51
|
Giersch K, Dandri M. Hepatitis B and Delta Virus: Advances on Studies about Interactions between the Two Viruses and the Infected Hepatocyte. J Clin Transl Hepatol 2015; 3:220-9. [PMID: 26623269 PMCID: PMC4663204 DOI: 10.14218/jcth.2015.00018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/11/2015] [Accepted: 07/12/2015] [Indexed: 12/14/2022] Open
Abstract
The mechanisms determining persistence of hepatitis B virus (HBV) infection and long-term pathogenesis of HBV-associated liver disease appear to be multifactorial. Although viral replication can be efficiently suppressed by the antiviral treatments currently available, viral clearance is generally not achieved since HBV has developed unique replication strategies, enabling persistence of its genome within the infected hepatocytes. Moreover, no direct antiviral therapy exists for the more than 15 million people worldwide that are also coinfected with the hepatitis delta virus (HDV), a defective virus that needs the HBV envelope proteins for propagation. The limited availability of robust HBV and HDV infection systems has hindered the understanding of the complex network of virus-virus and virus-host interactions that are established in the course of infection and slowed down progress in drug development. Since chronic HBV/HDV coinfection leads to the most severe form of chronic viral hepatitis, elucidation of the molecular mechanisms regulating virus-host interplay and pathogenesis are urgently needed. This article summarizes the current knowledge regarding the interactions among HBV, HDV, and the infected target cell and discusses the dependence of HDV on HBV activity and possible future therapeutic approaches.
Collapse
Affiliation(s)
- Katja Giersch
- Department of Internal Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maura Dandri
- Department of Internal Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel site, Germany
- Correspondence to: Maura Dandri, Department of Internal Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246 Hamburg, Germany. Tel: +49-40741052949, Fax: +49-40741057232, E-mail:
| |
Collapse
|
52
|
Zeisel MB, Lucifora J, Mason WS, Sureau C, Beck J, Levrero M, Kann M, Knolle PA, Benkirane M, Durantel D, Michel ML, Autran B, Cosset FL, Strick-Marchand H, Trépo C, Kao JH, Carrat F, Lacombe K, Schinazi RF, Barré-Sinoussi F, Delfraissy JF, Zoulim F. Towards an HBV cure: state-of-the-art and unresolved questions--report of the ANRS workshop on HBV cure. Gut 2015; 64:1314-26. [PMID: 25670809 DOI: 10.1136/gutjnl-2014-308943] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/10/2015] [Indexed: 12/11/2022]
Abstract
HBV infection is a major cause of liver cirrhosis and hepatocellular carcinoma. Although HBV infection can be efficiently prevented by vaccination, and treatments are available, to date there is no reliable cure for the >240 million individuals that are chronically infected worldwide. Current treatments can only achieve viral suppression, and lifelong therapy is needed in the majority of infected persons. In the framework of the French National Agency for Research on AIDS and Viral Hepatitis 'HBV Cure' programme, a scientific workshop was held in Paris in June 2014 to define the state-of-the-art and unanswered questions regarding HBV pathobiology, and to develop a concerted strategy towards an HBV cure. This review summarises our current understanding of HBV host-interactions leading to viral persistence, as well as the roadblocks to be overcome to ultimately address unmet medical needs in the treatment of chronic HBV infection.
Collapse
Affiliation(s)
- Mirjam B Zeisel
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France Université de Strasbourg, Strasbourg, France
| | - Julie Lucifora
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
| | | | - Camille Sureau
- INTS, Laboratoire de Virologie Moléculaire, Paris, France
| | - Jürgen Beck
- Department of Internal Medicine 2/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Massimo Levrero
- Center for Life Nanosciences (CNLS)-IIT/Sapienza, Rome, Italy Laboratory of Gene Expression, Department of Internal Medicine (DMISM), Sapienza University of Rome, Italy
| | - Michael Kann
- Université de Bordeaux, Microbiologie fondamentale et Pathogénicité, UMR 5234, Bordeaux, France CNRS, Microbiologie fondamentale et Pathogénicité, UMR 5234, Bordeaux, France CHU de Bordeaux, Bordeaux, France
| | - Percy A Knolle
- Technische Universität München, Institut für Molekulare Immunologie, München, Germany
| | - Monsef Benkirane
- Institut de Génétique Humaine, Laboratoire de Virologie Moléculaire, CNRS UPR1142, Montpellier, France
| | - David Durantel
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
| | - Marie-Louise Michel
- Laboratoire de Pathogenèse des Virus de l'Hépatite B, Département de Virologie, Institut Pasteur, Paris, France
| | - Brigitte Autran
- Laboratory of Immunity and Infection, Inserm U945, Paris, France Laboratory of Immunity and Infection, UPMC University Paris 06, Unité mixte de recherche-S945, Paris, France Inserm, IFR 113, Immunité-Cancer-Infection, Paris, France
| | - François-Loïc Cosset
- CIRI-International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France. Inserm, U1111, Lyon, France Ecole Normale Supérieure de Lyon, Lyon, France. CNRS, UMR5308, Lyon, France LabEx Ecofect, Université de Lyon, Lyon, France
| | | | - Christian Trépo
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France Department of Hepatology, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| | - Jia-Horng Kao
- Department of Internal Medicine, Department of Medical Research, Graduate Institute of Clinical Medicine, and Hepatitis Research Center, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Fabrice Carrat
- Inserm, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France Sorbonne Universités, UPMC Univ Paris 06, Paris, France Assistance Publique Hôpitaux de Paris, Hôpital Saint Antoine, Paris, France
| | - Karine Lacombe
- Inserm, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France Sorbonne Universités, UPMC Univ Paris 06, Paris, France Assistance Publique Hôpitaux de Paris, Hôpital Saint Antoine, Paris, France
| | - Raymond F Schinazi
- Center for AIDS Research, Emory University School of Medicine and Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Françoise Barré-Sinoussi
- Inserm and Unit of Regulation of Retroviral Infections, Department of Virology, Institut Pasteur, Paris, France
| | | | - Fabien Zoulim
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France Department of Hepatology, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
53
|
Pei RJ, Chen XW, Lu MJ. Control of hepatitis B virus replication by interferons and Toll-like receptor signaling pathways. World J Gastroenterol 2014; 20:11618-11629. [PMID: 25206268 PMCID: PMC4155354 DOI: 10.3748/wjg.v20.i33.11618] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 12/23/2013] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection is one of the major causes of liver diseases, affecting more than 350 million people worldwide. The interferon (IFN)-mediated innate immune responses could restrict HBV replication at the different steps of viral life cycle. Indeed, IFN-α has been successfully used for treatment of patients with chronic hepatitis B. However, the role of the innate immune response in HBV replication and the mechanism of the anti-HBV effect of IFN-α are not completely explored. In this review, we summarized the currently available knowledge about the IFN-mediated anti-HBV effect in the HBV life cycle and the possible effectors downstream the IFN signaling pathway. The antiviral effect of Toll-like receptors (TLRs) in HBV replication is briefly discussed. The strategies exploited by HBV to evade the IFN- and TLR-mediated antiviral actions are summarized.
Collapse
|
54
|
Lupberger J, Schaedler S, Peiran A, Hildt E. Identification and characterization of a novel bipartite nuclear localization signal in the hepatitis B virus polymerase. World J Gastroenterol 2013; 19:8000-8010. [PMID: 24307793 PMCID: PMC3848147 DOI: 10.3748/wjg.v19.i44.8000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/10/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To characterize the nuclear import of hepatitis B virus (HBV) polymerase (P) and its relevance for the viral life cycle.
METHODS: Sequence analysis was performed to predict functional motives within P. Phosphorylation of P was analyzed by in vitro phosphorylation. Phosphorylation site and nuclear localization signal (NLS) were destroyed by site directed mutagenesis. Functionality of the identified NLS was analyzed by confocal fluorescence microscopy and characterizing the karyopherin binding. Relevance of the structural motives for viral life cycle was studied by infection of primary Tupaia hepatocytes with HBV.
RESULTS: We identified by sequence alignment and functional experiments a conserved bipartite NLS containing a casein kinase II (CKII) phosphorylation site located within the terminal protein domain (TP) of the HBV polymerase. Inhibition of CKII impairs the functionality of this NLS and thereby prevents the nuclear import of the polymerase. Binding of the import factor karyopherin-α2 to the polymerase depends on its CKII-mediated phosphorylation of the bipartite NLS. In HBV-infected primary Tupaia hepatocytes CKII inhibition in the early phase (post entry phase) of the infection process prevents the establishment of the infection.
CONCLUSION: Based on these data it is suggested that during HBV infection the final import of the genome complex into the nucleus is mediated by a novel bipartite NLS localized in the TP domain of HBV polymerase.
Collapse
|
55
|
Viral subversion of the nuclear pore complex. Viruses 2013; 5:2019-42. [PMID: 23959328 PMCID: PMC3761240 DOI: 10.3390/v5082019] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/29/2013] [Accepted: 08/08/2013] [Indexed: 12/17/2022] Open
Abstract
The nuclear pore complex (NPC) acts as a selective barrier between the nucleus and the cytoplasm and is responsible for mediating communication by regulating the transport of RNA and proteins. Numerous viral pathogens have evolved different mechanisms to hijack the NPC in order to regulate trafficking of viral proteins, genomes and even capsids into and out of the nucleus thus promoting virus replication. The present review examines the different strategies and the specific nucleoporins utilized during viral infections as a means of promoting their life cycle and inhibiting host viral defenses.
Collapse
|
56
|
Inactivation of hepatitis B virus replication in cultured cells and in vivo with engineered transcription activator-like effector nucleases. Mol Ther 2013; 21:1889-97. [PMID: 23883864 PMCID: PMC3808145 DOI: 10.1038/mt.2013.170] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/14/2013] [Indexed: 02/07/2023] Open
Abstract
Chronic hepatitis B virus (HBV) infection remains an important global health problem. Stability of the episomal covalently closed circular HBV DNA (cccDNA) is largely responsible for the modest curative efficacy of available therapy. Since licensed anti-HBV drugs have a post-transcriptional mechanism of action, disabling cccDNA is potentially of therapeutic benefit. To develop this approach, we engineered mutagenic transcription activator-like effector nucleases (TALENs) that target four HBV-specific sites within the viral genome. TALENs with cognate sequences in the S or C open-reading frames (ORFs) efficiently disrupted sequences at the intended sites and suppressed markers of viral replication. Following triple transfection of cultured HepG2.2.15 cells under mildly hypothermic conditions, the S TALEN caused targeted mutation in ~35% of cccDNA molecules. Markers of viral replication were also inhibited in vivo in a murine hydrodynamic injection model of HBV replication. HBV target sites within S and C ORFs of the injected HBV DNA were mutated without evidence of toxicity. These findings are the first to demonstrate a targeted nuclease-mediated disruption of HBV cccDNA. Efficacy in vivo also indicates that these engineered nucleases have potential for use in treatment of chronic HBV infection.
Collapse
|
57
|
Ceelen L, Lieveld M, Forsyth R, Vinken M. The HepaRG cell line: a valuable in vitro tool for hepatitis virus infection studies. Hepatol Int 2013. [PMID: 26201773 DOI: 10.1007/s12072-013-9428-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hepatitis virus infections, mainly hepatitis B virus (HBV) and hepatitis C virus (HCV) infections, constitute a major problem for public health since they have a worldwide distribution and because they are associated with hepatocellular carcinoma and death. Current anti-HBV vaccines seem to be effective in the majority of people. However, an important issue waiting to be tackled nowadays is how to cure patients with chronic hepatitis B. Moreover, no vaccine is available today for the prevention of HCV infection. Therefore, the use of adequate in vitro infection systems is a prerequisite for the molecular understanding of the infection events of these viruses, which could result in the development of novel powerful therapeutics. In this respect, the HepaRG cell line exhibits a hepatocyte-like morphology and displays drug metabolism capacity similar to that of primary hepatocytes. HepaRG cells have yet been proven to be a useful tool in the study of viral infections, particularly for deciphering the mechanism of HBV entry into hepatocytes.
Collapse
Affiliation(s)
| | | | | | - Mathieu Vinken
- Department of Toxicology, Faculty of Medicine and Pharmacy, Center for Pharmaceutical Research, Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090, Brussels, Belgium
| |
Collapse
|
58
|
Paganelli M, Dallmeier K, Nyabi O, Scheers I, Kabamba B, Neyts J, Goubau P, Najimi M, Sokal EM. Differentiated umbilical cord matrix stem cells as a new in vitro model to study early events during hepatitis B virus infection. Hepatology 2013; 57:59-69. [PMID: 22898823 DOI: 10.1002/hep.26006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Accepted: 07/30/2012] [Indexed: 01/08/2023]
Abstract
UNLABELLED The role of cell differentiation state on hepatitis B virus (HBV) replication has been well demonstrated, whereas how it determines cell susceptibility to HBV entry is far less understood. We previously showed that umbilical cord matrix stem cells (UCMSC) can be differentiated towards hepatocyte-like cells in vitro. In this study we infected undifferentiated (UD-) and differentiated (D-) UCMSCs with HBV and studied the infection kinetics, comparing them to primary human hepatocytes (PHHs). UD-UCMSCs, although permissive to viral binding, had a very limited uptake capacity, whereas D-UCMSCs showed binding and uptake capabilities similar to PHHs. Likewise, asialoglycoprotein receptor (ASGPR) was up-regulated in UCMSCs upon differentiation. In D-UCMSCs, a dose-dependent inhibition of HBV binding and uptake was observed when ASGPR was saturated with known specific ligands. Subsequent viral replication was shown in D-UCMSCs but not in UD-UCMSCs. Susceptibility of UCMSCs to viral replication correlated with the degree of differentiation. Replication efficiency was low compared to PHHs, but was confirmed by (1) a dose-dependent inhibition by specific antiviral treatment using tenofovir; (2) the increase of viral RNAs along time; (3) de novo synthesis of viral proteins; and (4) secretion of infectious viral progeny. CONCLUSION UCMSCs become supportive of the entire HBV life cycle upon in vitro hepatic differentiation. Despite low replication efficiency, D-UCMSCs proved to be fully capable of HBV uptake. Overall, UCMSCs are a unique human, easily available, nontransformed, in vitro model of HBV infection that could prove useful to study early infection events and the role of the cell differentiation state on such events.
Collapse
Affiliation(s)
- Massimiliano Paganelli
- Pediatric Gastroenterology & Hepatology Unit, Université catholique de Louvain and Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Abdul F, Ndeboko B, Buronfosse T, Zoulim F, Kann M, Nielsen PE, Cova L. Potent inhibition of late stages of hepadnavirus replication by a modified cell penetrating peptide. PLoS One 2012; 7:e48721. [PMID: 23173037 PMCID: PMC3500254 DOI: 10.1371/journal.pone.0048721] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 09/28/2012] [Indexed: 12/17/2022] Open
Abstract
Cationic cell-penetrating peptides (CPPs) and their lipid domain-conjugates (CatLip) are agents for the delivery of (uncharged) biologically active molecules into the cell. Using infection and transfection assays we surprisingly discovered that CatLip peptides were able to inhibit replication of Duck Hepatitis B Virus (DHBV), a reference model for human HBV. Amongst twelve CatLip peptides we identified Deca-(Arg)8 having a particularly potent antiviral activity, leading to a drastic inhibition of viral particle secretion without detectable toxicity. Inhibition of virion secretion was correlated with a dose-dependent increase in intracellular viral DNA. Deca-(Arg)8 peptide did neither interfere with DHBV entry, nor with formation of mature nucleocapsids nor with their travelling to the nucleus. Instead, Deca-(Arg)8 caused envelope protein accumulation in large clusters as revealed by confocal laser scanning microscopy indicating severe structural changes of preS/S. Sucrose gradient analysis of supernatants from Deca-(Arg)8-treated cells showed unaffected naked viral nucleocapsids release, which was concomitant with a complete arrest of virion and surface protein-containing subviral particle secretion. This is the first report showing that a CPP is able to drastically block hepadnaviral release from infected cells by altering late stages of viral morphogenesis via interference with enveloped particle formation, without affecting naked nucleocapsid egress, thus giving a view inside the mode of inhibition. Deca-(Arg)8 may be a useful tool for elucidating the hepadnaviral secretory pathway, which is not yet fully understood. Moreover we provide the first evidence that a modified CPP displays a novel antiviral mechanism targeting another step of viral life cycle compared to what has been so far described for other enveloped viruses.
Collapse
Affiliation(s)
- Fabien Abdul
- Université de Lyon 1, Lyon, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
- CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Bénédicte Ndeboko
- Université de Lyon 1, Lyon, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
- CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Thierry Buronfosse
- Université de Lyon 1, Lyon, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
- CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- VetAgro-Sup, Marcy l'Etoile, France
| | - Fabien Zoulim
- Université de Lyon 1, Lyon, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
- CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Michael Kann
- Université de Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
- CNRS, Microbiologie fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
- CHU de Bordeaux, Bordeaux, France
| | - Peter E. Nielsen
- Department of Cellular and Molecular Medicine and Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | - Lucyna Cova
- Université de Lyon 1, Lyon, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
- CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- * E-mail:
| |
Collapse
|
60
|
Kariithi HM, Ince IA, Boeren S, Abd-Alla AMM, Parker AG, Aksoy S, Vlak JM, van Oers MM. The salivary secretome of the tsetse fly Glossina pallidipes (Diptera: Glossinidae) infected by salivary gland hypertrophy virus. PLoS Negl Trop Dis 2011; 5:e1371. [PMID: 22132244 PMCID: PMC3222630 DOI: 10.1371/journal.pntd.0001371] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/05/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The competence of the tsetse fly Glossina pallidipes (Diptera; Glossinidae) to acquire salivary gland hypertrophy virus (SGHV), to support virus replication and successfully transmit the virus depends on complex interactions between Glossina and SGHV macromolecules. Critical requisites to SGHV transmission are its replication and secretion of mature virions into the fly's salivary gland (SG) lumen. However, secretion of host proteins is of equal importance for successful transmission and requires cataloging of G. pallidipes secretome proteins from hypertrophied and non-hypertrophied SGs. METHODOLOGY/PRINCIPAL FINDINGS After electrophoretic profiling and in-gel trypsin digestion, saliva proteins were analyzed by nano-LC-MS/MS. MaxQuant/Andromeda search of the MS data against the non-redundant (nr) GenBank database and a G. morsitans morsitans SG EST database, yielded a total of 521 hits, 31 of which were SGHV-encoded. On a false discovery rate limit of 1% and detection threshold of least 2 unique peptides per protein, the analysis resulted in 292 Glossina and 25 SGHV MS-supported proteins. When annotated by the Blast2GO suite, at least one gene ontology (GO) term could be assigned to 89.9% (285/317) of the detected proteins. Five (∼1.8%) Glossina and three (∼12%) SGHV proteins remained without a predicted function after blast searches against the nr database. Sixty-five of the 292 detected Glossina proteins contained an N-terminal signal/secretion peptide sequence. Eight of the SGHV proteins were predicted to be non-structural (NS), and fourteen are known structural (VP) proteins. CONCLUSIONS/SIGNIFICANCE SGHV alters the protein expression pattern in Glossina. The G. pallidipes SG secretome encompasses a spectrum of proteins that may be required during the SGHV infection cycle. These detected proteins have putative interactions with at least 21 of the 25 SGHV-encoded proteins. Our findings opens venues for developing novel SGHV mitigation strategies to block SGHV infections in tsetse production facilities such as using SGHV-specific antibodies and phage display-selected gut epithelia-binding peptides.
Collapse
Affiliation(s)
- Henry M. Kariithi
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
- Insect Pest Control Laboratory, Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Ikbal A. Ince
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University, Wageningen, The Netherlands
| | - Adly M. M. Abd-Alla
- Insect Pest Control Laboratory, Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Andrew G. Parker
- Insect Pest Control Laboratory, Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Serap Aksoy
- Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Just M. Vlak
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | | |
Collapse
|
61
|
|
62
|
Bardens A, Döring T, Stieler J, Prange R. Alix regulates egress of hepatitis B virus naked capsid particles in an ESCRT-independent manner. Cell Microbiol 2010; 13:602-19. [PMID: 21129143 PMCID: PMC7162389 DOI: 10.1111/j.1462-5822.2010.01557.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hepatitis B virus (HBV) is an enveloped DNA virus that exploits the endosomal sorting complexes required for transport (ESCRT) pathway for budding. In addition to infectious particles, HBV‐replicating cells release non‐enveloped (nucleo)capsids, but their functional implication and pathways of release are unclear. Here, we focused on the molecular mechanisms and found that the sole expression of the HBV core protein is sufficient for capsid release. Unexpectedly, released capsids are devoid of a detectable membrane bilayer, implicating a non‐vesicular exocytosis process. Unlike virions, naked capsid budding does not require the ESCRT machinery. Rather, we identified Alix, a multifunctional protein with key roles in membrane biology, as a regulator of capsid budding. Ectopic overexpression of Alix enhanced capsid egress, while its depletion inhibited capsid release. Notably, the loss of Alix did not impair HBV production, furthermore indicating that virions and capsids use diverse export routes. By mapping of Alix domains responsible for its capsid release‐mediating activity, its Bro1 domain was found to be required and sufficient. Alix binds to core via its Bro1 domain and retained its activity even if its ESCRT‐III binding site is disrupted. Together, the boomerang‐shaped Bro1 domain of Alix appears to escort capsids without ESCRT.
Collapse
Affiliation(s)
- Andreas Bardens
- Department of Medical Microbiology and Hygiene,University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | | | | |
Collapse
|
63
|
Cohen S, Au S, Panté N. How viruses access the nucleus. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:1634-45. [PMID: 21167871 DOI: 10.1016/j.bbamcr.2010.12.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 11/24/2010] [Accepted: 12/08/2010] [Indexed: 10/25/2022]
Abstract
Many viruses depend on nuclear proteins for replication. Therefore, their viral genome must enter the nucleus of the host cell. In this review we briefly summarize the principles of nucleocytoplasmic transport, and then describe the diverse strategies used by viruses to deliver their genomes into the host nucleus. Some of the emerging mechanisms include: (1) nuclear entry during mitosis, when the nuclear envelope is disassembled, (2) viral genome release in the cytoplasm followed by entry of the genome through the nuclear pore complex (NPC), (3) capsid docking at the cytoplasmic side of the NPC, followed by genome release, (4) nuclear entry of intact capsids through the NPC, followed by genome release, and (5) nuclear entry via virus-induced disruption of the nuclear envelope. Which mechanism a particular virus uses depends on the size and structure of the virus, as well as the cellular cues used by the virus to trigger capsid disassembly and genome release. This article is part of a Special Issue entitled: Regulation of Signaling and Cellular Fate through Modulation of Nuclear Protein Import.
Collapse
Affiliation(s)
- Sarah Cohen
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
64
|
Abstract
In vitro hepatocyte models represent very useful systems in both fundamental research and various application areas. Primary hepatocytes appear as the closest model for the liver in vivo. However, they are phenotypically unstable, have a limited life span and in addition, exhibit large interdonor variability when of human origin. Hepatoma cell lines appear as an alternative but only the HepaRG cell line exhibits various functions, including major cytochrome P450 activities, at levels close to those found in primary hepatocytes. In vitro hepatocyte models have brought a substantial contribution to the understanding of the biochemistry, physiology, and cell biology of the normal and diseased liver and in various application domains such as xenobiotic metabolism and toxicity, virology, parasitology, and more generally cell therapies. In the future, new well-differentiated hepatocyte cell lines derived from tumors or from either embryonic or adult stem cells might be expected and although hepatocytes will continue to be used in various fields, these in vitro liver models should allow marked advances, especially in cell-based therapies and predictive and mechanistic hepatotoxicity of new drugs and other chemicals. All models will benefit from new developments in throughput screening based on cell chips coupled with high-content imaging and in toxicogenomics technologies.
Collapse
|
65
|
Initiation of duck hepatitis B virus infection requires cleavage by a furin-like protease. J Virol 2010; 84:4569-78. [PMID: 20181690 DOI: 10.1128/jvi.02281-09] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The entry mechanism of hepatitis B virus (HBV) has not been defined, and this impedes development of antiviral therapies aimed at an early step in the viral life cycle. HBV infection has both host and tissue specificities. For the related duck hepatitis B virus (DHBV), duck carboxypeptidase D (DCPD) has been proposed as the species-specific docking receptor, while glycine decarboxylase (DGD) may serve as a tissue-specific cofactor or secondary receptor. DGD binds to several truncated versions of the viral large envelope protein but not to the full-length protein, suggesting a need for proteolytic cleavage of the envelope protein by a furin-like proprotein convertase. In the present study, we found that transfected DCPD could confer DHBV binding to non-duck cell lines but that this was followed by rapid virus release from cells. Coexpression of furin led to DCPD cleavage and increased virus retention. Treatment of DHBV particles with endosome prepared from duck liver led to cleavage of the large envelope protein, and such viral preparation could generate a small amount of covalently closed circular DNA in LMH cells, a chicken hepatoma cell line resistant to DHBV infection. A furin inhibitor composed of decanoyl-RVKR-chloromethylketone blocked endosomal cleavage of the large envelope protein in vitro and suppressed DHBV infection of primary duck hepatocytes in vivo. These findings suggest that furin or a furin-like proprotein convertase facilitates DHBV infection by cleaving both the docking receptor and the viral large envelope protein.
Collapse
|
66
|
Schmitz A, Schwarz A, Foss M, Zhou L, Rabe B, Hoellenriegel J, Stoeber M, Panté N, Kann M. Nucleoporin 153 arrests the nuclear import of hepatitis B virus capsids in the nuclear basket. PLoS Pathog 2010; 6:e1000741. [PMID: 20126445 PMCID: PMC2813275 DOI: 10.1371/journal.ppat.1000741] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Accepted: 12/28/2009] [Indexed: 12/14/2022] Open
Abstract
Virtually all DNA viruses including hepatitis B viruses (HBV) replicate their genome inside the nucleus. In non-dividing cells, the genome has to pass through the nuclear pore complexes (NPCs) by the aid of nuclear transport receptors as e.g. importin β (karyopherin). Most viruses release their genome in the cytoplasm or at the cytosolic face of the NPC, as the diameter of their capsids exceeds the size of the NPC. The DNA genome of HBV is derived from reverse transcription of an RNA pregenome. Genome maturation occurs in cytosolic capsids and progeny capsids can deliver the genome into the nucleus causing nuclear genome amplification. The karyophilic capsids are small enough to pass the NPC, but nuclear entry of capsids with an immature genome is halted in the nuclear basket on the nuclear side of the NPC, and the genome remains encapsidated. In contrast, capsids with a mature genome enter the basket and consequently liberate the genome. Investigating the difference between immature and mature capsids, we found that mature capsids had to disintegrate in order to leave the nuclear basket. The arrest of a karyophilic cargo at the nuclear pore is a rare phenomenon, which has been described for only very few cellular proteins participating in nuclear entry. We analyzed the interactions causing HBV capsid retention. By pull-down assays and partial siRNA depletion, we showed that HBV capsids directly interact with nucleoporin 153 (Nup153), an essential protein of the nuclear basket which participates in nuclear transport via importin β. The binding sites of importin β and capsids were shown to overlap but capsid binding was 150-fold stronger. In cellulo experiments using digitonin-permeabilized cells confirmed the interference between capsid binding and nuclear import by importin β. Collectively, our findings describe a unique nuclear import strategy not only for viruses but for all karyophilic cargos. Viral capsids facilitate protection of the enclosed viral genome and participate in the intracellular transport of the genome. At the site of replication capsids have to release the genome. The particular factors triggering genome liberation are not well understood. Like other karyophilic cargos, hepatitis B virus (HBV) capsids are transported through the nuclear pore using nuclear transport receptors of the importin ß superfamily. Unlike physiological cargos, HBV capsids become arrested within the nuclear basket, which is a filamentous structure on the nuclear side of the nuclear pore. Asking which interaction causes this unique strategy, we found that the capsids bind to a protein of the basket periphery, nucleoporin 153 (Nup153). The findings were confirmed in situ using digitonin-permeabilized cells that support physiological genome delivery into the nucleus. We observed that HBV capsids bound to Nup153 irrespective of the maturation of the encapsidated genome. But while capsids with an immature genome remained in arrested state, capsids with a mature genome disassembled and released their DNA.
Collapse
Affiliation(s)
- André Schmitz
- Institute of Medical Virology, Justus Liebig University, Giessen, Germany
| | - Alexandra Schwarz
- Institute of Medical Virology, Justus Liebig University, Giessen, Germany
| | - Michael Foss
- UMR-CNRS 5234 MCMP, Université Bordeaux 2, Bordeaux, France
| | - Lixin Zhou
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Birgit Rabe
- Institute of Medical Virology, Justus Liebig University, Giessen, Germany
| | | | - Miriam Stoeber
- UMR-CNRS 5234 MCMP, Université Bordeaux 2, Bordeaux, France
| | - Nelly Panté
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Kann
- Institute of Medical Virology, Justus Liebig University, Giessen, Germany
- UMR-CNRS 5234 MCMP, Université Bordeaux 2, Bordeaux, France
- * E-mail:
| |
Collapse
|
67
|
Wittkop L, Schwarz A, Cassany A, Grün-Bernhard S, Delaleau M, Rabe B, Cazenave C, Gerlich W, Glebe D, Kann M. Inhibition of protein kinase C phosphorylation of hepatitis B virus capsids inhibits virion formation and causes intracellular capsid accumulation. Cell Microbiol 2010; 12:962-75. [PMID: 20109160 DOI: 10.1111/j.1462-5822.2010.01444.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Capsids of hepatitis B virus and other hepadnaviruses contain a cellular protein kinase, which phosphorylates the capsid protein. Some phosphorylation sites are shown to be essential for distinct steps of viral replication as pregenome packaging or plus strand DNA synthesis. Although different protein kinases have been reported to phosphorylate the capsid protein, varying experimental approaches do not allow direct comparison. Furthermore, the activity of a specific protein kinase has not yet been correlated to steps in the hepadnaviral life cycle. In this study we show that capsids from various sources encapsidate active protein kinase Calpha, irrespective of hepatitis B virus genotype and host cell. Treatment of a virion expressing cell line with a pseudosubstrate inhibitor showed that inhibition of protein kinase C phosphorylation did not affect genome maturation but resulted in capsid accumulation and inhibited virion release to the medium. Our results imply that different protein kinases have distinct functions within the hepadnaviral life cycle.
Collapse
Affiliation(s)
- Linda Wittkop
- Institute of Medical Virology, Justus Liebig University, Frankfurter Strasse 107, D-35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Investigation of the relationship between apolipoprotein E gene polymorphisms and hepatitis B virus infection in northern China. Clin Chem Lab Med 2010; 48:1803-7. [DOI: 10.1515/cclm.2010.354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
69
|
Interaction of hepatitis B virus core protein with human GIPC1. Arch Virol 2009; 155:247-50. [PMID: 20091192 DOI: 10.1007/s00705-009-0561-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 10/29/2009] [Indexed: 12/29/2022]
Abstract
Up to now, little is known about hepatitis B virus core protein (HBc) interactions with host-cell proteins, although such interactions might be essential for virus propagation and pathogenicity. In this work, a human liver cDNA library was screened for proteins interacting with HBc. Among several HBc-interacting partners selected, it interacted most strongly with the human protein GIPC1. A common protein interaction domain, PDZ, was identified as the region that is sufficient for the interaction with HBc. The core protein has a putative C-terminal PDZ-interacting motif, and this sequence proved to be important for the interaction with GIPC1.
Collapse
|
70
|
Abstract
DNA-tumor viruses comprise enveloped and non-enveloped agents that cause malignancies in a large variety of cell types and tissues by interfering with cell cycle control and immortalization. Those DNA-tumor viruses that replicate in the nucleus use cellular mechanisms to transport their genome and newly synthesized viral proteins into the nucleus. This requires cytoplasmic transport and nuclear import of their genome. Agents that employ this strategy include adenoviruses, hepadnaviruses, herpesviruses, and likely also papillomaviruses, and polyomaviruses, but not poxviruses which replicate in the cytoplasm. Here, we discuss how DNA-tumor viruses enter cells, take advantage of cytoplasmic transport, and import their DNA genome through the nuclear pore complex into the nucleus. Remarkably, nuclear import of incoming genomes does not necessarily follow the same pathways used by the structural proteins of the viruses during the replication and assembly phases of the viral life cycle. Understanding the mechanisms of DNA nuclear import can identify new pathways of cell regulation and anti-viral therapies.
Collapse
Affiliation(s)
- Urs F Greber
- Institute of Zoology, University of Zürich, Switzerland
| | | |
Collapse
|
71
|
HBV life cycle: entry and morphogenesis. Viruses 2009; 1:185-209. [PMID: 21994545 PMCID: PMC3185491 DOI: 10.3390/v1020185] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2009] [Revised: 07/31/2009] [Accepted: 08/13/2009] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B virus (HBV) is a major cause of liver disease. HBV primarily infects hepatocytes by a still poorly understood mechanism. After an endocytotic process, the nucleocapsids are released into the cytoplasm and the relaxed circular rcDNA genome is transported towards the nucleus where it is converted into covalently closed circular cccDNA. Replication of the viral genome occurs via an RNA pregenome (pgRNA) that binds to HBV polymerase (P). P initiates pgRNA encapsidation and reverse transcription inside the capsid. Matured, rcDNA containing nucleocapsids can re-deliver the RC-DNA to the nucleus, or be secreted via interaction with the envelope proteins as progeny virions.
Collapse
|
72
|
Rabe B, Delaleau M, Bischof A, Foss M, Sominskaya I, Pumpens P, Cazenave C, Castroviejo M, Kann M. Nuclear entry of hepatitis B virus capsids involves disintegration to protein dimers followed by nuclear reassociation to capsids. PLoS Pathog 2009; 5:e1000563. [PMID: 19714236 PMCID: PMC2727048 DOI: 10.1371/journal.ppat.1000563] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 08/03/2009] [Indexed: 12/21/2022] Open
Abstract
Assembly and disassembly of viral capsids are essential steps in the viral life cycle. Studies on their kinetics are mostly performed in vitro, allowing application of biochemical, biophysical and visualizing techniques. In vivo kinetics are poorly understood and the transferability of the in vitro models to the cellular environment remains speculative. We analyzed capsid disassembly of the hepatitis B virus in digitonin-permeabilized cells which support nuclear capsid entry and subsequent genome release. Using gradient centrifugation, size exclusion chromatography and immune fluorescence microscopy of digitonin-permeabilized cells, we showed that capsids open and close reversibly. In the absence of RNA, capsid re-assembly slows down; the capsids remain disintegrated and enter the nucleus as protein dimers or irregular polymers. Upon the presence of cellular RNA, capsids re-assemble in the nucleus. We conclude that reversible genome release from hepatitis B virus capsids is a unique strategy different from that of other viruses, which employs irreversible capsid destruction for genome release. The results allowed us to propose a model of HBV genome release in which the unique environment of the nuclear pore favors HBV capsid disassembly reaction, while both cytoplasm and nucleus favor capsid assembly. Viral capsids facilitate protection of the enclosed viral genome and participate in the intracellular transport of the genome. At the site of replication capsids have to release the genome, but after replication new capsids have to be assembled for encapsidation of the progeny genomes. Detailed data on stability of capsids and kinetics of their formation and dissociation are obtained for several viruses in vitro, allowing biophysical or electron microscopical techniques. These approaches, however, do not consider the impact of cellular interaction partners. Using digitonin-permeabilized cells which support hepadnaviral genome release actively, we analyzed the disassembly kinetic of the hepatitis B virus (HBV) capsid. Using different analytical methods we found that HBV capsids disintegrate to protein dimers which reassemble to capsids inside the nucleus. The study provides a link between in vitro and in vivo data showing that HBV uses a unique strategy. We propose a model in which the unique environment of the nuclear pore favors the disassembly reaction, while both cytoplasm and nucleus favor assembly.
Collapse
Affiliation(s)
- Birgit Rabe
- Institute of Virology, Justus Liebig University, Giessen, Germany
| | | | - Andreas Bischof
- Institute of Virology, Justus Liebig University, Giessen, Germany
| | - Michael Foss
- UMR 5234 CNRS-University Bordeaux 2 MCMP, Bordeaux, France
| | | | - Paul Pumpens
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | | - Michael Kann
- Institute of Virology, Justus Liebig University, Giessen, Germany
- UMR 5234 CNRS-University Bordeaux 2 MCMP, Bordeaux, France
- * E-mail:
| |
Collapse
|
73
|
Rizzetto M, Zoulim F. Viral Hepatitis. TEXTBOOK OF HEPATOLOGY 2007:819-956. [DOI: 10.1002/9780470691861.ch9a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
74
|
Li SH, Huang WG, Huang B, Chen XG. High expression of hepatitis B virus based vector with reporter gene in hepatitis B virus infection system. World J Gastroenterol 2007; 13:2490-5. [PMID: 17552034 PMCID: PMC4146769 DOI: 10.3748/wjg.v13.i17.2490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct a hepatitis B virus (HBV)-based vector with a reporter gene and to establish an HBV infection system to evaluate the availability of the vector.
METHODS: The HBV-based vectors with green fluorescence protein (GFP) were packaged into the liver of immunodeficient mice through transfer and helper plasmid using hydrodynamic technology. Wild type HBV (wt HBV) was provided by plasmid MC2009. Primary human hepatocytes (PHH) were isolated and infected with recombinant HBV (rHBV) or wt HBV. GFP expression was monitored by confocal and flow cytometry. HBV DNA and HBV surface antigen (HBSAg) were analyzed by PCR and ELISA.
RESULTS: 3 × 107 wt HBV copies/mL and 5 × 106 rHBV copies/mL were collected from mice serum. In the wt HBV infected group, HBV progeny was 2 × 107 copies/mL and HBSAg was 770 ng/mL. In the rHBV infected group, GFP fluorescence was detected on d 3 post-infection and over 85% of the parenchymal cells expressed green fluorescence on d 12 post-infection. Compared with wt HBV in the PHH infection system, no rHBV DNA or HBSAg were detected in PHH culture media.
CONCLUSION: An effective HBV based vector was developed, which proved to be a useful HBV infection system. This vector and infection system can be applied to develop a therapeutic vector and study the HBV life cycle and viral pathogenesis.
Collapse
Affiliation(s)
- Shi-Hong Li
- Center of Experimental Animals, Sun Yat-sen University, 58 Zhongshan 2 Road, Guangzhou 510080, Guangdong Province, China
| | | | | | | |
Collapse
|
75
|
Kann M, Schmitz A, Rabe B. Intracellular transport of hepatitis B virus. World J Gastroenterol 2007; 13:39-47. [PMID: 17206753 PMCID: PMC4065875 DOI: 10.3748/wjg.v13.i1.39] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 09/25/2006] [Accepted: 11/21/2006] [Indexed: 02/06/2023] Open
Abstract
For genome multiplication hepadnaviruses use the transcriptional machinery of the cell that is found within the nucleus. Thus the viral genome has to be transported through the cytoplasm and nuclear pore. The intracytosolic translocation is facilitated by the viral capsid that surrounds the genome and that interacts with cellular microtubules. The subsequent passage through the nuclear pore complexes (NPC) is mediated by the nuclear transport receptors importin alpha and beta. Importin alpha binds to the C-terminus of the capsid protein that comprises a nuclear localization signal (NLS). The exposure of the NLS is regulated and depends upon genome maturation and/or phosphorylation of the capsid protein. As for other karyophilic cargos using this pathway importin alpha interacts with importin beta that facilitates docking of the import complex to the NPC and the passage through the pore. Being a unique strategy, the import of the viral capsid is incomplete in that it becomes arrested inside the nuclear basket, which is a cage-like structure on the karyoplasmic face of the NPC. Presumably only this compartment provides the factors that are required for capsid disassembly and genome release that is restricted to those capsids comprising a mature viral DNA genome.
Collapse
Affiliation(s)
- Michael Kann
- CNRS-REGER (UMR 5097), Bâtiment 3A, Université Bordeaux 2, 146 rue Leo Saignat, F-33076 Bordeaux Cedex, France.
| | | | | |
Collapse
|
76
|
Abstract
Hepadnaviridae is a family of hepatotropic DNA viruses that is divided into the genera orthohepadnavirus of mammals and avihepadnavirus of birds. All members of this family can cause acute and chronic hepatic infection, which in the case of human hepatitis B virus (HBV) constitutes a major global health problem. Although our knowledge about the molecular biology of these highly liver-specific viruses has profoundly increased in the last two decades, the mechanisms of attachment and productive entrance into the differentiated host hepatocytes are still enigmatic. The difficulties in studying hepadnaviral entry were primarily caused by the lack of easily accessible in vitro infection systems. Thus, for more than twenty years, differentiated primary hepatocytes from the respective species were the only in vitro models for both orthohepadnaviruses (e.g. HBV) and avihepadnaviruses (e.g. duck hepatitis B virus [DHBV]). Two important discoveries have been made recently regarding HBV: (1) primary hepatocytes from tree-shrews; i.e., Tupaia belangeri, can be substituted for primary human hepatocytes, and (2) a human hepatoma cell line (HepaRG) was established that gains susceptibility for HBV infection upon induction of differentiation in vitro. A number of potential HBV receptor candidates have been described in the past, but none of them have been confirmed to function as a receptor. For DHBV and probably all other avian hepadnaviruses, carboxypeptidase D (CPD) has been shown to be indispensable for infection, although the exact role of this molecule is still under debate. While still restricted to the use of primary duck hepatocytes (PDH), investigations performed with DHBV provided important general concepts on the first steps of hepadnaviral infection. However, with emerging data obtained from the new HBV infection systems, the hope that DHBV utilizes the same mechanism as HBV only partially held true. Nevertheless, both HBV and DHBV in vitro infection systems will help to: (1) functionally dissect the hepadnaviral entry pathways, (2) perform reverse genetics (e.g. test the fitness of escape mutants), (3) titrate and map neutralizing antibodies, (4) improve current vaccines to combat acute and chronic infections of hepatitis B, and (5) develop entry inhibitors for future clinical applications.
Collapse
Affiliation(s)
- Dieter Glebe
- Institute of Medical Virology, Justus-Liebig University of Giessen, Frankfurter Strasse 107, D-35392 Giessen, Germany.
| | | |
Collapse
|