Xrn1 in regulation of hepatitis C virus replication. Proc Natl Acad Sci U S A 2012; 110:1881-6. [PMID: 23248316 DOI: 10.1073/pnas.1213515110] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C virus (HCV) replication is dependent on microRNA 122 (miR-122), a liver-specific microRNA that recruits Argonaute 2 to the 5' end of the viral genome, stabilizing it and slowing its decay both in cell-free reactions and in infected cells. Here we describe the RNA degradation pathways against which miR-122 provides protection. Transfected HCV RNA is degraded by both the 5' exonuclease Xrn1 and 3' exonuclease exosome complex, whereas replicating RNA within infected cells is degraded primarily by Xrn1 with no contribution from the exosome. Consistent with this, sequencing of the 5' and 3' ends of RNA degradation intermediates in infected cells confirmed that 5' decay is the primary pathway for HCV RNA degradation. Xrn1 knockdown enhances HCV replication, indicating that Xrn1 decay and the viral replicase compete to set RNA abundance within infected cells. Xrn1 knockdown and miR-122 supplementation have equal, redundant, and nonadditive effects on the rate of viral RNA decay, indicating that miR-122 protects HCV RNA from 5' decay. Nevertheless, Xrn1 knockdown does not rescue replication of a viral mutant defective in miR-122 binding, indicating that miR-122 has additional yet uncharacterized function(s) in the viral life cycle.
Collapse
|
69
|
Romero-Brey I, Merz A, Chiramel A, Lee JY, Chlanda P, Haselman U, Santarella-Mellwig R, Habermann A, Hoppe S, Kallis S, Walther P, Antony C, Krijnse-Locker J, Bartenschlager R. Three-dimensional architecture and biogenesis of membrane structures associated with hepatitis C virus replication. PLoS Pathog 2012; 8:e1003056. [PMID: 23236278 PMCID: PMC3516559 DOI: 10.1371/journal.ppat.1003056] [Citation(s) in RCA: 398] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 10/11/2012] [Indexed: 02/08/2023] Open
Abstract
All positive strand RNA viruses are known to replicate their genomes in close association with intracellular membranes. In case of the hepatitis C virus (HCV), a member of the family Flaviviridae, infected cells contain accumulations of vesicles forming a membranous web (MW) that is thought to be the site of viral RNA replication. However, little is known about the biogenesis and three-dimensional structure of the MW. In this study we used a combination of immunofluorescence- and electron microscopy (EM)-based methods to analyze the membranous structures induced by HCV in infected cells. We found that the MW is derived primarily from the endoplasmic reticulum (ER) and contains markers of rough ER as well as markers of early and late endosomes, COP vesicles, mitochondria and lipid droplets (LDs). The main constituents of the MW are single and double membrane vesicles (DMVs). The latter predominate and the kinetic of their appearance correlates with kinetics of viral RNA replication. DMVs are induced primarily by NS5A whereas NS4B induces single membrane vesicles arguing that MW formation requires the concerted action of several HCV replicase proteins. Three-dimensional reconstructions identify DMVs as protrusions from the ER membrane into the cytosol, frequently connected to the ER membrane via a neck-like structure. In addition, late in infection multi-membrane vesicles become evident, presumably as a result of a stress-induced reaction. Thus, the morphology of the membranous rearrangements induced in HCV-infected cells resemble those of the unrelated picorna-, corona- and arteriviruses, but are clearly distinct from those of the closely related flaviviruses. These results reveal unexpected similarities between HCV and distantly related positive-strand RNA viruses presumably reflecting similarities in cellular pathways exploited by these viruses to establish their membranous replication factories. All positive-strand RNA viruses replicate in the cytoplasm in distinct membranous compartments acting as ‘replication factories’. Membranes building up these factories are recruited from different sources and serve as platforms for the assembly of multi-subunit protein complexes (the ‘replicase’) that catalyze the amplification of the viral RNA genome. In this study we found that hepatitis C virus (HCV), a major causative agent of chronic liver disease, induces profound remodeling of primarily endoplasmic reticulum-derived membranes. Surprisingly, the 3D architecture of these membrane rearrangements is similar to those induced by the unrelated picorna- and coronaviruses, but in striking contrast to the closely related flaviviruses. Early in infection HCV induces double membrane vesicles (DMVs) that emerge as protrusions of the ER; later on, HCV induces in addition multi-membrane vesicles that are probably the result of a cellular stress reaction and that are reminiscent to an autophagic response. These profound membrane rearrangements are induced by the concerted action of HCV-encoded nonstructural proteins of which NS5A is the only one capable to induce DMVs. These results provide important insights into the 3D architecture of the membrane alterations induced by HCV and reveal unexpected similarities between HCV and the very distantly related picorna- and coronaviruses.
Collapse
Affiliation(s)
- Inés Romero-Brey
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Andreas Merz
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Abhilash Chiramel
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Ji-Young Lee
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Petr Chlanda
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Uta Haselman
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Anja Habermann
- Electron Microscopy Core Facility, University of Heidelberg, Heidelberg, Germany
| | - Simone Hoppe
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
- Electron Microscopy Core Facility, University of Heidelberg, Heidelberg, Germany
| | - Stephanie Kallis
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Paul Walther
- Central Electron Microscopy Facility, Ulm University, Ulm, Germany
| | - Claude Antony
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jacomine Krijnse-Locker
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
- Electron Microscopy Core Facility, University of Heidelberg, Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
70
|
Abstract
Coronaviruses are positive-strand RNA viruses that are important infectious agents of both animals and humans. A common feature among positive-strand RNA viruses is their assembly of replication-transcription complexes in association with cytoplasmic membranes. Upon infection, coronaviruses extensively rearrange cellular membranes into organelle-like replicative structures that consist of double-membrane vesicles and convoluted membranes to which the nonstructural proteins involved in RNA synthesis localize. Double-stranded RNA, presumably functioning as replicative intermediate during viral RNA synthesis, has been detected at the double-membrane vesicle interior. Recent studies have provided new insights into the assembly and functioning of the coronavirus replicative structures. This review will summarize the current knowledge on the biogenesis of the replicative structures, the membrane anchoring of the replication-transcription complexes, and the location of viral RNA synthesis, with particular focus on the dynamics of the coronavirus replicative structures and individual replication-associated proteins.
Collapse
|
71
|
Grangeon R, Jiang J, Laliberté JF. Host endomembrane recruitment for plant RNA virus replication. Curr Opin Virol 2012; 2:683-90. [PMID: 23123078 PMCID: PMC7185485 DOI: 10.1016/j.coviro.2012.10.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/03/2012] [Accepted: 10/04/2012] [Indexed: 01/27/2023]
Abstract
Although there is a significant amount of literature that deals with the identification of plant viral proteins involved in membrane remodeling and vesicle production in infected cells, there are very few investigations that report on the impact that infection has on the overall architecture and dynamics of the early secretory endomembranes. Recent investigations have shown that for some viruses the endoplasmic reticulum, Golgi bodies and other organelles are heavily recruited into virus-induced perinuclear structures. These structures are not isolated organelles and are dynamically connected to the bulk of non-modified endomembranes. They also have a functional link with peripheral motile vesicles involved in virus intracellular movement. The full molecular events that consubstantiate with this endomembrane recruitment in virus-induced structures remain to be elucidated but viral genome replication and virion assembly are probably taking place within these structures.
Collapse
Affiliation(s)
- Romain Grangeon
- INRS-Institut Armand Frappier, 531 Boulevard des Prairies, Laval, Québec H7V 1B7, Canada
| | | | | |
Collapse
|
72
|
Grangeon R, Agbeci M, Chen J, Grondin G, Zheng H, Laliberté JF. Impact on the endoplasmic reticulum and Golgi apparatus of turnip mosaic virus infection. J Virol 2012; 86:9255-65. [PMID: 22718813 PMCID: PMC3416146 DOI: 10.1128/jvi.01146-12] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 06/08/2012] [Indexed: 01/10/2023] Open
Abstract
The impact of turnip mosaic virus (TuMV) infection on the endomembranes of the host early secretory pathway was investigated using an infectious clone that has been engineered for tagging viral membrane structures with a fluorescent protein fused to the viral protein 6K(2). TuMV infection led to the amalgamation of the endoplasmic reticulum (ER), Golgi apparatus, COPII coatamers, and chloroplasts into a perinuclear globular structure that also contained viral proteins. One consequence of TuMV infection was that protein secretion was blocked at the ER-Golgi interface. Fluorescence recovery after photobleaching (FRAP) experiments indicated that the perinuclear structure cannot be restocked in viral components but was dynamically connected to the bulk of the Golgi apparatus and the ER. Experiments with 6K(2) fused to photoactivable green fluorescent protein (GFP) showed that production of motile peripheral 6K(2) vesicles was functionally linked to the perinuclear structure. Disruption of the early secretory pathway did not prevent the formation of the perinuclear globular structure, enhanced the clustering of peripheral 6K(2) vesicles with COPII coatamers, and led to inhibition of cell-to-cell virus movement. This suggests that a functional secretory pathway is not required for the formation of the TuMV perinuclear globular structure and peripheral vesicles but is needed for successful viral intercellular propagation.
Collapse
Affiliation(s)
- Romain Grangeon
- INRS-Institut Armand-Frappier, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Maxime Agbeci
- INRS-Institut Armand-Frappier, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Jun Chen
- Department of Biology, McGill University, Montréal, Québec, Canada
| | - Gilles Grondin
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Huanquan Zheng
- Department of Biology, McGill University, Montréal, Québec, Canada
| | - Jean-François Laliberté
- INRS-Institut Armand-Frappier, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| |
Collapse
|
73
|
Liu HM, Aizaki H, Machida K, Ou JHJ, Lai MMC. Hepatitis C virus translation preferentially depends on active RNA replication. PLoS One 2012; 7:e43600. [PMID: 22937067 PMCID: PMC3427374 DOI: 10.1371/journal.pone.0043600] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/26/2012] [Indexed: 12/19/2022] Open
Abstract
Hepatitis C virus (HCV) RNA initiates its replication on a detergent-resistant membrane structure derived from the endoplasmic reticulum (ER) in the HCV replicon cells. By performing a pulse-chase study of BrU-labeled HCV RNA, we found that the newly-synthesized HCV RNA traveled along the anterograde-membrane traffic and moved away from the ER. Presumably, the RNA moved to the site of translation or virion assembly in the later steps of viral life cycle. In this study, we further addressed how HCV RNA translation was regulated by HCV RNA trafficking. When the movement of HCV RNA from the site of RNA synthesis to the Golgi complex was blocked by nocodazole, an inhibitor of ER-Golgi transport, HCV protein translation was surprisingly enhanced, suggesting that the translation of viral proteins occurred near the site of RNA synthesis. We also found that the translation of HCV proteins was dependent on active RNA synthesis: inhibition of viral RNA synthesis by an NS5B inhibitor resulted in decreased HCV viral protein synthesis even when the total amount of intracellular HCV RNA remained unchanged. Furthermore, the translation activity of the replication-defective HCV replicons or viral RNA with an NS5B mutation was greatly reduced as compared to that of the corresponding wildtype RNA. By performing live cell labeling of newly synthesized HCV RNA and proteins, we further showed that the newly synthesized HCV proteins colocalized with the newly synthesized viral RNA, suggesting that HCV RNA replication and protein translation take place at or near the same site. Our findings together indicate that the translation of HCV RNA is coupled to RNA replication and that the both processes may occur at the same subcellular membrane compartments, which we term the replicasome.
Collapse
Affiliation(s)
- Helene Minyi Liu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America.
| | | | | | | | | |
Collapse
|
74
|
Phosphatidylinositol 4-kinases: hostages harnessed to build panviral replication platforms. Trends Biochem Sci 2012; 37:293-302. [PMID: 22633842 PMCID: PMC3389303 DOI: 10.1016/j.tibs.2012.03.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 03/27/2012] [Accepted: 03/30/2012] [Indexed: 12/20/2022]
Abstract
Several RNA viruses have recently been shown to hijack members of the host phosphatidylinositol (PtdIns) 4-kinase (PI4K) family of enzymes. They use PI4K to generate membranes enriched in phosphatidylinositide 4-phosphate (PtdIns4P or PI4P) lipids, which can be used as replication platforms. Viral replication machinery is assembled on these platforms as a supramolecular complex and PtdIns4P lipids regulate viral RNA synthesis. This article highlights these recent studies on the regulation of viral RNA synthesis by PtdIns4P lipids. It explores the potential mechanisms by which PtdIns4P lipids can contribute to viral replication and discusses the therapeutic potential of developing antiviral molecules that target host PI4Ks as a form of panviral therapy.
Collapse
|
75
|
Role for TBC1D20 and Rab1 in hepatitis C virus replication via interaction with lipid droplet-bound nonstructural protein 5A. J Virol 2012; 86:6491-502. [PMID: 22491470 DOI: 10.1128/jvi.00496-12] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Replication and assembly of hepatitis C virus (HCV) depend on the host's secretory and lipid-biosynthetic machinery. Viral replication occurs on endoplasmic reticulum (ER)-derived modified membranes, while viral assembly is thought to occur on lipid droplets (LDs). A physical association and coordination between the viral replication and assembly complexes are prerequisites for efficient viral production. Nonstructural protein 5A (NS5A), which localizes both to the ER and LDs, is an ideal candidate for this function. Here, the interaction of NS5A with host cell membranes and binding partners was characterized in living cells. The binding of NS5A to LDs is apparently irreversible, both in HCV-infected cells and when ectopically expressed. In HCV-infected cells, NS5A fluorescence was observed around the LDs and in perinuclear structures that were incorporated into a highly immobile platform superimposed over the ER membrane. Moreover, TBC1D20 and its cognate GTPase Rab1 are recruited by NS5A to LDs. The NS5A-TBC1D20 interaction was shown to be essential for the viral life cycle. In cells, expression of the Rab1 dominant negative (Rab1DN) GTPase mutant abolished steady-state LDs. In infected cells, Rab1DN induced the elimination of NS5A from viral replication sites. Our results demonstrate the significance of the localization of NS5A to LDs and support a model whereby its interaction with TBC1D20 and Rab1 affects lipid droplet metabolism to promote the viral life cycle.
Collapse
|
76
|
Horner SM, Park HS, Gale M. Control of innate immune signaling and membrane targeting by the Hepatitis C virus NS3/4A protease are governed by the NS3 helix α0. J Virol 2012; 86:3112-20. [PMID: 22238314 PMCID: PMC3302330 DOI: 10.1128/jvi.06727-11] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/03/2012] [Indexed: 12/28/2022] Open
Abstract
Hepatitis C virus (HCV) infection is sensed in the host cell by the cytosolic pathogen recognition receptor RIG-I. RIG-I signaling is propagated through its signaling adaptor protein MAVS to drive activation of innate immunity. However, HCV blocks RIG-I signaling through viral NS3/4A protease cleavage of MAVS on the mitochondrion-associated endoplasmic reticulum (ER) membrane (MAM). The multifunctional HCV NS3/4A serine protease is associated with intracellular membranes, including the MAM, through membrane-targeting domains within NS4A and also at the amphipathic helix α(0) of NS3. The serine protease domain of NS3 is required for both cleavage of MAVS, a tail-anchored membrane protein, and processing the HCV polyprotein. Here, we show that hydrophobic amino acids in the NS3 helix α(0) are required for selective cleavage of membrane-anchored portions of the HCV polyprotein and for cleavage of MAVS for control of RIG-I pathway signaling of innate immunity. Further, we found that the hydrophobic composition of NS3 helix α(0) is essential to establish HCV replication and infection. Alanine substitution of individual hydrophobic amino acids in the NS3 helix α(0) impaired HCV RNA replication in cells with a functional RIG-I pathway, but viral RNA replication was rescued in cells lacking RIG-I signaling. Therefore, the hydrophobic amphipathic helix α(0) of NS3 is required for NS3/4A control of RIG-I signaling and HCV replication by directing the membrane targeting of both viral and cellular substrates.
Collapse
Affiliation(s)
- Stacy M Horner
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | | | | |
Collapse
|
77
|
Shapira A, Shapira S, Gal-Tanamy M, Zemel R, Tur-Kaspa R, Benhar I. Removal of hepatitis C virus-infected cells by a zymogenized bacterial toxin. PLoS One 2012; 7:e32320. [PMID: 22359682 PMCID: PMC3281143 DOI: 10.1371/journal.pone.0032320] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 01/26/2012] [Indexed: 11/19/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver disease and has become a global health threat. No HCV vaccine is currently available and treatment with antiviral therapy is associated with adverse side effects. Moreover, there is no preventive therapy for recurrent hepatitis C post liver transplantation. The NS3 serine protease is necessary for HCV replication and represents a prime target for developing anti HCV therapies. Recently we described a therapeutic approach for eradication of HCV infected cells that is based on protein delivery of two NS3 protease-activatable recombinant toxins we named "zymoxins". These toxins were inactivated by fusion to rationally designed inhibitory peptides via NS3-cleavable linkers. Once delivered to cells where NS3 protease is present, the inhibitory peptide is removed resulting in re-activation of cytotoxic activity. The zymoxins we described suffered from two limitations: they required high levels of protease for activation and had basal activities in the un-activated form that resulted in a narrow potential therapeutic window. Here, we present a solution that overcame the major limitations of the "first generation zymoxins" by converting MazF ribonuclease, the toxic component of the E. coli chromosomal MazEF toxin-antitoxin system, into an NS3-activated zymoxin that is introduced to cells by means of gene delivery. We constructed an expression cassette that encodes for a single polypeptide that incorporates both the toxin and a fragment of its potent natural antidote, MazE, linked via an NS3-cleavable linker. While covalently paired to its inhibitor, the ribonuclease is well tolerated when expressed in naïve, healthy cells. In contrast, activating proteolysis that is induced by even low levels of NS3, results in an eradication of NS3 expressing model cells and HCV infected cells. Zymoxins may thus become a valuable tool in eradicating cells infected by intracellular pathogens that express intracellular proteases.
Collapse
Affiliation(s)
- Assaf Shapira
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel
- Molecular Hepatology Research Laboratory, Sackler School of Medicine, Felsenstein Medical Research Center, Tel-Aviv University, Petah Tikva, Israel
| | - Shiran Shapira
- The Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel-Aviv, Israel
- Sackler School of Medicine, Tel-Aviv University, Ramat Aviv, Israel
| | - Meital Gal-Tanamy
- Molecular Hepatology Research Laboratory, Sackler School of Medicine, Felsenstein Medical Research Center, Tel-Aviv University, Petah Tikva, Israel
| | - Romy Zemel
- Molecular Hepatology Research Laboratory, Sackler School of Medicine, Felsenstein Medical Research Center, Tel-Aviv University, Petah Tikva, Israel
| | - Ran Tur-Kaspa
- Molecular Hepatology Research Laboratory, Sackler School of Medicine, Felsenstein Medical Research Center, Tel-Aviv University, Petah Tikva, Israel
- Department of Medicine D and Liver Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Itai Benhar
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel
- * E-mail:
| |
Collapse
|
78
|
Nakabayashi J. A compartmentalization model of hepatitis C virus replication: an appropriate distribution of HCV RNA for the effective replication. J Theor Biol 2012; 300:110-7. [PMID: 22286015 DOI: 10.1016/j.jtbi.2012.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 01/10/2012] [Accepted: 01/11/2012] [Indexed: 01/28/2023]
Abstract
Hepatitis C virus (HCV) infection is a major cause of liver disease. Ten to twenty percent of chronic hepatitis C will develop complications of chronic liver diseases such as liver cirrhosis and hepatocellular carcinoma. The culture system of HCV is established by the specific combination between HCV strain and a host cell. Some chimeras substituting core to NS2 into the analogous region of JFH1 strain fail to effectively replicate. Core to NS2 of HCV gene mainly encodes the structural protein of HCV virion and contributes to the virion assembly, while other regions mainly contribute to the genome replication. The balance between the virion assembly and the genome replication of chimera may differ from that of reference strain. We construct a mathematical model of the whole replication process of HCV in single infected cell. It is revealed by this model that there are two replication patterns of HCV, explosive and arrested replication. In the explosive replication, HCV can continue to exponentially reproduce its progeny. The explosive replication is caused by the effect of the positive feedback in the replication cycle. On the other hand, in the arrested replication, the replication is stalled after sufficiently long time has passed from the infection because of the depletion of the genome RNA of HCV. To avoid the arrest of replication, HCV RNA must be appropriately distributed to three distinct functions as a template for the genome replication, as a template for the translation of viral proteins and as a component of the viral particle. When the genome replication and the translation of viral proteins precede to the virion assembly, HCV can effectively replicate by explosive replication. It is suggested that some chimeras of HCV fail to effectively replicate because of the inappropriate distribution of HCV RNA to these functions.
Collapse
Affiliation(s)
- Jun Nakabayashi
- Department of Mathematical Informatics, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyoku, Tokyo, Japan.
| |
Collapse
|
79
|
Popescu CI, Rouillé Y, Dubuisson J. Hepatitis C virus assembly imaging. Viruses 2011; 3:2238-54. [PMID: 22163343 PMCID: PMC3230850 DOI: 10.3390/v3112238] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C Virus (HCV) assembly process is the least understood step in the virus life cycle. The functional data revealed by forward and reverse genetics indicated that both structural and non-structural proteins are involved in the assembly process. Using confocal and electron microscopy different groups determined the subcellular localization of different viral proteins and they identified the lipid droplets (LDs) as the potential viral assembly site. Here, we aim to review the mechanisms that govern the viral proteins recruitment to LDs and discuss the current model of HCV assembly process. Based on previous examples, this review will also discuss advanced imaging techniques as potential means to extend our present knowledge of HCV assembly process.
Collapse
Affiliation(s)
- Costin-Ioan Popescu
- Institute of Biochemistry, The Romanian Academy, Splaiul Independentei 296, 060031 Bucharest 17, Romania
| | - Yves Rouillé
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille 59021, France; E-Mails: (Y.R.); (J.D.)
| | - Jean Dubuisson
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille (CIIL), Institut Pasteur de Lille, Université Lille Nord de France, Lille 59021, France; E-Mails: (Y.R.); (J.D.)
| |
Collapse
|
80
|
Ghosh S, Ahrens WA, Phatak SU, Hwang S, Schrum LW, Bonkovsky HL. Association of filamin A and vimentin with hepatitis C virus proteins in infected human hepatocytes. J Viral Hepat 2011; 18:e568-77. [PMID: 21914078 DOI: 10.1111/j.1365-2893.2011.01487.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Chronic hepatitis C (CHC) infection caused by hepatitis C virus (HCV) is a major cause of liver disease and remains a major therapeutic challenge. A variety of host proteins interact with HCV proteins. The definitive role of cytoskeletal (CS) proteins in HCV infection remains to be determined. In this study, our aim was to determine the expression profile of differentially regulated and expressed selected CS proteins and their association with HCV proteins in infected hepatocytes as possible therapeutic targets. Using proteomics, qRT-PCR, Western blot and immunofluorescence techniques, we revealed that filamin A (fila) and vimentin (vim) were prominently increased proteins in HCV-expressing human hepatoma cells compared with parental cells and in liver biopsies from patients with CHC vs controls. HCV nonstructural (NS) 3 and NS5A proteins were associated with fila, while core protein partially with fila and vim. Immunoprecipitation showed interactions among fila and NS3 and NS5A proteins. Cells treated with interferon-α showed a dose- and time-dependent decrease in CS and HCV proteins. NS proteins clustered at the perinuclear region following cytochalasin b treatment, whereas disperse cytoplasmic and perinuclear distribution was observed in the no-treatment group. This study demonstrates and signifies that changes occur in the expression of CS proteins in HCV-infected hepatocytes and, for the first time, shows the up-regulation and interaction of fila with HCV proteins. Association between CS and HCV proteins may have implications in future design of CS protein-targeted therapy for the treatment for HCV infection.
Collapse
Affiliation(s)
- S Ghosh
- The Liver-Biliary-Pancreatic Center and Liver, Digestive and Metabolic Disorders Laboratory, Carolinas Medical Center, Charlotte, NC 28203, USA.
| | | | | | | | | | | |
Collapse
|
81
|
Hepatitis C virus-induced cancer stem cell-like signatures in cell culture and murine tumor xenografts. J Virol 2011; 85:12292-303. [PMID: 21937640 DOI: 10.1128/jvi.05920-11] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C virus (HCV) infection is a prominent risk factor for the development of hepatocellular carcinoma (HCC). Similar to most solid tumors, HCCs are believed to contain poorly differentiated cancer stem cell-like cells (CSCs) that initiate tumorigenesis and confer resistance to chemotherapy. In these studies, we demonstrate that the expression of an HCV subgenomic replicon in cultured cells results in the acquisition of CSC traits. These traits include enhanced expression of doublecortin and CaM kinase-like-1 (DCAMKL-1), Lgr5, CD133, α-fetoprotein, cytokeratin-19 (CK19), Lin28, and c-Myc. Conversely, curing of the replicon from these cells results in diminished expression of these factors. The putative stem cell marker DCAMKL-1 is also elevated in response to the overexpression of a cassette of pluripotency factors. The DCAMKL-1-positive cells isolated from hepatoma cell lines by fluorescence-activated cell sorting (FACS) form spheroids in Matrigel. The HCV RNA abundance and NS5B levels are significantly reduced by the small interfering RNA (siRNA)-led depletion of DCAMKL-1. We further demonstrate that HCV replicon-expressing cells initiate distinct tumor phenotypes compared to the tumors initiated by parent cells lacking the replicon. This HCV-induced phenotype is characterized by high-level expression/coexpression of DCAMKL-1, CK19, α-fetoprotein, and active c-Src. The results obtained by the analysis of liver tissues from HCV-positive patients and liver tissue microarrays reiterate these observations. In conclusion, chronic HCV infection appears to predispose cells toward the path of acquiring cancer stem cell-like traits by inducing DCAMKL-1 and hepatic progenitor and stem cell-related factors. DCAMKL-1 also represents a novel cellular target for combating HCV-induced hepatocarcinogenesis.
Collapse
|
82
|
Martín-Acebes MA, Blázquez AB, Jiménez de Oya N, Escribano-Romero E, Saiz JC. West Nile virus replication requires fatty acid synthesis but is independent on phosphatidylinositol-4-phosphate lipids. PLoS One 2011; 6:e24970. [PMID: 21949814 PMCID: PMC3176790 DOI: 10.1371/journal.pone.0024970] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 08/19/2011] [Indexed: 12/19/2022] Open
Abstract
West Nile virus (WNV) is a neurovirulent mosquito-borne flavivirus, which main natural hosts are birds but it also infects equines and humans, among other mammals. As in the case of other plus-stranded RNA viruses, WNV replication is associated to intracellular membrane rearrangements. Based on results obtained with a variety of viruses, different cellular processes have been shown to play important roles on these membrane rearrangements for efficient viral replication. As these processes are related to lipid metabolism, fatty acid synthesis, as well as generation of a specific lipid microenvironment enriched in phosphatidylinositol-4-phosphate (PI4P), has been associated to it in other viral models. In this study, intracellular membrane rearrangements following infection with a highly neurovirulent strain of WNV were addressed by means of electron and confocal microscopy. Infection of WNV, and specifically viral RNA replication, were dependent on fatty acid synthesis, as revealed by the inhibitory effect of cerulenin and C75, two pharmacological inhibitors of fatty acid synthase, a key enzyme of this process. However, WNV infection did not induce redistribution of PI4P lipids, and PI4P did not localize at viral replication complex. Even more, WNV multiplication was not inhibited by the use of the phosphatidylinositol-4-kinase inhibitor PIK93, while infection by the enterovirus Coxsackievirus B5 was reduced. Similar features were found when infection by other flavivirus, the Usutu virus (USUV), was analyzed. These features of WNV replication could help to design specific antiviral approaches against WNV and other related flaviviruses.
Collapse
Affiliation(s)
- Miguel A Martín-Acebes
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain.
| | | | | | | | | |
Collapse
|
83
|
Distinct functions of NS5A in hepatitis C virus RNA replication uncovered by studies with the NS5A inhibitor BMS-790052. J Virol 2011; 85:7312-20. [PMID: 21593143 DOI: 10.1128/jvi.00253-11] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BMS-790052, targeting nonstructural protein 5A (NS5A), is the most potent hepatitis C virus (HCV) inhibitor described to date. It is highly effective against genotype 1 replicons and also displays robust genotype 1 anti-HCV activity in the clinic (M. Gao et al., Nature 465:96-100, 2010). BMS-790052 inhibits genotype 2a JFH1 replicon cells and cell culture infectious virus with 50% effective concentrations (EC(50)s) of 46.8 and 16.1 pM, respectively. Resistance selection studies with the JFH1 replicon and virus systems identified drug-induced mutations within the N-terminal region of NS5A. F28S, L31M, C92R, and Y93H were the major resistance mutations identified; the impact of these mutations on inhibitor sensitivity between the replicon and virus was very similar. The C92R and Y93H mutations negatively impacted fitness of the JFH1 virus. Second-site replacements at NS5A residue 30 (K30E/Q) restored efficient replication of the C92R viral variant, thus demonstrating a genetic interaction between NS5A residues 30 and 92. By using a trans-complementation assay with JFH1 replicons encoding inhibitor-sensitive and inhibitor-resistant NS5A proteins, we provide genetic evidence that NS5A performs the following two distinct functions in HCV RNA replication: a cis-acting function that likely occurs as part of the HCV replication complex and a trans-acting function that may occur outside the replication complex. The cis-acting function is likely performed by basally phosphorylated NS5A, while the trans-acting function likely requires hyperphosphorylation. Our data indicate that BMS-790052 blocks the cis-acting function of NS5A. Since BMS-790052 also impairs JFH1 NS5A hyperphosphorylation, it likely also blocks the trans-acting function.
Collapse
|
84
|
Hepatitis C virus co-opts Ras-GTPase-activating protein-binding protein 1 for its genome replication. J Virol 2011; 85:6996-7004. [PMID: 21561913 DOI: 10.1128/jvi.00013-11] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We recently reported that Ras-GTPase-activating protein-binding protein 1 (G3BP1) interacts with hepatitis C virus (HCV) nonstructural protein (NS)5B and the 5' end of the HCV minus-strand RNA. In the current study we confirmed these observations using immunoprecipitation and RNA pulldown assays, suggesting that G3BP1 might be an HCV replication complex (RC) component. In replicon cells, transfected G3BP1 interacts with multiple HCV nonstructural proteins. Using immunostaining and confocal microscopy, we demonstrate that G3BP1 is colocalized with HCV RCs in replicon cells. Small interfering RNA (siRNA)-mediated knockdown of G3BP1 moderately reduces established HCV RNA replication in HCV replicon cells and dramatically reduces HCV replication-dependent colony formation and cell-culture-produced HCV (HCVcc) infection. In contrast, knockdown of G3BP2 has no effect on HCVcc infection. Transient replication experiments show that G3BP1 is involved in HCV genome amplification. Thus, G3BP1 is associated with HCV RCs and may be co-opted as a functional RC component for viral replication. These findings may facilitate understanding of the molecular mechanisms of HCV genome replication.
Collapse
|
85
|
Small molecules targeting hepatitis C virus-encoded NS5A cause subcellular redistribution of their target: insights into compound modes of action. J Virol 2011; 85:6353-68. [PMID: 21507963 DOI: 10.1128/jvi.00215-11] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The current standard of care for hepatitis C virus (HCV)-infected patients consists of lengthy treatment with interferon and ribavirin. To increase the effectiveness of HCV therapy, future regimens will incorporate multiple direct-acting antiviral (DAA) drugs. Recently, the HCV-encoded NS5A protein has emerged as a promising DAA target. Compounds targeting NS5A exhibit remarkable potency in vitro and demonstrate early clinical promise, suggesting that NS5A inhibitors could feature in future DAA combination therapies. Since the mechanisms through which these molecules operate are unknown, we have used NS5A inhibitors as tools to investigate their modes of action. Analysis of replicon-containing cells revealed dramatic phenotypic alterations in NS5A localization following treatment with NS5A inhibitors; NS5A was redistributed from the endoplasmic reticulum to lipid droplets. The NS5A relocalization did not occur in cells treated with other classes of HCV inhibitors, and NS5A-targeting molecules did not cause similar alterations in the localization of other HCV-encoded proteins. Time course analysis of the redistribution of NS5A revealed that the transfer of protein to lipid droplets was concomitant with the onset of inhibition, as judged by the kinetic profiles for these compounds. Furthermore, analysis of the kinetic profile of inhibition for a panel of test molecules permitted the separation of compounds into different kinetic classes based on their modes of action. Results from this approach suggested that NS5A inhibitors perturbed the function of new replication complexes, rather than acting on preformed complexes. Taken together, our data reveal novel biological consequences of NS5A inhibition, which may help enable the development of future assay platforms for the identification of new and/or different NS5A inhibitors.
Collapse
|
86
|
Watanabe N, Aizaki H, Matsuura T, Kojima S, Wakita T, Suzuki T. Hepatitis C virus RNA replication in human stellate cells regulates gene expression of extracellular matrix-related molecules. Biochem Biophys Res Commun 2011; 407:135-40. [PMID: 21371436 DOI: 10.1016/j.bbrc.2011.02.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2011] [Accepted: 02/25/2011] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver disease, including chronic hepatitis, fibrosis, and cirrhosis. Fibrosis often develops in HCV-infected livers and ultimately leads to cirrhosis and carcinoma. During fibrosis, hepatic stellate cells (HSC) play important roles in the control of extracellular matrix synthesis and degradation in fibrotic livers. In this study, we established a subgenomic replicon (SGR) cell line with human hepatic stellate cells to investigate the effect of HCV RNA replication on HSC. Isolated SGR clones contained HCV RNA copy numbers ranging from 10(4) to 10(7) per μg total RNA, and long-term culture of low-copy number SGR clones resulted in markedly increased HCV RNA copy numbers. Furthermore, HCV RNA replication affected gene expression of extracellular matrix-related molecules in both hepatic stellate cells and hepatic cells, suggesting that HCV RNA replication and/or HCV proteins directly contribute to liver fibrosis. The HCV RNA-replicating hepatic stellate cell line isolated in this study will be useful for investigating hepatic stellate cell functions and HCV replication machinery.
Collapse
Affiliation(s)
- Noriyuki Watanabe
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
87
|
Ma Y, Anantpadma M, Timpe JM, Shanmugam S, Singh SM, Lemon SM, Yi M. Hepatitis C virus NS2 protein serves as a scaffold for virus assembly by interacting with both structural and nonstructural proteins. J Virol 2011; 85:86-97. [PMID: 20962101 PMCID: PMC3014171 DOI: 10.1128/jvi.01070-10] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 10/05/2010] [Indexed: 11/20/2022] Open
Abstract
Many aspects of the assembly of hepatitis C virus (HCV) remain incompletely understood. To characterize the role of NS2 in the production of infectious virus, we determined NS2 interaction partners among other HCV proteins during productive infection. Pulldown assays showed that NS2 forms complexes with both structural and nonstructural proteins, including E1, E2, p7, NS3, and NS5A. Confocal microscopy also demonstrated that NS2 colocalizes with E1, E2, and NS5A in dot-like structures near lipid droplets. However, NS5A did not coprecipitate with E2 and interacted only weakly with NS3 in pulldown assays. Also, there was no demonstrable interaction between p7 and E2 or NS3 in such assays. Therefore, NS2 is uniquely capable of interacting with both structural and nonstructural proteins. Among mutations in p7, NS2, and NS3 that prevent production of infectious virus, only p7 mutations significantly reduced NS2-mediated protein interactions. These p7 mutations altered the intracellular distribution of NS2 and E2 and appeared to modulate the membrane topology of the C-terminal domain of NS2. These results suggest that NS2 acts to coordinate virus assembly by mediating interactions between envelope proteins and NS3 and NS5A within replication complexes adjacent to lipid droplets, where virus particle assembly is thought to occur. p7 may play an accessory role by regulating NS2 membrane topology, which is important for NS2-mediated protein interactions and therefore NS2 function.
Collapse
Affiliation(s)
- Yinghong Ma
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0610
| | - Manu Anantpadma
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0610
| | - Jennifer M. Timpe
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0610
| | - Saravanabalaji Shanmugam
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0610
| | - Sher M. Singh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0610
| | - Stanley M. Lemon
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0610
| | - MinKyung Yi
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555-0610
| |
Collapse
|
88
|
Pezacki JP, Singaravelu R, Lyn RK. Host-virus interactions during hepatitis C virus infection: a complex and dynamic molecular biosystem. MOLECULAR BIOSYSTEMS 2010; 6:1131-42. [PMID: 20549003 DOI: 10.1039/b924668c] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The hepatitis C virus (HCV) is a global health issue with no vaccine available and limited clinical treatment options. Like other obligate parasites, HCV requires host cellular components of an infected individual to propagate. These host-virus interactions during HCV infection are complex and dynamic and involve the hijacking of host cell environments, enzymes and pathways. Understanding this unique molecular biosystem has the potential to yield new and exciting strategies for therapeutic intervention. Advances in genomics and proteomics have opened up new possibilities for the rapid measurement of global changes at the transcriptional and translational levels during infection. However, these techniques only yield snapshots of host-virus interactions during HCV infection. Other new methods that involve the imaging of biomolecular interactions during HCV infection are required to identify key interactions that may be transient and dynamic. Herein we highlight systems biology based strategies that have helped to identify key host-virus interactions during HCV replication and infection. Novel biophysical tools are also highlighted for identification and visualization of activities and interactions between HCV and its host hepatocyte. As some of these methods mature, we expect them to pave the way forward for further exploration of this complex biosystem and elucidation of mechanisms for HCV pathogenesis and carcinogenesis.
Collapse
Affiliation(s)
- John Paul Pezacki
- Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Dr., Ottawa, Ontario, Canada.
| | | | | |
Collapse
|
89
|
Lyn RK, Kennedy DC, Stolow A, Ridsdale A, Pezacki JP. Dynamics of lipid droplets induced by the hepatitis C virus core protein. Biochem Biophys Res Commun 2010; 399:518-24. [DOI: 10.1016/j.bbrc.2010.07.101] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 07/24/2010] [Indexed: 01/22/2023]
|
90
|
Tanaka Y, Mori Y, Tani H, Abe T, Moriishi K, Kojima H, Nagano T, Okabe T, Suzuki T, Tatsumi M, Matsuura Y. Establishment of an indicator cell system for hepatitis C virus. Microbiol Immunol 2010; 54:206-20. [PMID: 20377749 DOI: 10.1111/j.1348-0421.2010.00209.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Although a cell culture system for HCV JFH-1 strain has been developed, no robust cell culture system for serum-derived HCV is available. In this study, we have established systems capable of monitoring infection with JFH-1 virus based on specific reporter gene expression through proteolysis of chimeric transcription factors by HCV NS3/4A protease. We utilized a transcriptional factor Gal4-TBP that synergistically enhances transcription of the GAL4UAS and HIV-1 LTR tandem promoter with the Tat protein. We constructed chimeric Tat and Gal4-TBP transcription factors containing the HCV NS3/4A cleavage sequence of a mitochondria-resident IPS-1, but not those of the HCV polyprotein, and manipulated them to localize in the ER. Upon infection with JFH-1 virus, the transcription factors were efficiently cleaved by HCV protease, migrated into the nucleus and activated the reporter gene under the tandem promoter. Upon infection with JFH-1 virus, the Huh7OK1/TG-Luc cell line carrying the transcription factors and a luciferase gene under the promoter expressed luciferase in a dose-dependent manner in close correlation with HCV RNA replication. Huh7OK1/TG-LNGFR cells carrying the transcription factors and a cDNA of human low affinity nerve growth factor receptor under the promoter were selectively concentrated by immunomagnetic cell sorting upon infection with JFH-1 virus. These results indicate that the chimeric constructs bearing the ER-resident IPS-1 sequence are specifically recognized and efficiently cleaved by HCV protease and are harnessed for detection of HCV replication and for recovery of HCV-infected cells. This strategy may be applicable for the establishment of cell culture systems for the isolation of serum-derived HCV.
Collapse
Affiliation(s)
- Yoshinori Tanaka
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Hsu NY, Ilnytska O, Belov G, Santiana M, Chen YH, Takvorian PM, Pau C, van der Schaar H, Kaushik-Basu N, Balla T, Cameron CE, Ehrenfeld E, van Kuppeveld FJM, Altan-Bonnet N. Viral reorganization of the secretory pathway generates distinct organelles for RNA replication. Cell 2010; 141:799-811. [PMID: 20510927 PMCID: PMC2982146 DOI: 10.1016/j.cell.2010.03.050] [Citation(s) in RCA: 560] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 01/12/2010] [Accepted: 03/18/2010] [Indexed: 01/31/2023]
Abstract
Many RNA viruses remodel intracellular membranes to generate specialized sites for RNA replication. How membranes are remodeled and what properties make them conducive for replication are unknown. Here we show how RNA viruses can manipulate multiple components of the cellular secretory pathway to generate organelles specialized for replication that are distinct in protein and lipid composition from the host cell. Specific viral proteins modulate effector recruitment by Arf1 GTPase and its guanine nucleotide exchange factor GBF1, promoting preferential recruitment of phosphatidylinositol-4-kinase IIIbeta (PI4KIIIbeta) to membranes over coat proteins, yielding uncoated phosphatidylinositol-4-phosphate (PI4P) lipid-enriched organelles. The PI4P-rich lipid microenvironment is essential for both enteroviral and flaviviral RNA replication; PI4KIIIbeta inhibition interferes with this process; and enteroviral RNA polymerases specifically bind PI4P. These findings reveal how RNA viruses can selectively exploit specific elements of the host to form specialized organelles where cellular phosphoinositide lipids are key to regulating viral RNA replication.
Collapse
Affiliation(s)
- Nai-Yun Hsu
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Phosphatidylinositol 3-kinase-, actin-, and microtubule-dependent transport of Semliki Forest Virus replication complexes from the plasma membrane to modified lysosomes. J Virol 2010; 84:7543-57. [PMID: 20484502 DOI: 10.1128/jvi.00477-10] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Like other positive-strand RNA viruses, alphaviruses replicate their genomes in association with modified intracellular membranes. Alphavirus replication sites consist of numerous bulb-shaped membrane invaginations (spherules), which contain the double-stranded replication intermediates. Time course studies with Semliki Forest virus (SFV)-infected cells were combined with live-cell imaging and electron microscopy to reveal that the replication complex spherules of SFV undergo an unprecedented large-scale movement between cellular compartments. The spherules first accumulated at the plasma membrane and were then internalized using an endocytic process that required a functional actin-myosin network, as shown by blebbistatin treatment. Wortmannin and other inhibitors indicated that the internalization of spherules also required the activity of phosphatidylinositol 3-kinase. The spherules therefore represent an unusual type of endocytic cargo. After endocytosis, spherule-containing vesicles were highly dynamic and had a neutral pH. These primary carriers fused with acidic endosomes and moved long distances on microtubules, in a manner prevented by nocodazole. The result of the large-scale migration was the formation of a very stable compartment, where the spherules were accumulated on the outer surfaces of unusually large and static acidic vacuoles localized in the pericentriolar region. Our work highlights both fundamental similarities and important differences in the processes that lead to the modified membrane compartments in cells infected by distinct groups of positive-sense RNA viruses.
Collapse
|
93
|
Shetty S, Kim S, Shimakami T, Lemon SM, Mihailescu MR. Hepatitis C virus genomic RNA dimerization is mediated via a kissing complex intermediate. RNA (NEW YORK, N.Y.) 2010; 16:913-25. [PMID: 20360391 PMCID: PMC2856886 DOI: 10.1261/rna.1960410] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 02/07/2010] [Indexed: 05/18/2023]
Abstract
With over 200 million people infected with hepatitis C virus (HCV) worldwide, there is a need for more effective and better-tolerated therapeutic strategies. The HCV genome is a positive-sense; single-stranded RNA encoding a large polyprotein cleaved at multiple sites to produce at least ten proteins, among them an error-prone RNA polymerase that confers a high mutation rate. Despite considerable overall sequence diversity, in the 3'-untranslated region of the HCV genomic RNA there is a 98-nucleotide (nt) sequence named X RNA, the first 55 nt of which (X55 RNA) are 100% conserved among all HCV strains. The X55 region has been suggested to be responsible for in vitro dimerization of the genomic RNA in the presence of the viral core protein, although the mechanism by which this occurs is unknown. In this study, we analyzed the X55 region and characterized the mechanism by which it mediates HCV genomic RNA dimerization. Similar to a mechanism proposed previously for the human immunodeficiency 1 virus (HIV-1) genome, we show that dimerization of the HCV genome involves formation of a kissing complex intermediate, which is converted to a more stable extended duplex conformation in the presence of the core protein. Mutations in the dimer linkage sequence loop sequence that prevent RNA dimerization in vitro significantly reduced but did not completely ablate the ability of HCV RNA to replicate or produce infectious virus in transfected cells.
Collapse
Affiliation(s)
- Sumangala Shetty
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, Pennsylvania 15282, USA
| | | | | | | | | |
Collapse
|
94
|
Abente EJ, Sosnovtsev SV, Bok K, Green KY. Visualization of feline calicivirus replication in real-time with recombinant viruses engineered to express fluorescent reporter proteins. Virology 2010; 400:18-31. [PMID: 20137802 PMCID: PMC2855553 DOI: 10.1016/j.virol.2009.12.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 12/15/2009] [Accepted: 12/31/2009] [Indexed: 11/30/2022]
Abstract
Caliciviruses are non-enveloped, icosahedral viruses with a single-stranded, positive sense RNA genome. Transposon-mediated insertional mutagenesis was used to insert a transprimer sequence into random sites of an infectious full-length cDNA clone of the feline calicivirus (FCV) genome. A site in the LC gene (encoding the capsid leader protein) of the FCV genome was identified that could tolerate foreign insertions, and two viable recombinant FCV variants expressing LC fused either to AcGFP, or DsRedFP were recovered. The effects of the insertions on LC processing, RNA replication, and stability of the viral genome were analyzed, and the progression of a calicivirus single infection and co-infection were captured by real-time imaging fluorescent microscopy. The ability to engineer viable recombinant caliciviruses expressing foreign markers enables new approaches to investigate virus and host cell interactions, as well as studies of viral recombination, one of the driving forces of calicivirus evolution.
Collapse
Affiliation(s)
- Eugenio J. Abente
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20892
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | | | - Karin Bok
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20892
| | - Kim Y. Green
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20892
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| |
Collapse
|
95
|
Horner SM, Gale M. Intracellular innate immune cascades and interferon defenses that control hepatitis C virus. J Interferon Cytokine Res 2010; 29:489-98. [PMID: 19708811 DOI: 10.1089/jir.2009.0063] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hepatitis C virus (HCV) is a global public health problem that mediates a persistent infection in nearly 200 million people. HCV is efficient in establishing chronicity due in part to the inefficiency of the host immune system in controlling and counteracting HCV-mediated evasion strategies. HCV persistence is linked to the ability of the virus to suppress the RIG-I pathway and interferon production from infected hepatocytes, thus evading innate immune defenses within the infected cell. This review describes the virus and host processes that regulate the RIG-I pathway during HCV infection. An understanding of these HCV-host interactions could lead to more effective therapies for HCV designed to reactivate the host immune response following HCV infection.
Collapse
Affiliation(s)
- Stacy M Horner
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | | |
Collapse
|
96
|
Regulation of de novo-initiated RNA synthesis in hepatitis C virus RNA-dependent RNA polymerase by intermolecular interactions. J Virol 2010; 84:5923-35. [PMID: 20375156 DOI: 10.1128/jvi.02446-09] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The hepatitis C virus (HCV) RNA-dependent RNA polymerase (RdRp) has been proposed to change conformations in association with RNA synthesis and to interact with cellular proteins. In vitro, the RdRp can initiate de novo from the ends of single-stranded RNA or extend a primed RNA template. The interactions between the Delta1 loop and thumb domain in NS5B are required for de novo initiation, although it is unclear whether these interactions are within an NS5B monomer or are part of a higher-order NS5B oligomeric complex. This work seeks to address how polymerase conformation and/or oligomerization affects de novo initiation. We have shown that an increasing enzyme concentration increases de novo initiation by the genotype 1b and 2a RdRps while primer extension reactions are not affected or inhibited under similar conditions. Initiation-defective mutants of the HCV polymerase can increase de novo initiation by the wild-type (WT) polymerase. GTP was also found to stimulate de novo initiation. Our results support a model in which the de novo initiation-competent conformation of the RdRp is stimulated by oligomeric contacts between individual subunits. Using electron microscopy and single-molecule reconstruction, we attempted to visualize the low-resolution conformations of a dimer of a de novo initiation-competent HCV RdRp.
Collapse
|
97
|
Berke JM, Fenistein D, Pauwels F, Bobbaers R, Lenz O, Lin TI, Krausz E, Fanning G. Development of a high-content screening assay to identify compounds interfering with the formation of the hepatitis C virus replication complex. J Virol Methods 2010; 165:268-76. [PMID: 20153373 DOI: 10.1016/j.jviromet.2010.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 01/29/2010] [Accepted: 02/03/2010] [Indexed: 10/19/2022]
Abstract
The hepatitis C virus (HCV) replicates its genome on a membrane-associated replication complex. These complexes are represented by "dot-like" structures on the endoplasmic reticulum when standard fluorescence microscopy techniques are applied. To screen compound libraries for inhibitors interfering with the formation of the HCV replication complex independent of RNA replication, an image-based high-content screening assay was developed utilizing inducible expression of the HCV non-structural proteins NS3-5B in an U2-OS Tet-On cell line. An eGFP was fused to NS5A for the detection of replication complexes. The cell line was tightly regulated and the eGFP insertion within NS5A did not alter polyprotein processing. The NS5AeGFP signal colocalized with other non-structural proteins in "dot-like" structures. Accompanying image analysis tools were developed enabling the detection of changes in replication complex formation. Finally, the addition of a HCV NS3/4A protease inhibitor resulted in a dose-dependent reduction of "dot-like" structures demonstrating the practicability of the assay.
Collapse
|
98
|
Joyce MA, Tyrrell DLJ. The cell biology of hepatitis C virus. Microbes Infect 2010; 12:263-71. [PMID: 20080204 DOI: 10.1016/j.micinf.2009.12.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 12/22/2009] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus infects 3% of the world's population and has a variable disease course with potentially sever outcomes, liver failure and hepatocellular carcinoma. The influence of HCV the biology of infected hepatocytes is now just becoming known. This review will focus on effect of HCV on host cells.
Collapse
Affiliation(s)
- Michael A Joyce
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada.
| | | |
Collapse
|
99
|
Gouttenoire J, Penin F, Moradpour D. Hepatitis C virus nonstructural protein 4B: a journey into unexplored territory. Rev Med Virol 2010; 20:117-29. [DOI: 10.1002/rmv.640] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
100
|
Abstract
This review focuses on the extensive membrane and organelle rearrangements that have been observed in plant cells infected with RNA viruses. The modifications generally involve the formation of spherules, vesicles, and/or multivesicular bodies associated with various organelles such as the endoplasmic reticulum and peroxisomes. These virus-induced organelles house the viral RNA replication complex and are known as virus factories or viroplasms. Membrane and organelle alterations are attributed to the action of one or two viral proteins, which additionally act as a scaffold for the assembly of a large complex of proteins of both viral and host origin and viral RNA. Some virus factories have been shown to align with and traffic along microfilaments. In addition to viral RNA replication, the factories may be involved in other processes such as viral RNA translation and cell-to-cell virus transport. Confining the process of RNA replication to a specific location may also prevent the activation of certain host defense functions.
Collapse
Affiliation(s)
- Jean-François Laliberté
- INRS-Institut Armand-Frappier, Institut National de la Recherche Scientifique, Laval, Québec, Canada H7V 1B7.
| | | |
Collapse
|