1
|
Lozada C, Barlow TMA, Gonzalez S, Lubin-Germain N, Ballet S. Identification and Characteristics of Fusion Peptides Derived From Enveloped Viruses. Front Chem 2021; 9:689006. [PMID: 34497798 PMCID: PMC8419435 DOI: 10.3389/fchem.2021.689006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/10/2021] [Indexed: 01/28/2023] Open
Abstract
Membrane fusion events allow enveloped viruses to enter and infect cells. The study of these processes has led to the identification of a number of proteins that mediate this process. These proteins are classified according to their structure, which vary according to the viral genealogy. To date, three classes of fusion proteins have been defined, but current evidence points to the existence of additional classes. Despite their structural differences, viral fusion processes follow a common mechanism through which they exert their actions. Additional studies of the viral fusion proteins have demonstrated the key role of specific proteinogenic subsequences within these proteins, termed fusion peptides. Such peptides are able to interact and insert into membranes for which they hold interest from a pharmacological or therapeutic viewpoint. Here, the different characteristics of fusion peptides derived from viral fusion proteins are described. These criteria are useful to identify new fusion peptides. Moreover, this review describes the requirements of synthetic fusion peptides derived from fusion proteins to induce fusion by themselves. Several sequences of the viral glycoproteins E1 and E2 of HCV were, for example, identified to be able to induce fusion, which are reviewed here.
Collapse
Affiliation(s)
- Camille Lozada
- BioCIS, CNRS, CY Cergy-Paris Université, Cergy-Pontoise, France
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas M. A. Barlow
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| | - Simon Gonzalez
- BioCIS, CNRS, CY Cergy-Paris Université, Cergy-Pontoise, France
| | | | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
2
|
Hu Z, Rolt A, Hu X, Ma CD, Le DJ, Park SB, Houghton M, Southall N, Anderson DE, Talley DC, Lloyd JR, Marugan JC, Liang TJ. Chlorcyclizine Inhibits Viral Fusion of Hepatitis C Virus Entry by Directly Targeting HCV Envelope Glycoprotein 1. Cell Chem Biol 2020; 27:780-792.e5. [PMID: 32386595 PMCID: PMC7368827 DOI: 10.1016/j.chembiol.2020.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/04/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022]
Abstract
Chlorcyclizine (CCZ) is a potent hepatitis C virus (HCV) entry inhibitor, but its molecular mechanism is unknown. Here, we show that CCZ directly targets the fusion peptide of HCV E1 and interferes with the fusion process. Generation of CCZ resistance-associated substitutions of HCV in vitro revealed six missense mutations in the HCV E1 protein, five being in the putative fusion peptide. A viral fusion assay demonstrated that CCZ blocked HCV entry at the membrane fusion step and that the mutant viruses acquired resistance to CCZ's action in blocking membrane fusion. UV cross-linking of photoactivatable CCZ-diazirine-biotin in both HCV-infected cells and recombinant HCV E1/E2 protein demonstrated direct binding to HCV E1 glycoprotein. Mass spectrometry analysis revealed that CCZ cross-linked to an E1 sequence adjacent to the putative fusion peptide. Docking simulations demonstrate a putative binding model, wherein CCZ binds to a hydrophobic pocket of HCV E1 and forms extensive interactions with the fusion peptide.
Collapse
Affiliation(s)
- Zongyi Hu
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Adam Rolt
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Xin Hu
- Division of Pre-Clinical Innovations, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Christopher D Ma
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Derek J Le
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Seung Bum Park
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Michael Houghton
- Li Ka Shing Virology Institute, University of Alberta, Edmonton, Canada
| | - Noel Southall
- Division of Pre-Clinical Innovations, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - D Eric Anderson
- Advanced Mass Spectrometry Facility, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Daniel C Talley
- Division of Pre-Clinical Innovations, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - John R Lloyd
- Advanced Mass Spectrometry Facility, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Juan C Marugan
- Division of Pre-Clinical Innovations, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - T Jake Liang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Abstract
The membrane fusion properties of HCV envelope glycoproteins can be evaluated using several assays. Fusion assays generally require contacts between glycoproteins expressed on a donor membrane, such as those from a cell or a viral particle, and an acceptor membrane that may or may not express cognate viral receptor, such as those from an indicator cell or a liposome. In this chapter, we describe three well-established methods in the field that use either cell surface expression of glycoproteins, HCV pseudoparticles (HCVpp), or cell culture-grown HCV (HCVcc) particles for donor membrane and cells or liposomes as acceptor membrane in which specific components can be included to monitor and quantify fusion. We provide details of cell-cell fusion assay, virus-liposome fusion assay, and finally virus-plasma membrane fusion assay. We also describe inhibitors that can block HCV envelope membrane fusion.
Collapse
|
4
|
Gonzalez S, Gallier F, Kellouche S, Carreiras F, Novellino E, Carotenuto A, Chassaing G, Rovero P, Uziel J, Lubin-Germain N. Studies of membranotropic and fusogenic activity of two putative HCV fusion peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:50-61. [PMID: 30343120 DOI: 10.1016/j.bbamem.2018.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 11/19/2022]
Abstract
Over the past decades, membranotropic peptides such as positively charged cell-penetrating peptides (CPPs) or amphipathic antimicrobial peptides (AMPs) have received increasing interest in order to improve therapeutic agent cellular uptake. As far as we are concerned, we were interested in studying HCV fusion peptides as putative anchors. Two peptides, HCV6 and HCV7, were identified and conjugated to a fluorescent tag NBD and tested for their interaction with liposomes as model membranes. DSC and spectrofluorescence analyses demonstrate HCV7 propensity to insert or internalize in vesicles containing anionic lipids DMPG whereas no activity was observed with zwitterionic DMPC. This behavior could be explained by the peptide sequence containing a cationic arginine residue. On the contrary, HCV6 did not exhibit any membranotropic activity but was the only sequence able to induce liposomes' fusion or aggregation monitored by spectrofluorescence and DLS. This two peptides mild activity was related to their inefficient structuration in contact with membrane mimetics, which was demonstrated by CD and NMR experiments. Altogether, our data allowed us to identify two promising membrane-active peptides from E1 and E2 HCV viral proteins, one fusogenic (HCV6) and the other membranotropic (HCV7). The latter was also confirmed by fluorescence microscopy with CHO cells, indicating that HCV7 could cross the plasma membrane via an endocytosis process. Therefore, this study provides new evidences supporting the identification of HCV6 as the HCV fusion peptide as well as insights on a novel membranotropic peptide from the HCV-E2 viral protein.
Collapse
Affiliation(s)
- Simon Gonzalez
- Laboratoire de Chimie Biologique, University of Cergy-Pontoise, 5 mail Gay-Lussac, Cergy-Pontoise, France
| | - Florian Gallier
- Laboratoire de Chimie Biologique, University of Cergy-Pontoise, 5 mail Gay-Lussac, Cergy-Pontoise, France
| | - Sabrina Kellouche
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Institut des Matériaux, I-MAT (FD4122), University of Cergy-Pontoise, MIR, rue Descartes, 95031, Neuville sur Oise Cedex, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Institut des Matériaux, I-MAT (FD4122), University of Cergy-Pontoise, MIR, rue Descartes, 95031, Neuville sur Oise Cedex, France
| | - Ettore Novellino
- Department of Pharmacy, University of Naples 'Federico II', Naples 80131, Italy
| | - Alfonso Carotenuto
- Department of Pharmacy, University of Naples 'Federico II', Naples 80131, Italy
| | - Gérard Chassaing
- Sorbonne Universités, UPMC University Paris 06, LBM, 4 place Jussieu, F-75005 Paris, France
| | - Paolo Rovero
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Jacques Uziel
- Laboratoire de Chimie Biologique, University of Cergy-Pontoise, 5 mail Gay-Lussac, Cergy-Pontoise, France
| | - Nadège Lubin-Germain
- Laboratoire de Chimie Biologique, University of Cergy-Pontoise, 5 mail Gay-Lussac, Cergy-Pontoise, France.
| |
Collapse
|
5
|
Uddin R, Downard KM. Molecular basis of benzimidazole inhibitors to hepatitis C virus envelope glycoprotein. Chem Biol Drug Des 2018; 92:1638-1646. [DOI: 10.1111/cbdd.13329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/18/2018] [Accepted: 04/21/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Reaz Uddin
- Infectious Disease Responses Laboratory; University of New South Wales; Sydney NSW Australia
| | - Kevin M. Downard
- Infectious Disease Responses Laboratory; University of New South Wales; Sydney NSW Australia
| |
Collapse
|
6
|
Fusogenic properties of the Ectodomain of HCV E2 envelope protein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:728-736. [DOI: 10.1016/j.bbamem.2017.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 01/04/2023]
|
7
|
Gopal R, Jackson K, Tzarum N, Kong L, Ettenger A, Guest J, Pfaff JM, Barnes T, Honda A, Giang E, Davidson E, Wilson IA, Doranz BJ, Law M. Probing the antigenicity of hepatitis C virus envelope glycoprotein complex by high-throughput mutagenesis. PLoS Pathog 2017; 13:e1006735. [PMID: 29253863 PMCID: PMC5749897 DOI: 10.1371/journal.ppat.1006735] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/02/2018] [Accepted: 11/04/2017] [Indexed: 12/12/2022] Open
Abstract
The hepatitis C virus (HCV) envelope glycoproteins E1 and E2 form a non-covalently linked heterodimer on the viral surface that mediates viral entry. E1, E2 and the heterodimer complex E1E2 are candidate vaccine antigens, but are technically challenging to study because of difficulties in producing natively folded proteins by standard protein expression and purification methods. To better comprehend the antigenicity of these proteins, a library of alanine scanning mutants comprising the entirety of E1E2 (555 residues) was created for evaluating the role of each residue in the glycoproteins. The mutant library was probed, by a high-throughput flow cytometry-based assay, for binding with the co-receptor CD81, and a panel of 13 human and mouse monoclonal antibodies (mAbs) that target continuous and discontinuous epitopes of E1, E2, and the E1E2 complex. Together with the recently determined crystal structure of E2 core domain (E2c), we found that several residues in the E2 back layer region indirectly impact binding of CD81 and mAbs that target the conserved neutralizing face of E2. These findings highlight an unexpected role for the E2 back layer in interacting with the E2 front layer for its biological function. We also identified regions of E1 and E2 that likely located at or near the interface of the E1E2 complex, and determined that the E2 back layer also plays an important role in E1E2 complex formation. The conformation-dependent reactivity of CD81 and the antibody panel to the E1E2 mutant library provides a global view of the influence of each amino acid (aa) on E1E2 expression and folding. This information is valuable for guiding protein engineering efforts to enhance the antigenic properties and stability of E1E2 for vaccine antigen development and structural studies.
Collapse
Affiliation(s)
- Radhika Gopal
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Kelli Jackson
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Netanel Tzarum
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Leopold Kong
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Andrew Ettenger
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | - Johnathan Guest
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | - Jennifer M. Pfaff
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | - Trevor Barnes
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | - Andrew Honda
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Erick Giang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Edgar Davidson
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States of America
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, United States of America
| | | | - Mansun Law
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States of America
- * E-mail:
| |
Collapse
|
8
|
Naik AS, Owsianka A, Palmer BA, O’Halloran CJ, Walsh N, Crosbie O, Kenny-Walsh E, Patel AH, Fanning LJ. Reverse epitope mapping of the E2 glycoprotein in antibody associated hepatitis C virus. PLoS One 2017; 12:e0175349. [PMID: 28558001 PMCID: PMC5448734 DOI: 10.1371/journal.pone.0175349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/24/2017] [Indexed: 12/20/2022] Open
Abstract
The humoral immune system responds to chronic hepatitis C virus (HCV) infection by producing neutralising antibodies (nAb). In this study we generated three HCV pseudoparticles in which E1E2 glycoprotein sequence was targeted by the host humoral immune system. We used patient derived virus free Fabs (VF-Fabs) obtained from HCV genotype 1a (n = 3), genotype 1b (n = 7) and genotype 3a (n = 1) for neutralisation of HCVpp produced in this study both individually and in combination. Based on the available anti-HCV monoclonal nAb mapping information we selected amino acid region 384-619 for conformational epitope mapping. Amongst our notable findings, we observed significant reduction in HCVpp infectivity (p<0.05) when challenged with a combination of inter genotype and subtype VF-Fabs. We also identified five binding motifs targeted by patient derived VF-Fab upon peptide mapping, of which two shared the residues with previously reported epitopes. One epitope lies within an immunodominant HVR1 and two were novel. In summary, we used a reverse epitope mapping strategy to identify preferred epitopes by the host humoral immune system. Additionally, we have combined different VF-Fabs to further reduce the HCVpp infectivity. Our data indicates that combining the antigen specificity of antibodies may be a useful strategy to reduce (in-vitro) infectivity.
Collapse
Affiliation(s)
- Amruta S. Naik
- Department of Medicine, University College Cork, Cork, Ireland
| | - Ania Owsianka
- MRC—University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | | | | | - Nicole Walsh
- Department of Medicine, University College Cork, Cork, Ireland
| | - Orla Crosbie
- Department of Hepatology, Cork University Hospital, Cork, Ireland
| | | | - Arvind H. Patel
- MRC—University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - Liam J. Fanning
- Department of Medicine, University College Cork, Cork, Ireland
- APC-Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
9
|
Computational Prediction of the Heterodimeric and Higher-Order Structure of gpE1/gpE2 Envelope Glycoproteins Encoded by Hepatitis C Virus. J Virol 2017; 91:JVI.02309-16. [PMID: 28148799 DOI: 10.1128/jvi.02309-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/25/2017] [Indexed: 12/24/2022] Open
Abstract
Despite the recent success of newly developed direct-acting antivirals against hepatitis C, the disease continues to be a global health threat due to the lack of diagnosis of most carriers and the high cost of treatment. The heterodimer formed by glycoproteins E1 and E2 within the hepatitis C virus (HCV) lipid envelope is a potential vaccine candidate and antiviral target. While the structure of E1/E2 has not yet been resolved, partial crystal structures of the E1 and E2 ectodomains have been determined. The unresolved parts of the structure are within the realm of what can be modeled with current computational modeling tools. Furthermore, a variety of additional experimental data is available to support computational predictions of E1/E2 structure, such as data from antibody binding studies, cryo-electron microscopy (cryo-EM), mutational analyses, peptide binding analysis, linker-scanning mutagenesis, and nuclear magnetic resonance (NMR) studies. In accordance with these rich experimental data, we have built an in silico model of the full-length E1/E2 heterodimer. Our model supports that E1/E2 assembles into a trimer, which was previously suggested from a study by Falson and coworkers (P. Falson, B. Bartosch, K. Alsaleh, B. A. Tews, A. Loquet, Y. Ciczora, L. Riva, C. Montigny, C. Montpellier, G. Duverlie, E. I. Pecheur, M. le Maire, F. L. Cosset, J. Dubuisson, and F. Penin, J. Virol. 89:10333-10346, 2015, https://doi.org/10.1128/JVI.00991-15). Size exclusion chromatography and Western blotting data obtained by using purified recombinant E1/E2 support our hypothesis. Our model suggests that during virus assembly, the trimer of E1/E2 may be further assembled into a pentamer, with 12 pentamers comprising a single HCV virion. We anticipate that this new model will provide a useful framework for HCV envelope structure and the development of antiviral strategies.IMPORTANCE One hundred fifty million people have been estimated to be infected with hepatitis C virus, and many more are at risk for infection. A better understanding of the structure of the HCV envelope, which is responsible for attachment and fusion, could aid in the development of a vaccine and/or new treatments for this disease. We draw upon computational techniques to predict a full-length model of the E1/E2 heterodimer based on the partial crystal structures of the envelope glycoproteins E1 and E2. E1/E2 has been widely studied experimentally, and this provides valuable data, which has assisted us in our modeling. Our proposed structure is used to suggest the organization of the HCV envelope. We also present new experimental data from size exclusion chromatography that support our computational prediction of a trimeric oligomeric state of E1/E2.
Collapse
|
10
|
Freedman H, Logan MR, Law JLM, Houghton M. Structure and Function of the Hepatitis C Virus Envelope Glycoproteins E1 and E2: Antiviral and Vaccine Targets. ACS Infect Dis 2016; 2:749-762. [PMID: 27933781 DOI: 10.1021/acsinfecdis.6b00110] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The hepatitis C virus (HCV) envelope glycoproteins E1 and E2 are critical in viral attachment and cell fusion, and studies of these proteins may provide valuable insights into their potential uses in vaccines and antiviral strategies. Progress has included elucidating the crystal structures of portions of their ectodomains, as well as many other studies of hypervariable regions, stem regions, glycosylation sites, and the participation of E1/E2 in viral fusion with the endosomal membrane. The available structural data have shed light on the binding sites of cross-neutralizing antibodies. A large amount of information has been discovered concerning heterodimerization, including the roles of transmembrane domains, disulfide bonding, and heptad repeat regions. The possible organization of higher order oligomers within the HCV virion has also been evaluated on the basis of experimental data. In this review, E1/E2 structure and function is discussed, and some important issues requiring further study are highlighted.
Collapse
Affiliation(s)
- Holly Freedman
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael R. Logan
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - John Lok Man Law
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Houghton
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Lombana L, Ortega-Atienza S, Gómez-Gutiérrez J, Yélamos B, Peterson DL, Gavilanes F. The deletion of residues 268-292 of E1 impairs the ability of HCV envelope proteins to induce pore formation. Virus Res 2016; 217:63-70. [PMID: 26945847 DOI: 10.1016/j.virusres.2016.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 12/25/2022]
Abstract
We have obtained a chimeric protein containing the ectodomains of hepatitis C virus (HCV) envelope proteins but lacking the region 268-292 of E1. All its structural properties are coincident with those of the corresponding full length chimera. The deleted and entire chimeras were compared in terms of their membrane destabilizing properties. No differences were found in their ability to induce vesicle aggregation and lipid mixing but the deleted chimera showed a reduced capacity to promote leakage. The role of the deletion was also studied by obtaining HCV pseudoparticles (HCVpp). Both E1 and E2, and also the E1 deleted mutant, were incorporated into HCVpp to a similar level. However, HCVpp containing the E1 deleted protein are almost unable to infect Huh7 cells. These results point to the involvement of the region 268-292 in the formation of pores in the membrane necessary for the complete fusion of the membranes.
Collapse
Affiliation(s)
- Laura Lombana
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sara Ortega-Atienza
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Julián Gómez-Gutiérrez
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Belén Yélamos
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Darrell L Peterson
- Department of Biochemistry and Molecular Biology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Francisco Gavilanes
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| |
Collapse
|
12
|
Alves NS, Mendes YS, Souza TLF, Bianconi ML, Silva JL, Gomes AMO, Oliveira AC. A biophysical characterization of the interaction of a hepatitis C virus membranotropic peptide with micelles. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:359-71. [PMID: 26773352 DOI: 10.1016/j.bbapap.2016.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 01/02/2016] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
Abstract
Membrane fusion is a highly regulated process that allows enveloped viruses to enter cells and replicate. Viral glycoproteins trigger membrane fusion by means of internal sequences known as fusion peptides. The hepatitis C virus (HCV) genome encodes the envelope glycoproteins E1 and E2, but their specific roles in the fusion step and the localization of the fusion peptide remain uncharacterized. Here, we studied the biophysics of the interactions between the glycoprotein E2 peptide HCV421-445 and four different micellar systems providing ionic, non-ionic and zwitterionic surfaces to investigate the importance of electrostatic interactions for peptide-membrane binding. Circular dichroism, fluorescence spectroscopy and calorimetry were used to characterize peptide-micelle interactions and structural changes. Fluorescence quenching showed that HCV421-445 interacts with SDS or CTAB ionic, n-OGP non-ionic and DPC zwitterionic micelles. The indole ring of Trp seems to anchor the peptide in micelles. Trp residues seem to be more deeply inserted in ionic and non-ionic micelles where peptide interactions are more stable than with DPC zwitterionic micelles. The interaction with zwitterionic micelles appears to occur at the surface. Both interaction types are exothermic because of peptide-micelle interactions and a gain of secondary structure in the helical conformation. HCV421-445 interacts with detergent monomers and micelles. Peptide-micelle interaction is pH-independent. HCV421-445 interacts with membranes, promoting aggregation and coalescence of vesicles with content leakage, suggesting that HCV421-445 may participate in membrane fusion. This structural characterization contributes to our understanding of the molecular process that promotes fusion, which is important in the further development of new antiviral therapies.
Collapse
Affiliation(s)
- N S Alves
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Y S Mendes
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - T L F Souza
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - M L Bianconi
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - J L Silva
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - A M O Gomes
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - A C Oliveira
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| |
Collapse
|
13
|
Perin PM, Haid S, Brown RJ, Doerrbecker J, Schulze K, Zeilinger C, von Schaewen M, Heller B, Vercauteren K, Luxenburger E, Baktash YM, Vondran FW, Speerstra S, Awadh A, Mukhtarov F, Schang LM, Kirschning A, Müller R, Guzman CA, Kaderali L, Randall G, Meuleman P, Ploss A, Pietschmann T. Flunarizine prevents hepatitis C virus membrane fusion in a genotype-dependent manner by targeting the potential fusion peptide within E1. Hepatology 2016; 63:49-62. [PMID: 26248546 PMCID: PMC4688136 DOI: 10.1002/hep.28111] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/05/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED To explore mechanisms of hepatitis C viral (HCV) replication we screened a compound library including licensed drugs. Flunarizine, a diphenylmethylpiperazine used to treat migraine, inhibited HCV cell entry in vitro and in vivo in a genotype-dependent fashion. Analysis of mosaic viruses between susceptible and resistant strains revealed that E1 and E2 glycoproteins confer susceptibility to flunarizine. Time of addition experiments and single particle tracking of HCV demonstrated that flunarizine specifically prevents membrane fusion. Related phenothiazines and pimozide also inhibited HCV infection and preferentially targeted HCV genotype 2 viruses. However, phenothiazines and pimozide exhibited improved genotype coverage including the difficult to treat genotype 3. Flunarizine-resistant HCV carried mutations within the alleged fusion peptide and displayed cross-resistance to these compounds, indicating that these drugs have a common mode of action. CONCLUSION These observations reveal novel details about HCV membrane fusion; moreover, flunarizine and related compounds represent first-in-class HCV fusion inhibitors that merit consideration for repurposing as a cost-effective component of HCV combination therapies.
Collapse
Affiliation(s)
- Paula M. Perin
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection ResearchHannoverGermany
| | - Sibylle Haid
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection ResearchHannoverGermany
| | - Richard J.P. Brown
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection ResearchHannoverGermany
| | - Juliane Doerrbecker
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection ResearchHannoverGermany
| | - Kai Schulze
- Department of Vaccinology and Applied MicrobiologyHelmholtz Centre of Infection ResearchBraunschweigGermany
| | - Carsten Zeilinger
- Institute of Organic Chemistry and Center of Biomolecular Drug Research, Leibniz UniversitätHannoverGermany
| | | | - Brigitte Heller
- Department of Molecular BiologyPrinceton UniversityPrincetonNJ
| | | | - Eva Luxenburger
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
| | | | - Florian W.R. Vondran
- ReMediES, Department of GeneralVisceral and Transplantation Surgery, Hannover Medical SchoolHannoverGermany,German Centre for Infection Research, Hannover‐BraunschweigGermany
| | - Sietkse Speerstra
- Department of BiochemistryUniversity of AlbertaEdmontonABCanada,Li Ka Shing Institute of Virology, University of AlbertaEdmontonABCanada
| | - Abdullah Awadh
- Li Ka Shing Institute of Virology, University of AlbertaEdmontonABCanada,Department of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
| | - Furkat Mukhtarov
- Department of BiochemistryUniversity of AlbertaEdmontonABCanada,Li Ka Shing Institute of Virology, University of AlbertaEdmontonABCanada
| | - Luis M. Schang
- Department of BiochemistryUniversity of AlbertaEdmontonABCanada,Li Ka Shing Institute of Virology, University of AlbertaEdmontonABCanada,Department of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
| | - Andreas Kirschning
- Institute of Organic Chemistry and Center of Biomolecular Drug Research, Leibniz UniversitätHannoverGermany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
| | - Carlos A. Guzman
- Department of Vaccinology and Applied MicrobiologyHelmholtz Centre of Infection ResearchBraunschweigGermany
| | | | - Glenn Randall
- Department of MicrobiologyThe University of ChicagoChicagoIL
| | | | - Alexander Ploss
- Department of Molecular BiologyPrinceton UniversityPrincetonNJ
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection ResearchHannoverGermany,German Centre for Infection Research, Hannover‐BraunschweigGermany
| |
Collapse
|
14
|
Kong L, Jackson KN, Wilson IA, Law M. Capitalizing on knowledge of hepatitis C virus neutralizing epitopes for rational vaccine design. Curr Opin Virol 2015; 11:148-57. [PMID: 25932568 PMCID: PMC4507806 DOI: 10.1016/j.coviro.2015.04.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/08/2015] [Indexed: 12/13/2022]
Abstract
Hepatitis C virus infects nearly 3% of the world's population and is often referred as a silent epidemic. It is a leading cause of liver cirrhosis and hepatocellular carcinoma in endemic countries. Although antiviral drugs are now available, they are not readily accessible to marginalized social groups and developing nations that are disproportionally impacted by HCV. To stop the HCV pandemic, a vaccine is needed. Recent advances in HCV research have provided new opportunities for studying HCV neutralizing antibodies and their subsequent use for rational vaccine design. It is now recognized that neutralizing antibodies to conserved antigenic sites of the virus can cross-neutralize diverse HCV genotypes and protect against infection in vivo. Structural characterization of the neutralizing epitopes has provided valuable information for design of candidate immunogens.
Collapse
Affiliation(s)
- Leopold Kong
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kelli N Jackson
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mansun Law
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
15
|
Galdiero S, Falanga A, Morelli G, Galdiero M. gH625: a milestone in understanding the many roles of membranotropic peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:16-25. [PMID: 25305339 PMCID: PMC7124228 DOI: 10.1016/j.bbamem.2014.10.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/26/2014] [Accepted: 10/01/2014] [Indexed: 12/05/2022]
Abstract
Here, we review the current knowledge about viral derived membranotropic peptides, and we discuss how they may be used for many therapeutic applications. While they have been initially discovered in viral fusion proteins and have been involved in the mechanism of viral entry, it is now clear that their features and their mode of interaction with membrane bilayers can be exploited to design viral inhibitors as well as to favor delivery of cargos across the cell membrane and across the blood–brain barrier. The peptide gH625 has been extensively used for all these purposes and provides a significant contribution to the field. We describe the roles of this sequence in order to close the gap between the many functions that are now emerging for membranotropic peptides. Membranotropic peptides and their therapeutic applications Membrane fusion, viral inhibition, drug delivery gH625, a peptide derived from Herpes simplex virus type I: a case study gH625 in vitro and in vivo delivery across the blood–brain barrier
Collapse
Affiliation(s)
- Stefania Galdiero
- Department of Pharmacy, University of Naples "Federico II", Via Mezzocannone 16, 80134 Naples, Italy; DFM Scarl, University of Naples "Federico II", Via Mezzocannone 16, 80134 Naples, Italy.
| | - Annarita Falanga
- Department of Pharmacy, University of Naples "Federico II", Via Mezzocannone 16, 80134 Naples, Italy; DFM Scarl, University of Naples "Federico II", Via Mezzocannone 16, 80134 Naples, Italy
| | - Giancarlo Morelli
- Department of Pharmacy, University of Naples "Federico II", Via Mezzocannone 16, 80134 Naples, Italy; DFM Scarl, University of Naples "Federico II", Via Mezzocannone 16, 80134 Naples, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, II University of Naples, Via De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
16
|
Unexpected structure for the N-terminal domain of hepatitis C virus envelope glycoprotein E1. Nat Commun 2014; 5:4874. [PMID: 25224686 PMCID: PMC4175578 DOI: 10.1038/ncomms5874] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/31/2014] [Indexed: 12/21/2022] Open
Abstract
Hepatitis C virus (HCV) infection remains a major health problem worldwide. HCV entry into host cells and membrane fusion are achieved by two envelope glycoproteins, E1 and E2. We report here the 3.5-Å resolution crystal structure of the N-terminal domain of the HCV E1 ectodomain, which reveals a complex network of covalently linked intertwined homodimers that do not harbour the expected truncated class II fusion protein fold. Hepatitis C virus (HCV) gains entry into host cells via envelope glycoproteins E1 and E2. Here, El Omari et al. present the crystal structure of the N terminus of the E1 ectodomain of HCV and show that it adopts a different fold than predicted.
Collapse
|
17
|
Abstract
![]()
The Dengue virus (DENV) NS2A protein,
essential for viral replication,
is a poorly characterized membrane protein. NS2A displays both protein/protein
and membrane/protein interactions, yet neither its functions in the
viral cycle nor its active regions are known with certainty. To highlight
the different membrane-active regions of NS2A, we characterized the
effects of peptides derived from a peptide library encompassing this
protein’s full length on different membranes by measuring their
membrane leakage induction and modulation of lipid phase behavior.
Following this initial screening, one region, peptide dens25, had
interesting effects on membranes; therefore, we sought to thoroughly
characterize this region’s interaction with membranes. This
peptide presents an interfacial/hydrophobic pattern characteristic
of a membrane-proximal segment. We show that dens25 strongly interacts
with membranes that contain a large proportion of lipid molecules
with a formal negative charge, and that this effect has a major electrostatic
contribution. Considering its membrane modulating capabilities, this
region might be involved in membrane rearrangements and thus be important
for the viral cycle.
Collapse
Affiliation(s)
- Henrique Nemésio
- Molecular and Cellular Biology Institute, Universitas "Miguel Hernández" , E-03202 Elche-Alicante, Spain
| | | |
Collapse
|
18
|
Shalom-Elazari H, Zazrin-Greenspon H, Shaked H, Chill JH. Global fold and backbone dynamics of the hepatitis C virus E2 glycoprotein transmembrane domain determined by NMR. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:2919-28. [PMID: 25109935 DOI: 10.1016/j.bbamem.2014.07.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 06/30/2014] [Accepted: 07/22/2014] [Indexed: 01/13/2023]
Abstract
E1 and E2 are two hepatitis C viral envelope glycoproteins that assemble into a heterodimer that is essential for membrane fusion and penetration into the target cell. Both extracellular and transmembrane (TM) glycoprotein domains contribute to this interaction, but study of TM-TM interactions has been limited because synthesis and structural characterization of these highly hydrophobic segments present significant challenges. In this NMR study, by successful expression and purification of the E2 transmembrane domain as a fusion construct we have determined the global fold and characterized backbone motions for this peptide incorporated in phospholipid micelles. Backbone resonance frequencies, relaxation rates and solvent exposure measurements concur in showing this domain to adopt a helical conformation, with two helical segments spanning residues 717-726 and 732-746 connected by an unstructured linker containing the charged residues D728 and R730 involved in E1 binding. Although this linker exhibits increased local motions on the ps timescale, the dominating contribution to its relaxation is the global tumbling motion with an estimated correlation time of 12.3ns. The positioning of the helix-linker-helix architecture within the mixed micelle was established by paramagnetic NMR spectroscopy and phospholipid-peptide cross relaxation measurements. These indicate that while the helices traverse the hydrophobic interior of the micelle, the linker lies closer to the micelle perimeter to accommodate its charged residues. These results lay the groundwork for structure determination of the E1/E2 complex and a molecular understanding of glycoprotein heterodimerization.
Collapse
Affiliation(s)
| | | | - Hadassa Shaked
- Department of Chemistry, Bar Ilan University, Ramat Gan 52900, Israel
| | - Jordan H Chill
- Department of Chemistry, Bar Ilan University, Ramat Gan 52900, Israel.
| |
Collapse
|
19
|
Nemésio H, Villalaín J. Membranotropic Regions of the Dengue Virus prM Protein. Biochemistry 2014; 53:5280-9. [DOI: 10.1021/bi500724k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Henrique Nemésio
- Instituto de Biología
Molecular y Celular, Universidad “Miguel Hernández”, E-03202 Elche-Alicante, Spain
| | - José Villalaín
- Instituto de Biología
Molecular y Celular, Universidad “Miguel Hernández”, E-03202 Elche-Alicante, Spain
| |
Collapse
|
20
|
Unexpected structural features of the hepatitis C virus envelope protein 2 ectodomain. J Virol 2014; 88:10280-8. [PMID: 24991010 DOI: 10.1128/jvi.00874-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatitis C virus (HCV), a member of the family Flaviviridae, is a leading cause of chronic liver disease and cancer. Recent advances in HCV therapeutics have resulted in improved cure rates, but an HCV vaccine is not available and is urgently needed to control the global pandemic. Vaccine development has been hampered by the lack of high-resolution structural information for the two HCV envelope glycoproteins, E1 and E2. Recently, Kong and coworkers (Science 342:1090-1094, 2013, doi:10.1126/science.1243876) and Khan and coworkers (Nature 509[7500]:381-384, 2014, doi:10.1038/nature13117) independently determined the structure of the HCV E2 ectodomain core with some unexpected and informative results. The HCV E2 ectodomain core features a globular architecture with antiparallel β-sheets forming a central β sandwich. The residues comprising the epitopes of several neutralizing and nonneutralizing human monoclonal antibodies were also determined, which is an essential step toward obtaining a fine map of the human humoral response to HCV. Also clarified were the regions of E2 that directly bind CD81, an important HCV cellular receptor. While it has been widely assumed that HCV E2 is a class II viral fusion protein (VFP), the newly determined structure suggests that the HCV E2 ectodomain shares structural and functional similarities only with domain III of class II VFPs. The new structural determinations suggest that the HCV glycoproteins use a different mechanism than that used by class II fusion proteins for cell fusion.
Collapse
|
21
|
Identification of conserved residues in hepatitis C virus envelope glycoprotein E2 that modulate virus dependence on CD81 and SRB1 entry factors. J Virol 2014; 88:10584-97. [PMID: 24990994 DOI: 10.1128/jvi.01402-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED In spite of the high variability of its sequence, hepatitis C virus (HCV) envelope glycoprotein E2 contains several conserved regions. In this study, we explored the structural and functional features of the highly conserved E2 segment from amino acid (aa) 502 to 520, which had been proposed as a fusion peptide and shown to strongly overlap a potential conserved neutralizing epitope. For this purpose, we used reverse genetics to introduce point mutations within this region, and we characterized the phenotypes of these mutants in the light of the recently published structure of E2. The functional analyses showed that their phenotypes are in agreement with the positions of the corresponding residues in the E2 crystal structure. In contrast, our data ruled out the involvement of this region in membrane fusion, and they indicate that alternative conformations would be necessary to expose the potential neutralizing epitope present in this segment. Of particular interest, we identified three specific mutations (Y507L, V514A, and V515A) located within this neutralizing epitope which only mildly reduced infectivity and showed no assembly defect. These mutations modulated HCV dependence on the viral receptor SRB1, and/or they also modulated virion sensitivity to neutralizing antibodies. Importantly, their characterization also showed that amino acids Y507, V514, and V515 contribute to E2 interaction with HCV receptor CD81. In conclusion, our data show that the highly conserved E2 segment from aa 502 to 520 plays a key role in cell entry by influencing the association of the viral particle with coreceptors and neutralizing antibodies. IMPORTANCE Hepatitis C virus (HCV) envelope proteins E1 and E2 exhibit sequence variability. However, some segments of the envelope proteins are highly conserved, suggesting that these sequences play a key role at some steps of the HCV life cycle. In this work, we characterized the function and structure of a highly conserved E2 region that is targeted by neutralizing antibodies and had been proposed as a fusion peptide. Our data ruled out the involvement of this region in membrane fusion but allowed for the identification of new residues modulating the interaction of the virus with entry factors and its sensitivity to neutralizing antibodies. Moreover, structural data suggest that alternative conformations could exist for E2, which would explain the presence of a partially masked neutralizing epitope in this segment in the currently available E2 structure. Overall, our findings highlight the importance of conserved regions in the sequences of HCV envelope proteins.
Collapse
|
22
|
Tello D, Rodríguez-Rodríguez M, Ortega S, Lombana L, Yélamos B, Gómez-Gutiérrez J, Peterson DL, Gavilanes F. Fusogenic properties of the ectodomains of hepatitis C virus envelope proteins. FEBS J 2014; 281:2558-69. [DOI: 10.1111/febs.12802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 03/21/2014] [Accepted: 03/31/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Daniel Tello
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas; Universidad Complutense; Madrid Spain
| | - Mar Rodríguez-Rodríguez
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas; Universidad Complutense; Madrid Spain
| | - Sara Ortega
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas; Universidad Complutense; Madrid Spain
| | - Laura Lombana
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas; Universidad Complutense; Madrid Spain
| | - Belén Yélamos
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas; Universidad Complutense; Madrid Spain
| | - Julián Gómez-Gutiérrez
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas; Universidad Complutense; Madrid Spain
| | - Darrell L. Peterson
- Department of Biochemistry and Molecular Biology; Medical College of Virginia; Virginia Commonwealth University; Richmond VA USA
| | - Francisco Gavilanes
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas; Universidad Complutense; Madrid Spain
| |
Collapse
|
23
|
Apellániz B, Huarte N, Largo E, Nieva JL. The three lives of viral fusion peptides. Chem Phys Lipids 2014; 181:40-55. [PMID: 24704587 PMCID: PMC4061400 DOI: 10.1016/j.chemphyslip.2014.03.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/19/2014] [Accepted: 03/20/2014] [Indexed: 02/07/2023]
Abstract
The presence of a fusion peptide (FP) is a hallmark of viral fusion glycoproteins. Structure–function relationships underlying FP conservation remain greatly unknown. FPs establish interactions satisfying their folding within pre-fusion glycoproteins. Upon fusion activation FPs insert into and restructure target membranes. FPs can finally combine with transmembrane domains to form integral membrane bundles.
Fusion peptides comprise conserved hydrophobic domains absolutely required for the fusogenic activity of glycoproteins from divergent virus families. After 30 years of intensive research efforts, the structures and functions underlying their high degree of sequence conservation are not fully elucidated. The long-hydrophobic viral fusion peptide (VFP) sequences are structurally constrained to access three successive states after biogenesis. Firstly, the VFP sequence must fulfill the set of native interactions required for (meta) stable folding within the globular ectodomains of glycoprotein complexes. Secondly, at the onset of the fusion process, they get transferred into the target cell membrane and adopt specific conformations therein. According to commonly accepted mechanistic models, membrane-bound states of the VFP might promote the lipid bilayer remodeling required for virus-cell membrane merger. Finally, at least in some instances, several VFPs co-assemble with transmembrane anchors into membrane integral helical bundles, following a locking movement hypothetically coupled to fusion-pore expansion. Here we review different aspects of the three major states of the VFPs, including the functional assistance by other membrane-transferring glycoprotein regions, and discuss briefly their potential as targets for clinical intervention.
Collapse
Affiliation(s)
- Beatriz Apellániz
- Biophysics Unit (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain
| | - Nerea Huarte
- Biophysics Unit (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain
| | - Eneko Largo
- Biophysics Unit (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain
| | - José L Nieva
- Biophysics Unit (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain.
| |
Collapse
|
24
|
Zazrin H, Shaked H, Chill JH. Architecture of the hepatitis C virus E1 glycoprotein transmembrane domain studied by NMR. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:784-92. [PMID: 24192053 DOI: 10.1016/j.bbamem.2013.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 10/24/2013] [Accepted: 10/28/2013] [Indexed: 10/26/2022]
Abstract
Oligomerization of hepatitis C viral envelope proteins E1 and E2 is essential to virus fusion and assembly. Although interactions within the transmembrane (TM) domains of these glycoproteins have proven contributions to the E1/E2 heterodimerization process and consequent infectivity, there is little structural information on this entry mechanism. Here, as a first step towards our long-term goal of understanding the interaction between E1 and E2 TM-domains, we have expressed, purified and characterized E1-TM using structural biomolecular NMR methods. An MBP-fusion expression system yielded sufficient quantities of pure E1-TM, which was solubilized in two membrane-mimicking environments, SDS- and LPPG-micelles, affording samples amenable to NMR studies. Triple resonance assignment experiments and relaxation measurements provided information on the secondary structure and global fold of E1-TM in these environments. In SDS micelles E1-TM adopts a helical conformation, with helical stretches at residues 354-363 and 371-379 separated by a more flexible segment of residues 364-370. In LPPG micelles a helical conformation was observed for residues 354-377 with greater flexibility in the 366-367 dyad, suggesting LPPG provides a more native environment for the peptide. Replacement of key positively charged residue K370 with an alanine did not affect the secondary structure of E1-TM but did change the relative positioning within the micelle of the two helices. These results lay the foundation for structure determination of E1-TM and a molecular understanding of how E1-TM flexibility enhances its interaction with E2-TM during heterodimerization and membrane fusion.
Collapse
Affiliation(s)
- Hadas Zazrin
- Department of Chemistry, Bar Ilan University, Ramat Gan 52900, Israel
| | - Hadassa Shaked
- Department of Chemistry, Bar Ilan University, Ramat Gan 52900, Israel
| | - Jordan H Chill
- Department of Chemistry, Bar Ilan University, Ramat Gan 52900, Israel.
| |
Collapse
|
25
|
Palomares-Jerez MF, Nemesio H, Franquelim HG, Castanho MARB, Villalaín J. N-terminal AH2 segment of protein NS4B from hepatitis C virus. Binding to and interaction with model biomembranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1938-52. [PMID: 23639583 DOI: 10.1016/j.bbamem.2013.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 01/30/2023]
Abstract
HCV NS4B, a highly hydrophobic protein involved in the alteration of the intracellular host membranes forming the replication complex, plays a critical role in the HCV life cycle. NS4B is a multifunctional membrane protein that possesses different regions where diverse and significant functions are located. One of these important regions is the AH2 segment, which besides being highly conserved has been shown to play a significant role in NS4B functioning. We have carried out an in-depth biophysical study aimed at the elucidation of the capacity of this region to interact, modulate and disrupt membranes, as well as to study the structural and dynamic features relevant for that disruption. We show that a peptide derived from this region, NS4BAH2, is capable of specifically binding phosphatidyl inositol phosphates with high affinity, and its interfacial properties suggest that this segment could behave similarly to a pre-transmembrane domain partitioning into and interacting with the membrane depending on the membrane composition and/or other proteins. Moreover, NS4BAH2 is capable of rupturing membranes even at very low peptide-to-lipid ratios and its membrane-activity is modulated by lipid composition. NS4BAH2 is located in a shallow position in the membrane but it is able to affect the lipid environment from the membrane surface down to the hydrophobic core. The NS4B region where peptide NS4BAH2 resides might have an essential role in the membrane replication and/or assembly of the viral particle through the modulation of the membrane structure and hence the replication complex.
Collapse
|
26
|
Iourin O, Harlos K, El Omari K, Lu W, Kadlec J, Iqbal M, Meier C, Palmer A, Jones I, Thomas C, Brownlie J, Grimes JM, Stuart DI. Expression, purification and crystallization of the ectodomain of the envelope glycoprotein E2 from Bovine viral diarrhoea virus. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 69:35-8. [PMID: 23295482 DOI: 10.1107/s1744309112049184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 11/29/2012] [Indexed: 01/17/2023]
Abstract
Bovine viral diarrhoea virus (BVDV) is an economically important animal pathogen which is closely related to Hepatitis C virus. Of the structural proteins, the envelope glycoprotein E2 of BVDV is the major antigen which induces neutralizing antibodies; thus, BVDV E2 is considered as an ideal target for use in subunit vaccines. Here, the expression, purification of wild-type and mutant forms of the ectodomain of BVDV E2 and subsequent crystallization and data collection of two crystal forms grown at low and neutral pH are reported. Native and multiple-wavelength anomalous dispersion (MAD) data sets have been collected and structure determination is in progress.
Collapse
Affiliation(s)
- Oleg Iourin
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, England
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Nemésio H, Palomares-Jerez F, Villalaín J. NS4A and NS4B proteins from dengue virus: Membranotropic regions. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2818-30. [DOI: 10.1016/j.bbamem.2012.06.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/26/2012] [Accepted: 06/27/2012] [Indexed: 10/28/2022]
|
28
|
Distinct roles in folding, CD81 receptor binding and viral entry for conserved histidine residues of hepatitis C virus glycoprotein E1 and E2. Biochem J 2012; 443:85-94. [DOI: 10.1042/bj20110868] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The protonation of histidine in acidic environments underpins its role in regulating the function of pH-sensitive proteins. For pH-sensitive viral fusion proteins, histidine protonation in the endosome leads to the activation of their membrane fusion function. The HCV (hepatitis C virus) glycoprotein E1–E2 heterodimer mediates membrane fusion within the endosome, but the roles of conserved histidine residues in the formation of a functional heterodimer and in sensing pH changes is unknown. We examined the functional roles of conserved histidine residues located within E1 and E2. The E1 mutations, H222A/R, H298R and H352A, disrupted E1–E2 heterodimerization and reduced virus entry. A total of five out of six histidine residues located within the E2 RBD (receptor-binding domain) were important for the E2 fold, and their substitution with arginine or alanine caused aberrant heterodimerization and/or CD81 binding. Distinct roles in E1–E2 heterodimerization and in virus entry were identified for His691 and His693 respectively within the membrane-proximal stem region. Viral entry and cell–cell fusion at neutral and low pH values were enhanced with H445R, indicating that the protonation state of His445 is a key regulator of HCV fusion. However, H445R did not overcome the block to virus entry induced by bafilomycin A1, indicating a requirement for an endosomal activation trigger in addition to acidic pH.
Collapse
|
29
|
The membrane-active regions of the dengue virus proteins C and E. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1808:2390-402. [DOI: 10.1016/j.bbamem.2011.06.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/23/2011] [Accepted: 06/28/2011] [Indexed: 12/24/2022]
|
30
|
Helle F, Duverlie G, Dubuisson J. The hepatitis C virus glycan shield and evasion of the humoral immune response. Viruses 2011; 3:1909-32. [PMID: 22069522 PMCID: PMC3205388 DOI: 10.3390/v3101909] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 09/28/2011] [Accepted: 10/01/2011] [Indexed: 12/14/2022] Open
Abstract
Despite the induction of effective immune responses, 80% of hepatitis C virus (HCV)-infected individuals progress from acute to chronic hepatitis. In contrast to the cellular immune response, the role of the humoral immune response in HCV clearance is still subject to debate. Indeed, HCV escapes neutralizing antibodies in chronically infected patients and reinfection has been described in human and chimpanzee. Studies of antibody-mediated HCV neutralization have long been hampered by the lack of cell-culture-derived virus and the absence of a small animal model. However, the development of surrogate models and recent progress in HCV propagation in vitro now enable robust neutralization assays to be performed. These advances are beginning to shed some light on the mechanisms of HCV neutralization. This review summarizes the current state of knowledge of the viral targets of anti-HCV-neutralizing antibodies and the mechanisms that enable HCV to evade the humoral immune response. The recent description of the HCV glycan shield that reduces the immunogenicity of envelope proteins and masks conserved neutralizing epitopes at their surface constitutes the major focus of this review.
Collapse
Affiliation(s)
- François Helle
- Laboratory of Virology, EA4294, Jules Verne University of Picardie, Amiens 80000, France; E-Mail:
| | - Gilles Duverlie
- Laboratory of Virology, EA4294, Jules Verne University of Picardie, Amiens 80000, France; E-Mail:
- Virology Department, Amiens University Hospital Center, South Hospital, Amiens 80000, 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 59021, France; E-Mail:
| |
Collapse
|
31
|
Fraser J, Boo I, Poumbourios P, Drummer HE. Hepatitis C virus (HCV) envelope glycoproteins E1 and E2 contain reduced cysteine residues essential for virus entry. J Biol Chem 2011; 286:31984-92. [PMID: 21768113 PMCID: PMC3173156 DOI: 10.1074/jbc.m111.269605] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/13/2011] [Indexed: 12/14/2022] Open
Abstract
The HCV envelope glycoproteins E1 and E2 contain eight and 18 highly conserved cysteine residues, respectively. Here, we examined the oxidation state of E1E2 heterodimers incorporated into retroviral pseudotyped particles (HCVpp) and investigated the significance of free sulfhydryl groups in cell culture-derived HCV (HCVcc) and HCVpp entry. Alkylation of free sulfhydryl groups on HCVcc/pp with a membrane-impermeable sulfhydryl-alkylating reagent 4-(N-maleimido)benzyl-α-trimethylammonium iodide (M135) prior to virus attachment to cells abolished infectivity in a dose-dependent manner. Labeling of HCVpp envelope proteins with EZ-Link maleimide-PEG2-biotin (maleimide-biotin) detected free thiol groups in both E1 and E2. Unlike retroviruses that employ disulfide reduction to facilitate virus entry, the infectivity of alkylated HCVcc could not be rescued by addition of exogenous reducing agents. Furthermore, the infectivity of HCVcc bound to target cells was not affected by addition of M135 indicative of a change in glycoprotein oxidation state from reduced to oxidized following virus attachment to cells. By contrast, HCVpp entry was reduced by 61% when treated with M135 immediately following attachment to cells, suggesting that the two model systems might demonstrate variations in oxidation kinetics. Glycoprotein oxidation was not altered following binding of HCVpp incorporated E1E2 to soluble heparin or recombinant CD81. These results suggest that HCV entry is dependent on the presence of free thiol groups in E1 and E2 prior to cellular attachment and reveals a new essential component of the HCV entry process.
Collapse
Affiliation(s)
- Johanna Fraser
- From the Burnet Institute, 85 Commercial Road, Melbourne 3004, Australia
- the Department of Microbiology, Monash University, Clayton 3800, Australia, and
| | - Irene Boo
- From the Burnet Institute, 85 Commercial Road, Melbourne 3004, Australia
| | - Pantelis Poumbourios
- From the Burnet Institute, 85 Commercial Road, Melbourne 3004, Australia
- the Department of Microbiology, Monash University, Clayton 3800, Australia, and
| | - Heidi E. Drummer
- From the Burnet Institute, 85 Commercial Road, Melbourne 3004, Australia
- the Department of Microbiology, Monash University, Clayton 3800, Australia, and
- the Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
32
|
Verstrepen BE, Depla E, Rollier CS, Mares G, Drexhage JAR, Priem S, Verschoor EJ, Koopman G, Granier C, Dreux M, Cosset FL, Maertens G, Heeney JL. Clearance of genotype 1b hepatitis C virus in chimpanzees in the presence of vaccine-induced E1-neutralizing antibodies. J Infect Dis 2011; 204:837-44. [PMID: 21849281 PMCID: PMC3156919 DOI: 10.1093/infdis/jir423] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 04/12/2011] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence indicates that neutralizing antibodies play an important role in protection from chronic hepatitis C virus (HCV) infection. Efforts to elicit such responses by immunization with intact heterodimeric E1E2 envelope proteins have met with limited success. To determine whether antigenic sites, which are not exposed by the combined E1E2 heterodimer structure, are capable of eliciting neutralizing antibody responses, we expressed and purified each as separate recombinant proteins E1 and E2, from which the immunodominant hypervariable region (HVR-1) was deleted. Immunization of chimpanzees with either E1 or E2 alone induced antigen-specific T-helper cytokines of similar magnitude. Unexpectedly, the capacity to neutralize HCV was observed in E1 but not in animals immunized with E2 devoid of HVR-1. Furthermore, in vivo only E1-vaccinated animals exposed to the heterologous HCV-1b inoculum cleared HCV infection.
Collapse
Affiliation(s)
- Babs E. Verstrepen
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Erik Depla
- Innogenetics N.V, Industriepark Zwijnaarde, Ghent, Belgium
| | - Christine S. Rollier
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Gwenny Mares
- Innogenetics N.V, Industriepark Zwijnaarde, Ghent, Belgium
| | - Joost A. R. Drexhage
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Sofie Priem
- Innogenetics N.V, Industriepark Zwijnaarde, Ghent, Belgium
| | - Ernst J. Verschoor
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Christelle Granier
- Université de Lyon, UCB Lyon-1, INSERM, U758, Human Virology laboratory, EVIR Team, Lyon; Ecole Normale Supérieure de Lyon, France
| | - Marlène Dreux
- Université de Lyon, UCB Lyon-1, INSERM, U758, Human Virology laboratory, EVIR Team, Lyon; Ecole Normale Supérieure de Lyon, France
| | - François L. Cosset
- Université de Lyon, UCB Lyon-1, INSERM, U758, Human Virology laboratory, EVIR Team, Lyon; Ecole Normale Supérieure de Lyon, France
| | - Geert Maertens
- Innogenetics N.V, Industriepark Zwijnaarde, Ghent, Belgium
| | - Jonathan L. Heeney
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
- Laboratory of Viral Zoonotics, University of Cambridge, CB3-OES, United Kingdom
| |
Collapse
|
33
|
Rychłowska M, Owsianka AM, Foung SKH, Dubuisson J, Bieńkowska-Szewczyk K, Patel AH. Comprehensive linker-scanning mutagenesis of the hepatitis C virus E1 and E2 envelope glycoproteins reveals new structure-function relationships. J Gen Virol 2011; 92:2249-2261. [PMID: 21697343 PMCID: PMC3347801 DOI: 10.1099/vir.0.034314-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite extensive research, many details about the structure and functions of hepatitis C virus (HCV) glycoproteins E1 and E2 are not fully understood, and their crystal structure remains to be determined. We applied linker-scanning mutagenesis to generate a panel of 34 mutants, each containing an insertion of 5 aa at a random position within the E1E2 sequence. The mutated glycoproteins were analysed by using a range of assays to identify regions critical for maintaining protein conformation, E1E2 complex assembly, CD81 receptor binding, membrane fusion and infectivity. The results, while supporting previously published data, provide several interesting new findings. Firstly, insertion at amino acid 587 or 596 reduced E1E2 heterodimerization without affecting reactivity with some conformation-sensitive mAbs or with CD81, thus implicating these residues in glycoprotein assembly. Secondly, insertions within a conserved region of E2, between amino acid residues 611 and 631, severely disrupted protein conformation and abrogated binding of all conformation-sensitive antibodies, suggesting that the structural integrity of this region is critical for the correct folding of E2. Thirdly, an insertion at Leu-682 specifically affected membrane fusion, providing direct evidence that the membrane-proximal ‘stem’ of E2 is involved in the fusion mechanism. Overall, our results show that the HCV glycoproteins generally do not tolerate insertions and that there are a very limited number of sites that can be changed without dramatic loss of function. Nevertheless, we identified two E2 insertion mutants, at amino acid residues 408 and 577, that were infectious in the murine leukemia virus-based HCV pseudoparticle system.
Collapse
Affiliation(s)
- Małgorzata Rychłowska
- MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, UK.,Department of Molecular Virology, University of Gdańsk, Kładki 24, 80-822 Gdańsk, Poland
| | - Ania M Owsianka
- MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, UK
| | - Steven K H Foung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Jean Dubuisson
- Inserm U1019, F-59019 Lille, France.,Université Lille Nord de France, F-59000 Lille, France.,Institut Pasteur de Lille, Centre for Infection & Immunity of Lille (CIIL), F-59019 Lille, France.,CNRS UMR8204, F-59021 Lille, France
| | | | - Arvind H Patel
- MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, UK
| |
Collapse
|
34
|
Albecka A, Montserret R, Krey T, Tarr AW, Diesis E, Ball JK, Descamps V, Duverlie G, Rey F, Penin F, Dubuisson J. Identification of new functional regions in hepatitis C virus envelope glycoprotein E2. J Virol 2011; 85:1777-92. [PMID: 21147916 PMCID: PMC3028898 DOI: 10.1128/jvi.02170-10] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 11/29/2010] [Indexed: 01/21/2023] Open
Abstract
Little is known about the structure of the envelope glycoproteins of hepatitis C virus (HCV). To identify new regions essential for the function of these glycoproteins, we generated HCV pseudoparticles (HCVpp) containing HCV envelope glycoproteins, E1 and E2, from different genotypes in order to detect intergenotypic incompatibilities between these two proteins. Several genotype combinations were nonfunctional for HCV entry. Of interest, a combination of E1 from genotype 2a and E2 from genotype 1a was nonfunctional in the HCVpp system. We therefore used this nonfunctional complex and the recently described structural model of E2 to identify new functional regions in E2 by exchanging protein regions between these two genotypes. The functionality of these chimeric envelope proteins in the HCVpp system and/or the cell-cultured infectious virus (HCVcc) was analyzed. We showed that the intergenotypic variable region (IgVR), hypervariable region 2 (HVR2), and another segment in domain II play a role in E1E2 assembly. We also demonstrated intradomain interactions within domain I. Importantly, we also identified a segment (amino acids [aa] 705 to 715 [segment 705-715]) in the stem region of E2, which is essential for HCVcc entry. Circular dichroism and nuclear magnetic resonance structural analyses of the synthetic peptide E2-SC containing this segment revealed the presence of a central amphipathic helix, which likely folds upon membrane binding. Due to its location in the stem region, segment 705-715 is likely involved in the reorganization of the glycoprotein complexes taking place during the fusion process. In conclusion, our study highlights new functional and structural regions in HCV envelope glycoprotein E2.
Collapse
Affiliation(s)
- Anna Albecka
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Roland Montserret
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Thomas Krey
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Alexander W. Tarr
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Eric Diesis
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Jonathan K. Ball
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Véronique Descamps
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Gilles Duverlie
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Felix Rey
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - François Penin
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Jean Dubuisson
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), F-59019 Lille, France; Inserm U1019, F-59019 Lille, France; CNRS UMR8204, F-59021 Lille, France; and Université Lille Nord de France, F-59000 Lille, France, Institut de Biologie et Chimie des Protéines, UMR-5086-CNRS, Université de Lyon, Lyon, France, Institut Pasteur, CNRS URA3015, Unité de Virologie Structurale, Paris, France, School of Molecular Medical Sciences, the University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom, Laboratoire de Virologie EA4294, Centre Hospitalier Universitaire d'Amiens, Université de Picardie Jules Verne, Amiens, France
| |
Collapse
|
35
|
Mechanism of inhibition of enveloped virus membrane fusion by the antiviral drug arbidol. PLoS One 2011; 6:e15874. [PMID: 21283579 PMCID: PMC3026800 DOI: 10.1371/journal.pone.0015874] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 11/28/2010] [Indexed: 12/13/2022] Open
Abstract
The broad-spectrum antiviral arbidol (Arb) inhibits cell entry of enveloped viruses by blocking viral fusion with host cell membrane. To better understand Arb mechanism of action, we investigated its interactions with phospholipids and membrane peptides. We demonstrate that Arb associates with phospholipids in the micromolar range. NMR reveals that Arb interacts with the polar head-group of phospholipid at the membrane interface. Fluorescence studies of interactions between Arb and either tryptophan derivatives or membrane peptides reconstituted into liposomes show that Arb interacts with tryptophan in the micromolar range. Interestingly, apparent binding affinities between lipids and tryptophan residues are comparable with those of Arb IC50 of the hepatitis C virus (HCV) membrane fusion. Since tryptophan residues of membrane proteins are known to bind preferentially at the membrane interface, these data suggest that Arb could increase the strength of virus glycoprotein's interactions with the membrane, due to a dual binding mode involving aromatic residues and phospholipids. The resulting complexation would inhibit the expected viral glycoprotein conformational changes required during the fusion process. Our findings pave the way towards the design of new drugs exhibiting Arb-like interfacial membrane binding properties to inhibit early steps of virus entry, i.e., attractive targets to combat viral infection.
Collapse
|
36
|
Palma-Guerrero J, Lopez-Jimenez JA, Pérez-Berná AJ, Huang IC, Jansson HB, Salinas J, Villalaín J, Read ND, Lopez-Llorca LV. Membrane fluidity determines sensitivity of filamentous fungi to chitosan. Mol Microbiol 2010; 75:1021-32. [DOI: 10.1111/j.1365-2958.2009.07039.x] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
37
|
Structural characterization of the transmembrane proximal region of the hepatitis C virus E1 glycoprotein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:344-53. [PMID: 19891955 DOI: 10.1016/j.bbamem.2009.10.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 10/16/2009] [Accepted: 10/27/2009] [Indexed: 11/21/2022]
Abstract
A detailed knowledge of the mechanism of virus entry represents one of the most promising approaches to develop new therapeutic strategies. However, viral fusion is a very complex process involving fusion glycoproteins present on the viral envelope. In the two hepatitis C virus envelope proteins, E1 and E2, several membranotropic regions with a potential role in the fusion process have been identified. Among these, we have selected the 314-342 E1 region. Circular Dichroism data indicate that the peptide exhibits a clear propensity to adopt a helical folding in different membrane mimicking media, such as mixtures of water with fluorinated alcohols and phospholipids, with a slight preference for negative charged bilayers. The 3D structure of E1(314-342) peptide, calculated by 2D-NMR in a low-polarity environment, consists of two helical stretches encompassing residues 319-323 and 329-338 respectively. The peptide, presenting a largely apolar character, interacts with liposomes, as indicated by fluorescence and electron spin resonance spectra. The strength of the interaction and the deepness of peptide insertion in the phospholipid membrane are modulated by the bilayer composition, the interaction with anionic phospholipids being among the strongest ever observed. The presence of cholesterol also affects the peptide-bilayer interaction, favoring the peptide positioning close to the bilayer surface. Overall, the experimental data support the idea that this region of E1 might be involved in membrane destabilization and viral fusion; therefore it may represent a good target to develop anti-viral molecules.
Collapse
|
38
|
Li HF, Huang CH, Ai LS, Chuang CK, Chen SSL. Mutagenesis of the fusion peptide-like domain of hepatitis C virus E1 glycoprotein: involvement in cell fusion and virus entry. J Biomed Sci 2009; 16:89. [PMID: 19778418 PMCID: PMC2759930 DOI: 10.1186/1423-0127-16-89] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Accepted: 09/24/2009] [Indexed: 01/19/2023] Open
Abstract
Background Envelope (E) glycoprotein E2 of the hepatitis C virus (HCV) mediates binding of the virus to target cell receptors. Nevertheless, the precise role of E1 in viral entry remains elusive. Methods To understand the involvement of the fusion peptide-like domain positioned at residues 264 to 290 within envelope glycoprotein E1 in HCV infection, mutants with Ala and Asn substitutions for residues conserved between HCV and E proteins of flaviviruses or the fusion proteins of paramyxoviruses were constructed by site-directed mutagenesis and their effects on membrane fusion and viral infectivity were examined. Results None of these mutations affected the synthesis or cell surface expression of envelope proteins, nor did they alter the formation of a non-covalent E1-E2 heterodimer or E2 binding to the large extracellular loop of CD81. The Cys residues located at positions 272 and 281 were unlikely involved in intra- or intermolecular disulfide bond formation. With the exception of the G267A mutant, which showed increased cell fusion, other mutants displayed reduced or marginally inhibited cell fusion capacities compared to the wild-type (WT) E1E2. The G267A mutant was also an exception in human immunodeficiency virus type 1 (HIV-1)/HCV E1E2 pseudotyping analyses, in that it showed higher one-cycle infectivity; all other mutants exhibited greatly or partially reduced viral entry versus the WT pseudotype. All but the G278A and D279N mutants showed a WT-like profile of E1E2 incorporation into HIV-1 particles. Since C272A, C281A, G282A, and G288A pseudotypes bound to Huh7 cells as effectively as did the WT pseudotype, the reduced infectivity of these pseudotypes was due to their ability to inhibit cell fusion. Conclusion Our results indicate that specific residues, but not the structure, of this fusion peptide-like domain are required for mediating cell fusion and viral entry.
Collapse
Affiliation(s)
- Hsiao-Fen Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.
| | | | | | | | | |
Collapse
|
39
|
Russell RS, Kawaguchi K, Meunier JC, Takikawa S, Faulk K, Bukh J, Purcell RH, Emerson SU. Mutational analysis of the hepatitis C virus E1 glycoprotein in retroviral pseudoparticles and cell-culture-derived H77/JFH1 chimeric infectious virus particles. J Viral Hepat 2009; 16:621-32. [PMID: 19302336 PMCID: PMC2770242 DOI: 10.1111/j.1365-2893.2009.01111.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Cell entry by enveloped viruses is mediated by viral glycoproteins, and generally involves a short hydrophobic peptide (fusion peptide) that inserts into the cellular membrane. An internal hydrophobic domain within E1 (aa262-290) of hepatitis C virus (HCV) may function as a fusion peptide. Retrovirus-based HCV-pseudotyped viruses (HCVpp; genotype 1a) containing Ala or Pro substitutions at conserved amino acid positions within this putative fusion peptide were generated. Mutation of conserved residues significantly reduced efficiency of HCVpp entry into Huh-7 cells. The majority of amino acid substitutions appeared to disrupt necessary interactions between E1 and E2. For some mutants, reductions in HCVpp-associated E1 were associated with the incorporation of a high molecular weight, hyperglycosylated E2 that displayed decreased CD81-binding. Other entry-deficient mutants displayed normal E1E2 incorporation into pseudoparticles and normal CD81-binding, and therefore might affect viral fusion. One mutant (S283P) consistently displayed two- to threefold higher infectivity than did wild-type. Three mutations that decreased HCVpp infectivity also reduced levels of HCVcc infectious virus production. However, the S283P mutation had a different effect in the two systems as it did not increase production of infectious HCVcc. This comprehensive mutational analysis of the putative HCV fusion peptide provides insight into the role of E1 in its interaction with E2 and in HCV entry.
Collapse
Affiliation(s)
- R. S. Russell
- Hepatitis Viruses, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - K. Kawaguchi
- Hepatitis Viruses, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J.-C. Meunier
- Hepatitis Viruses, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - S. Takikawa
- Hepatitis Viruses, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - K. Faulk
- Hepatitis Viruses, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J. Bukh
- Hepatitis Viruses, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA,Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, and Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R. H. Purcell
- Hepatitis Viruses, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - S. U. Emerson
- Molecular Hepatitis Sections, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
40
|
Pérez-Berná AJ, Pabst G, Laggner P, Villalaín J. Biophysical characterization of the fusogenic region of HCV envelope glycoprotein E1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:2183-93. [PMID: 19698697 DOI: 10.1016/j.bbamem.2009.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 07/27/2009] [Accepted: 08/04/2009] [Indexed: 01/08/2023]
Abstract
We have studied the binding and interaction of the peptide E1(FP) with various model membranes. E1(FP) is derived from the amino acid segment 274-291 of the hepatitis C virus envelope glycoprotein E1, which was previously proposed to host the peptide responsible for fusion to target membranes. In the present study we addressed the changes which take place upon E1(FP) binding in both the peptide and the phospholipid bilayer, respectively, through a series of complementary experiments. We show that peptide E1(FP) binds to and interacts with phospholipid model membranes, modulates the polymorphic phase behavior of membrane phospholipids, is localized in a shallow position in the membrane and interacts preferentially with cholesterol. The capability of modifying the biophysical properties of model membranes supports its role in HCV-mediated membrane fusion and suggests that the mechanism of membrane fusion elicited by class I and II fusion proteins might be similar.
Collapse
Affiliation(s)
- Ana J Pérez-Berná
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, E-03202 Alicante, Spain
| | | | | | | |
Collapse
|
41
|
Bruni R, Costantino A, Tritarelli E, Marcantonio C, Ciccozzi M, Rapicetta M, El Sawaf G, Giuliani A, Ciccaglione AR. A computational approach identifies two regions of Hepatitis C Virus E1 protein as interacting domains involved in viral fusion process. BMC STRUCTURAL BIOLOGY 2009; 9:48. [PMID: 19640267 PMCID: PMC2732612 DOI: 10.1186/1472-6807-9-48] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Accepted: 07/29/2009] [Indexed: 01/01/2023]
Abstract
Background The E1 protein of Hepatitis C Virus (HCV) can be dissected into two distinct hydrophobic regions: a central domain containing an hypothetical fusion peptide (FP), and a C-terminal domain (CT) comprising two segments, a pre-anchor and a trans-membrane (TM) region. In the currently accepted model of the viral fusion process, the FP and the TM regions are considered to be closely juxtaposed in the post-fusion structure and their physical interaction cannot be excluded. In the present study, we took advantage of the natural sequence variability present among HCV strains to test, by purely sequence-based computational tools, the hypothesis that in this virus the fusion process involves the physical interaction of the FP and CT regions of E1. Results Two computational approaches were applied. The first one is based on the co-evolution paradigm of interacting peptides and consequently on the correlation between the distance matrices generated by the sequence alignment method applied to FP and CT primary structures, respectively. In spite of the relatively low random genetic drift between genotypes, co-evolution analysis of sequences from five HCV genotypes revealed a greater correlation between the FP and CT domains than respect to a control HCV sequence from Core protein, so giving a clear, albeit still inconclusive, support to the physical interaction hypothesis. The second approach relies upon a non-linear signal analysis method widely used in protein science called Recurrence Quantification Analysis (RQA). This method allows for a direct comparison of domains for the presence of common hydrophobicity patterns, on which the physical interaction is based upon. RQA greatly strengthened the reliability of the hypothesis by the scoring of a lot of cross-recurrences between FP and CT peptides hydrophobicity patterning largely outnumbering chance expectations and pointing to putative interaction sites. Intriguingly, mutations in the CT region of E1, reducing the fusion process in vitro, strongly reduced the amount of cross-recurrence further supporting interaction between this region and FP. Conclusion Our results support a fusion model for HCV in which the FP and the C-terminal region of E1 are juxtaposed and interact in the post-fusion structure. These findings have general implications for viruses, as any visualization of the post-fusion FP-TM complex has been precluded by the impossibility to obtain crystallised viral fusion proteins containing the trans-membrane region. This limitation gives to sequence based modelling efforts a crucial role in the sketching of a molecular interpretation of the fusion process. Moreover, our data also have a more general relevance for cell biology as the mechanism of intracellular fusion showed remarkable similarities with viral fusion
Collapse
Affiliation(s)
- Roberto Bruni
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Guillén J, González-Alvarez A, Villalaín J. A membranotropic region in the C-terminal domain of hepatitis C virus protein NS4B interaction with membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:327-37. [PMID: 19631190 DOI: 10.1016/j.bbamem.2009.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 07/05/2009] [Accepted: 07/08/2009] [Indexed: 01/06/2023]
Abstract
We have identified a membrane-active region in the HCV NS4B protein by studying membrane rupture induced by a NS4B-derived peptide library on model membranes. This segment corresponds to one of two previously predicted amphipathic helix and define it as a new membrane association domain. We report the binding and interaction with model membranes of a peptide patterned after this segment, peptide NS4B(H2), and show that NS4B(H2) strongly partitions into phospholipid membranes, interacts with them, and is located in a shallow position in the membrane. Furthermore, changes in the primary sequence cause the disruption of the hydrophobicity along the structure and prevent the resulting peptide from interacting with the membrane. Our results suggest that the region where the NS4B(H2) is located might have an essential role in the membrane replication and/or assembly of the viral particle through the modulation of the replication complex. Our findings therefore identify an important region in the HCV NS4B protein which might be implicated in the HCV life cycle and possibly in the formation of the membranous web.
Collapse
Affiliation(s)
- Jaime Guillén
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, E-03202, Elche-Alicante, Spain
| | | | | |
Collapse
|
43
|
Hepatitis C virus NS2 protein contributes to virus particle assembly via opposing epistatic interactions with the E1-E2 glycoprotein and NS3-NS4A enzyme complexes. J Virol 2009; 83:8379-95. [PMID: 19515772 DOI: 10.1128/jvi.00891-09] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The hepatitis C virus NS2 protein has been recently implicated in virus particle assembly. To further understand the role of NS2 in this process, we conducted a reverse genetic analysis of NS2 in the context of a chimeric genotype 2a infectious cell culture system. Of 32 mutants tested, all were capable of RNA replication and 25 had moderate-to-severe defects in virus assembly. Through forward genetic selection for variants capable of virus spread, we identified second-site mutations in E1, E2, NS2, NS3, and NS4A that suppressed NS2 defects in assembly. Two suppressor mutations, E1 A78T and NS3 Q221L, were further characterized by additional genetic and biochemical experiments. Both mutations were shown to suppress other NS2 defects, often with mutual exclusivity. Thus, several NS2 mutants were enhanced by NS3 Q221L and inhibited by E1 A78T, while others were enhanced by E1 A78T and inhibited by NS3 Q221L. Furthermore, we show that the NS3 Q221L mutation lowers the affinity of native, full-length NS3-NS4A for functional RNA binding. These data reveal a complex network of interactions involving NS2 and other viral structural and nonstructural proteins during virus assembly.
Collapse
|
44
|
Pérez-Berná AJ, Pabst G, Laggner P, Villalaín J. Screening a peptide library by DSC and SAXD: comparison with the biological function of the parent proteins. PLoS One 2009; 4:e4356. [PMID: 19194494 PMCID: PMC2632743 DOI: 10.1371/journal.pone.0004356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 12/09/2008] [Indexed: 01/04/2023] Open
Abstract
We have recently identified the membranotropic regions of the hepatitis C virus proteins E1, E2, core and p7 proteins by observing the effect of protein-derived peptide libraries on model membrane integrity. We have studied in this work the ability of selected sequences of these proteins to modulate the Lβ-Lα and Lα-HII phospholipid phase transitions as well as check the viability of using both DSC and SAXD to screen a protein-derived peptide library. We demonstrate that it is feasible to screen a library of peptides corresponding to one or several proteins by both SAXD and DSC. This methodological combination should allow the identification of essential regions of membrane-interacting proteins which might be implicated in the molecular mechanism of membrane fusion and/or budding.
Collapse
Affiliation(s)
- Ana J. Pérez-Berná
- Instituto de Biología Molecular y Celular, Universidad “Miguel Hernández”, Alicante, Spain
| | - George Pabst
- Institute of Biophysics and Nanosystems Research, Austrian Academy of Sciences, Graz, Austria
| | - Peter Laggner
- Institute of Biophysics and Nanosystems Research, Austrian Academy of Sciences, Graz, Austria
| | - José Villalaín
- Instituto de Biología Molecular y Celular, Universidad “Miguel Hernández”, Alicante, Spain
- * E-mail:
| |
Collapse
|
45
|
The pre-transmembrane region of the HCV E1 envelope glycoprotein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2008; 1778:2069-80. [DOI: 10.1016/j.bbamem.2008.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 03/22/2008] [Accepted: 03/24/2008] [Indexed: 12/13/2022]
|
46
|
Gottwein JM, Bukh J. Cutting the gordian knot-development and biological relevance of hepatitis C virus cell culture systems. Adv Virus Res 2008; 71:51-133. [PMID: 18585527 DOI: 10.1016/s0065-3527(08)00002-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Worldwide approximately 180 million people are chronically infected with hepatitis C virus (HCV). HCV isolates exhibit extensive genetic heterogeneity and have been grouped in six genotypes and various subtypes. Additionally, several naturally occurring intergenotypic recombinants have been described. Research on the viral life cycle, efficient therapeutics, and a vaccine has been hampered by the absence of suitable cell culture systems. The first system permitting studies of the full viral life cycle was intrahepatic transfection of RNA transcripts of HCV consensus complementary DNA (cDNA) clones into chimpanzees. However, such full-length clones were not infectious in vitro. The development of the replicon system and HCV pseudo-particles allowed in vitro studies of certain aspects of the viral life cycle, RNA replication, and viral entry, respectively. Identification of the genotype 2 isolate JFH1, which for unknown reasons showed an exceptional replication capability and resulted in formation of infectious viral particles in the human hepatoma cell line Huh7, led in 2005 to the development of the first full viral life cycle in vitro systems. JFH1-based systems now enable in vitro studies of the function of viral proteins, their interaction with each other and host proteins, new antivirals, and neutralizing antibodies in the context of the full viral life cycle. However, several challenges remain, including development of cell culture systems for all major HCV genotypes and identification of other susceptible cell lines.
Collapse
Affiliation(s)
- Judith M Gottwein
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Copenhagen University Hospital, Hvidovre, Denmark
| | | |
Collapse
|
47
|
Kashiwada A, Matsuda K, Mizuno T, Tanaka T. Construction of a pH-Responsive Artificial Membrane Fusion System by Using Designed Coiled-Coil Polypeptides. Chemistry 2008; 14:7343-50. [DOI: 10.1002/chem.200701726] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
48
|
Guillén J, Kinnunen PKJ, Villalaín J. Membrane insertion of the three main membranotropic sequences from SARS-CoV S2 glycoprotein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2008; 1778:2765-74. [PMID: 18721794 PMCID: PMC7157930 DOI: 10.1016/j.bbamem.2008.07.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 07/09/2008] [Accepted: 07/23/2008] [Indexed: 12/02/2022]
Abstract
In order to complete the fusion process of SARS-CoV virus, several regions of the S2 virus envelope glycoprotein are necessary. Recent studies have identified three membrane-active regions in the S2 domain of SARS-CoV glycoprotein, one situated downstream of the minimum furin cleavage, which is considered the fusion peptide (SARSFP), an internal fusion peptide located immediately upstream of the HR1 region (SARSIFP) and the pre-transmembrane domain (SARSPTM). We have explored the capacity of these selected membrane-interacting regions of the S2 SARS-CoV fusion protein, alone or in equimolar mixtures, to insert into the membrane as well as to perturb the dipole potential of the bilayer. We show that the three peptides interact with lipid membranes depending on lipid composition and experiments using equimolar mixtures of these peptides show that different segments of the protein may act in a synergistic way suggesting that several membrane-active regions could participate in the fusion process of the SARS-CoV.
Collapse
Affiliation(s)
- Jaime Guillén
- Instituto de Biología Molecular y Celular, Campus de Elche, Universidad Miguel Hernández, E-03202 Elche-Alicante, Spain
| | | | | |
Collapse
|
49
|
Guillén J, de Almeida RFM, Prieto M, Villalaín J. Structural and dynamic characterization of the interaction of the putative fusion peptide of the S2 SARS-CoV virus protein with lipid membranes. J Phys Chem B 2008; 112:6997-7007. [PMID: 18489147 DOI: 10.1021/jp7118229] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The SARS coronavirus (SARS-CoV) envelope spike (S) glycoprotein, a Class I viral fusion protein, is responsible for the fusion between the membranes of the virus and the target cell. In the present work, we report a study of the binding and interaction with model membranes of a peptide pertaining to the putative fusion domain of SARS-CoV, SARS FP, as well as the structural changes that take place in both the phospholipid and the peptide molecules upon this interaction. From fluorescence and infrared spectroscopies, the peptide ability to induce membrane leakage, aggregation and fusion, as well as its affinity toward specific phospholipids, was assessed. We demonstrate that SARS FP strongly partitions into phospholipid membranes, more specifically with those containing negatively charged phospholipids, increasing the water penetration depth and displaying membrane-activity modulated by the lipid composition of the membrane. Interestingly, peptide organization is different depending if SARS FP is in water or bound to the membrane. These data suggest that SARS FP could be involved in the merging of the viral and target cell membranes by perturbing the membrane outer leaflet phospholipids and specifically interacting with negatively charged phospholipids located in the inner leaflet.
Collapse
Affiliation(s)
- Jaime Guillén
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Elche-Alicante, Spain
| | | | | | | |
Collapse
|
50
|
Galdiero S, Vitiello M, D'Isanto M, Falanga A, Cantisani M, Browne H, Pedone C, Galdiero M. The identification and characterization of fusogenic domains in herpes virus glycoprotein B molecules. Chembiochem 2008; 9:758-67. [PMID: 18311743 DOI: 10.1002/cbic.200700457] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The molecular mechanism of entry of herpes viruses requires a multicomponent fusion system. Virus entry and cell-cell fusion of Herpes simplex virus (HSV) requires four glycoproteins: gD, gB and gH/gL. The role of gB remained elusive until recently, when the crystal structure of HSV-1 gB became available. Glycoprotein B homologues represent the most highly conserved group of herpes virus glycoproteins; however, despite the high degree of sequence and structural conservation, differences in post-translational processing are observed for different members of this virus family. Whereas gB of HSV is not proteolytically processed after oligomerization, most other gB homologues are cleaved by a cellular protease into subunits that remain linked through disulfide bonds. Proteolytic cleavage is common for activation of many other viral fusion proteins, so it remains difficult to envisage a common role for different herpes virus gB structures in the fusion mechanism. We selected bovine herpes virus type 1 (BoHV-1) and herpes simplex virus type 1 (HSV-1) as representative viruses expressing cleaved and uncleaved gBs, and have screened their amino acid sequences for regions of highly interfacial hydrophobicity. Synthetic peptides corresponding to such regions were tested for their ability to induce the fusion of large unilamellar vesicles and to inhibit herpes virus infection. These results underline that several regions of the gB protein are involved in the mechanism of membrane interaction.
Collapse
Affiliation(s)
- Stefania Galdiero
- Department of Biological Sciences, Division of Biostructures, University of Naples Federico II, Via Mezzocannone 16, 80134 Napoli, Italy
| | | | | | | | | | | | | | | |
Collapse
|