1
|
Townsend JA, Fapohunda O, Wang Z, Pham H, Taylor MT, Kloss B, Ho Park S, Opella S, Aspinwall CA, Marty MT. Differences in Oligomerization of the SARS-CoV-2 Envelope Protein, Poliovirus VP4, and HIV Vpu. Biochemistry 2024; 63:241-250. [PMID: 38216552 PMCID: PMC10872257 DOI: 10.1021/acs.biochem.3c00437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2024]
Abstract
Viroporins constitute a class of viral membrane proteins with diverse roles in the viral life cycle. They can self-assemble and form pores within the bilayer that transport substrates, such as ions and genetic material, that are critical to the viral infection cycle. However, there is little known about the oligomeric state of most viroporins. Here, we use native mass spectrometry in detergent micelles to uncover the patterns of oligomerization of the full-length SARS-CoV-2 envelope (E) protein, poliovirus VP4, and HIV Vpu. Our data suggest that the E protein is a specific dimer, VP4 is exclusively monomeric, and Vpu assembles into a polydisperse mixture of oligomers under these conditions. Overall, these results revealed the diversity in the oligomerization of viroporins, which has implications for the mechanisms of their biological functions as well as their potential as therapeutic targets.
Collapse
Affiliation(s)
- Julia A. Townsend
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Oluwaseun Fapohunda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Zhihan Wang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Hieu Pham
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Michael T. Taylor
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Brian Kloss
- New York Consortium on Membrane Protein Structure, New York Structural Biology Center, New York, NY 10027, USA
| | - Sang Ho Park
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stanley Opella
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Craig A. Aspinwall
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
- Bio5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| | - Michael T. Marty
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
- Bio5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
2
|
Condezo GN, San Martín C. Maturation of Viruses. Subcell Biochem 2024; 105:503-531. [PMID: 39738956 DOI: 10.1007/978-3-031-65187-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Viral genomes are transported between cells using various structural solutions such as spherical or filamentous protein cages, alone or in combination with lipid envelopes, in assemblies of varying complexity. Morphogenesis of the new infectious particles (virions) encompasses capsid assembly from individual components (proteins, and membranes when required), genome packaging, and maturation. This final step is crucial for full infectivity. During maturation, structural and physical changes prepare the viral particles for delivering their genome into cells at the right time and location. The virion must be stabilized for travel across harsh extracellular conditions, while enabling disassembly for genome exposure to replication and translation machineries. That is, maturation has to produce metastable particles. Common maturation strategies include structural reordering, controlled proteolysis, or posttranslational modifications. Here we outline the maturation process in representative members of the six realms proposed by the latest virus taxonomic classification.
Collapse
Affiliation(s)
- Gabriela N Condezo
- Department of Macromolecular Structure, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| | - Carmen San Martín
- Department of Macromolecular Structure, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
3
|
Galitska G, Jassey A, Wagner MA, Pollack N, Miller K, Jackson WT. Enterovirus D68 capsid formation and stability requires acidic compartments. mBio 2023; 14:e0214123. [PMID: 37819109 PMCID: PMC10653823 DOI: 10.1128/mbio.02141-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE The respiratory picornavirus enterovirus D68 is a causative agent of acute flaccid myelitis, a childhood paralysis disease identified in the last decade. Poliovirus, another picornavirus associated with paralytic disease, is a fecal-oral virus that survives acidic environments when passing from host to host. Here, we follow up on our previous work showing a requirement for acidic intracellular compartments for maturation cleavage of poliovirus particles. Enterovirus D68 requires acidic vesicles for an earlier step, assembly, and maintenance of viral particles themselves. These data have strong implications for the use of acidification blocking treatments to combat enterovirus diseases.
Collapse
Affiliation(s)
- Ganna Galitska
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alagie Jassey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Michael A. Wagner
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Noah Pollack
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Katelyn Miller
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - William T. Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Andino R, Kirkegaard K, Macadam A, Racaniello VR, Rosenfeld AB. The Picornaviridae Family: Knowledge Gaps, Animal Models, Countermeasures, and Prototype Pathogens. J Infect Dis 2023; 228:S427-S445. [PMID: 37849401 DOI: 10.1093/infdis/jiac426] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023] Open
Abstract
Picornaviruses are nonenveloped particles with a single-stranded RNA genome of positive polarity. This virus family includes poliovirus, hepatitis A virus, rhinoviruses, and Coxsackieviruses. Picornaviruses are common human pathogens, and infection can result in a spectrum of serious illnesses, including acute flaccid myelitis, severe respiratory complications, and hand-foot-mouth disease. Despite research on poliovirus establishing many fundamental principles of RNA virus biology and the first transgenic animal model of disease for infection by a human virus, picornaviruses are understudied. Existing knowledge gaps include, identification of molecules required for virus entry, understanding cellular and humoral immune responses elicited during virus infection, and establishment of immune-competent animal models of virus pathogenesis. Such knowledge is necessary for development of pan-picornavirus countermeasures. Defining enterovirus A71 and D68, human rhinovirus C, and echoviruses 29 as prototype pathogens of this virus family may provide insight into picornavirus biology needed to establish public health strategies necessary for pandemic preparedness.
Collapse
Affiliation(s)
- Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Karla Kirkegaard
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, California, USA
| | - Andrew Macadam
- National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom
| | - Vincent R Racaniello
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Amy B Rosenfeld
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
5
|
Townsend JA, Fapohunda O, Wang Z, Pham H, Taylor MT, Kloss B, Park SH, Opella S, Aspinwall CA, Marty MT. Differences in Oligomerization of the SARS-CoV-2 Envelope Protein, Poliovirus VP4, and HIV Vpu. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.18.553902. [PMID: 37645758 PMCID: PMC10462163 DOI: 10.1101/2023.08.18.553902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Viroporins constitute a class of viral membrane proteins with diverse roles in the viral life cycle. They can self-assemble and form pores within the bilayer that transport substrates, such as ions and genetic material, that are critical to the viral infection cycle. However, there is little known about the oligomeric state of most viroporins. Here, we use native mass spectrometry (MS) in detergent micelles to uncover the patterns of oligomerization of the full-length SARS-CoV-2 envelope (E) protein, poliovirus VP4, and HIV Vpu. Our data suggest that the E protein is a specific dimer, VP4 is exclusively monomeric, and Vpu assembles into a polydisperse mixture of oligomers under these conditions. Overall, these results revealed the diversity in the oligomerization of viroporins, which has implications for mechanisms of their biological functions as well as their potential as therapeutic targets.
Collapse
Affiliation(s)
- Julia A. Townsend
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Oluwaseun Fapohunda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Zhihan Wang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Hieu Pham
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Michael T. Taylor
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Brian Kloss
- New York Consortium on Membrane Protein Structure, New York Structural Biology Center, New York, NY 10027, USA
| | - Sang Ho Park
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stanley Opella
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Craig A. Aspinwall
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
- Bio5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| | - Michael T. Marty
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
- Bio5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
6
|
Galitska G, Jassey A, Wagner MA, Pollack N, Jackson WT. Enterovirus D68 capsid formation and stability requires acidic compartments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544695. [PMID: 37398138 PMCID: PMC10312662 DOI: 10.1101/2023.06.12.544695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Enterovirus D68 (EV-D68), a picornavirus traditionally associated with respiratory infections, has recently been linked to a polio-like paralytic condition known as acute flaccid myelitis (AFM). EV-D68 is understudied, and much of the field's understanding of this virus is based on studies of poliovirus. For poliovirus, we previously showed that low pH promotes virus capsid maturation, but here we show that, for EV-D68, inhibition of compartment acidification during a specific window of infection causes a defect in capsid formation and maintenance. These phenotypes are accompanied by radical changes in the infected cell, with viral replication organelles clustering in a tight juxtanuclear grouping. Organelle acidification is critical during a narrow window from 3-4hpi, which we have termed the "transition point," separating translation and peak RNA replication from capsid formation, maturation and egress. Our findings highlight that acidification is crucial only when vesicles convert from RNA factories to virion crucibles.
Collapse
Affiliation(s)
- Ganna Galitska
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore St, Baltimore, MD 21201, USA
| | - Alagie Jassey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore St, Baltimore, MD 21201, USA
| | - Michael A Wagner
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore St, Baltimore, MD 21201, USA
| | - Noah Pollack
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore St, Baltimore, MD 21201, USA
| | - William T Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore St, Baltimore, MD 21201, USA
| |
Collapse
|
7
|
Miller LM, Bond KM, Draper BE, Jarrold MF. Characterization of Classical Vaccines by Charge Detection Mass Spectrometry. Anal Chem 2021; 93:11965-11972. [PMID: 34435777 DOI: 10.1021/acs.analchem.1c01893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Vaccines induce immunity by presenting disease antigens through several platforms ranging from individual protein subunits to whole viruses. Due to the large difference in antigen size, the analytical techniques employed for vaccine characterization are often platform-specific. A single, robust analytical technique capable of widespread, cross-platform use would be of great benefit and allow for comparisons across manufacturing processes. One method that spans the antigen mass range is charge detection mass spectrometry (CDMS). CDMS is a single-ion approach where the mass-to-charge ratio (m/z) and charge are measured simultaneously, allowing accurate mass distributions to be measured for heterogeneous analytes over a broad size range. In this work, CDMS was used to characterize the antigens from three classical multivalent vaccines, inactivated poliomyelitis vaccine (IPOL), RotaTeq, and Gardasil-9, directly from commercial samples. For each vaccine, the antigen purity was inspected, and in the whole virus vaccines, empty virus particles were detected. For IPOL, information on the extent of formaldehyde cross-linking was obtained. RotaTeq shows a narrow peak at 61.06 MDa. This is at a slightly lower mass than expected for the double-layer particle, suggesting that around 10 pentonal trimers are missing. For Gardasil-9, buffer exchange of the vaccine resulted in very broad mass distributions. However, removal of the virus-like particles from the aluminum adjuvant using a displacement reaction generated a spectrum with narrow peaks.
Collapse
Affiliation(s)
- Lohra M Miller
- Chemistry Department, Indiana University, 800 E Kirkwood Ave., Bloomington, Indiana 47405, United States
| | - Kevin M Bond
- Chemistry Department, Indiana University, 800 E Kirkwood Ave., Bloomington, Indiana 47405, United States
| | - Benjamin E Draper
- Megadalton Solutions, 3750 E Bluebird Lane, Bloomington, Indiana 47401, United States
| | - Martin F Jarrold
- Chemistry Department, Indiana University, 800 E Kirkwood Ave., Bloomington, Indiana 47405, United States
| |
Collapse
|
8
|
Phanthong S, Densumite J, Seesuay W, Thanongsaksrikul J, Teimoori S, Sookrung N, Poovorawan Y, Onvimala N, Guntapong R, Pattanapanyasat K, Chaicumpa W. Human Antibodies to VP4 Inhibit Replication of Enteroviruses Across Subgenotypes and Serotypes, and Enhance Host Innate Immunity. Front Microbiol 2020; 11:562768. [PMID: 33101238 PMCID: PMC7545151 DOI: 10.3389/fmicb.2020.562768] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Hand, foot, and mouth disease (HFMD) is a highly contagious disease that usually affects infants and young children (<5 years). HFMD outbreaks occur frequently in the Asia-Pacific region, and these outbreaks are associated with enormous healthcare and socioeconomic burden. There is currently no specific antiviral agent to treat HFMD and/or the severe complications that are frequently associated with the enterovirus of serotype EV71. Therefore, the development of a broadly effective and safe anti-enterovirus agent is an existential necessity. In this study, human single-chain antibodies (HuscFvs) specific to the EV71-internal capsid protein (VP4) were generated using phage display technology. VP4 specific-HuscFvs were linked to cell penetrating peptides to make them cell penetrable HuscFvs (transbodies), and readily accessible to the intracellular target. The transbodies, as well as the original HuscFvs that were tested, entered the enterovirus-infected cells, bound to intracellular VP4, and inhibited replication of EV71 across subgenotypes A, B, and C, and coxsackieviruses CVA16 and CVA6. The antibodies also enhanced the antiviral response of the virus-infected cells. Computerized simulation, indirect and competitive ELISAs, and experiments on cells infected with EV71 particles to which the VP4 and VP1-N-terminus were surface-exposed (i.e., A-particles that don’t require receptor binding for infection) indicated that the VP4 specific-antibodies inhibit virus replication by interfering with the VP4-N-terminus, which is important for membrane pore formation and virus genome release leading to less production of virus proteins, less infectious virions, and restoration of host innate immunity. The antibodies may inhibit polyprotein/intermediate protein processing and cause sterically strained configurations of the capsid pentamers, which impairs virus morphogenesis. These antibodies should be further investigated for application as a safe and broadly effective HFMD therapy.
Collapse
Affiliation(s)
- Siratcha Phanthong
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand
| | - Jaslan Densumite
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand
| | - Watee Seesuay
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand
| | - Jeeraphong Thanongsaksrikul
- Graduate Program in Biomedical Science, Faculty of Allied Health Sciences, Thammasat University, Bangkok, Thailand
| | - Salma Teimoori
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand
| | - Nitat Sookrung
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand.,Biomedical Research Incubator Unit, Department of Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yong Poovorawan
- Department of Pediatrics, Faculty of Medicine, Center of Excellence in Clinical Virology, Chulalongkorn University, Bangkok, Thailand
| | - Napa Onvimala
- Department of Medical Science, Ministry of Public Health, National Institute of Health, Nonthaburi, Thailand
| | - Ratigorn Guntapong
- Department of Medical Science, Ministry of Public Health, National Institute of Health, Nonthaburi, Thailand
| | - Kovit Pattanapanyasat
- Biomedical Research Incubator Unit, Department of Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand
| |
Collapse
|
9
|
San Martín C. Virus Maturation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1215:129-158. [DOI: 10.1007/978-3-030-14741-9_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
10
|
Corbic Ramljak I, Stanger J, Real-Hohn A, Dreier D, Wimmer L, Redlberger-Fritz M, Fischl W, Klingel K, Mihovilovic MD, Blaas D, Kowalski H. Cellular N-myristoyltransferases play a crucial picornavirus genus-specific role in viral assembly, virion maturation, and infectivity. PLoS Pathog 2018; 14:e1007203. [PMID: 30080883 PMCID: PMC6089459 DOI: 10.1371/journal.ppat.1007203] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/13/2018] [Accepted: 07/05/2018] [Indexed: 01/06/2023] Open
Abstract
In nearly all picornaviruses the precursor of the smallest capsid protein VP4 undergoes co-translational N-terminal myristoylation by host cell N-myristoyltransferases (NMTs). Curtailing this modification by mutation of the myristoylation signal in poliovirus has been shown to result in severe assembly defects and very little, if any, progeny virus production. Avoiding possible pleiotropic effects of such mutations, we here used pharmacological abrogation of myristoylation with the NMT inhibitor DDD85646, a pyrazole sulfonamide originally developed against trypanosomal NMT. Infection of HeLa cells with coxsackievirus B3 in the presence of this drug decreased VP0 acylation at least 100-fold, resulting in a defect both early and late in virus morphogenesis, which diminishes the yield of viral progeny by about 90%. Virus particles still produced consisted mainly of provirions containing RNA and uncleaved VP0 and, to a substantially lesser extent, of mature virions with cleaved VP0. This indicates an important role of myristoylation in the viral maturation cleavage. By electron microscopy, these RNA-filled particles were indistinguishable from virus produced under control conditions. Nevertheless, their specific infectivity decreased by about five hundred fold. Since host cell-attachment was not markedly impaired, their defect must lie in the inability to transfer their genomic RNA into the cytosol, likely at the level of endosomal pore formation. Strikingly, neither parechoviruses nor kobuviruses are affected by DDD85646, which appears to correlate with their native capsid containing only unprocessed VP0. Individual knockout of the genes encoding the two human NMT isozymes in haploid HAP1 cells further demonstrated the pivotal role for HsNMT1, with little contribution by HsNMT2, in the virus replication cycle. Our results also indicate that inhibition of NMT can possibly be exploited for controlling the infection by a wide spectrum of picornaviruses.
Collapse
Affiliation(s)
- Irena Corbic Ramljak
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Julia Stanger
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Antonio Real-Hohn
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Dominik Dreier
- Institute of Applied Synthetic Chemistry, TU Wien, Vienna, Austria
| | - Laurin Wimmer
- Institute of Applied Synthetic Chemistry, TU Wien, Vienna, Austria
| | | | - Wolfgang Fischl
- Haplogen GmbH, Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | | | - Dieter Blaas
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Heinrich Kowalski
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
11
|
Newman J, Asfor AS, Berryman S, Jackson T, Curry S, Tuthill TJ. The Cellular Chaperone Heat Shock Protein 90 Is Required for Foot-and-Mouth Disease Virus Capsid Precursor Processing and Assembly of Capsid Pentamers. J Virol 2018; 92:e01415-17. [PMID: 29212943 PMCID: PMC5809743 DOI: 10.1128/jvi.01415-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/29/2017] [Indexed: 12/14/2022] Open
Abstract
Productive picornavirus infection requires the hijacking of host cell pathways to aid with the different stages of virus entry, synthesis of the viral polyprotein, and viral genome replication. Many picornaviruses, including foot-and-mouth disease virus (FMDV), assemble capsids via the multimerization of several copies of a single capsid precursor protein into a pentameric subunit which further encapsidates the RNA. Pentamer formation is preceded by co- and posttranslational modification of the capsid precursor (P1-2A) by viral and cellular enzymes and the subsequent rearrangement of P1-2A into a structure amenable to pentamer formation. We have developed a cell-free system to study FMDV pentamer assembly using recombinantly expressed FMDV capsid precursor and 3C protease. Using this assay, we have shown that two structurally different inhibitors of the cellular chaperone heat shock protein 90 (hsp90) impeded FMDV capsid precursor processing and subsequent pentamer formation. Treatment of FMDV permissive cells with the hsp90 inhibitor prior to infection reduced the endpoint titer by more than 10-fold while not affecting the activity of a subgenomic replicon, indicating that translation and replication of viral RNA were unaffected by the drug.IMPORTANCE FMDV of the Picornaviridae family is a pathogen of huge economic importance to the livestock industry due to its effect on the restriction of livestock movement and necessary control measures required following an outbreak. The study of FMDV capsid assembly, and picornavirus capsid assembly more generally, has tended to be focused upon the formation of capsids from pentameric intermediates or the immediate cotranslational modification of the capsid precursor protein. Here, we describe a system to analyze the early stages of FMDV pentameric capsid intermediate assembly and demonstrate a novel requirement for the cellular chaperone hsp90 in the formation of these pentameric intermediates. We show the added complexity involved for this process to occur, which could be the basis for a novel antiviral control mechanism for FMDV.
Collapse
Affiliation(s)
- Joseph Newman
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Amin S Asfor
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | | | - Terry Jackson
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Stephen Curry
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | |
Collapse
|
12
|
Panjwani A, Asfor AS, Tuthill TJ. The conserved N-terminus of human rhinovirus capsid protein VP4 contains membrane pore-forming activity and is a target for neutralizing antibodies. J Gen Virol 2016; 97:3238-3242. [PMID: 27902347 PMCID: PMC5203672 DOI: 10.1099/jgv.0.000629] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human rhinovirus is the causative agent of the common cold and belongs to the non-enveloped picornavirus family. A trigger such as receptor binding or low pH initiates conformational changes in the capsid that allow the virus to attach to membranes and form a pore for the translocation of viral RNA into the cytoplasm. We previously showed that recombinant capsid protein VP4 was able to form membrane pores. In this study, we show the N-terminus but not C-terminus of VP4 formed pores with properties similar to full-length VP4 and consistent with the size required for transfer of RNA. Sera against the N-terminus but not C-terminus of VP4 were shown to neutralize virus infectivity. Together, this suggests that the N-terminus of VP4 is responsible for membrane activity. This study contributes to an improved understanding of the mechanisms for involvement of VP4 in entry and its potential as an antiviral target.
Collapse
Affiliation(s)
- Anusha Panjwani
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Amin S Asfor
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Tobias J Tuthill
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| |
Collapse
|
13
|
Triatoma virus recombinant VP4 protein induces membrane permeability through dynamic pores. J Virol 2015; 89:4645-54. [PMID: 25673713 DOI: 10.1128/jvi.00011-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
UNLABELLED In naked viruses, membrane breaching is a key step that must be performed for genome transfer into the target cells. Despite its importance, the mechanisms behind this process remain poorly understood. The small protein VP4, encoded by the genomes of most viruses of the order Picornavirales, has been shown to be involved in membrane alterations. Here we analyzed the permeabilization activity of the natively nonmyristoylated VP4 protein from triatoma virus (TrV), a virus belonging to the Dicistroviridae family within the Picornavirales order. The VP4 protein was produced as a C-terminal maltose binding protein (MBP) fusion to achieve its successful expression. This recombinant VP4 protein is able to produce membrane permeabilization in model membranes in a membrane composition-dependent manner. The induced permeability was also influenced by the pH, being greater at higher pH values. We demonstrate that the permeabilization activity elicited by the protein occurs through discrete pores that are inserted on the membrane. Sizing experiments using fluorescent dextrans, cryo-electron microscopy imaging, and other, additional techniques showed that recombinant VP4 forms heterogeneous proteolipidic pores rather than common proteinaceous channels. These results suggest that the VP4 protein may be involved in the membrane alterations required for genome transfer or cell entry steps during dicistrovirus infection. IMPORTANCE During viral infection, viruses need to overcome the membrane barrier in order to enter the cell and replicate their genome. In nonenveloped viruses membrane fusion is not possible, and hence, other mechanisms are implemented. Among other proteins, like the capsid-forming proteins and the proteins required for viral replication, several viruses of the order Picornaviridae contain a small protein called VP4 that has been shown to be involved in membrane alterations. Here we show that the triatoma virus VP4 protein is able to produce membrane permeabilization in model membranes by the formation of heterogeneous dynamic pores. These pores formed by VP4 may be involved in the genome transfer or cell entry steps during viral infection.
Collapse
|
14
|
Thibaut HJ, van der Linden L, Jiang P, Thys B, Canela MD, Aguado L, Rombaut B, Wimmer E, Paul A, Pérez-Pérez MJ, van Kuppeveld FJM, Neyts J. Binding of glutathione to enterovirus capsids is essential for virion morphogenesis. PLoS Pathog 2014; 10:e1004039. [PMID: 24722756 PMCID: PMC3983060 DOI: 10.1371/journal.ppat.1004039] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 02/17/2014] [Indexed: 11/18/2022] Open
Abstract
Enteroviruses (family of the Picornaviridae) cover a large group of medically important human pathogens for which no antiviral treatment is approved. Although these viruses have been extensively studied, some aspects of the viral life cycle, in particular morphogenesis, are yet poorly understood. We report the discovery of TP219 as a novel inhibitor of the replication of several enteroviruses, including coxsackievirus and poliovirus. We show that TP219 binds directly glutathione (GSH), thereby rapidly depleting intracellular GSH levels and that this interferes with virus morphogenesis without affecting viral RNA replication. The inhibitory effect on assembly was shown not to depend on an altered reducing environment. Using TP219, we show that GSH is an essential stabilizing cofactor during the transition of protomeric particles into pentameric particles. Sequential passaging of coxsackievirus B3 in the presence of low GSH-levels selected for GSH-independent mutants that harbored a surface-exposed methionine in VP1 at the interface between two protomers. In line with this observation, enteroviruses that already contained this surface-exposed methionine, such as EV71, did not rely on GSH for virus morphogenesis. Biochemical and microscopical analysis provided strong evidence for a direct interaction between GSH and wildtype VP1 and a role for this interaction in localizing assembly intermediates to replication sites. Consistently, the interaction between GSH and mutant VP1 was abolished resulting in a relocalization of the assembly intermediates to replication sites independent from GSH. This study thus reveals GSH as a novel stabilizing host factor essential for the production of infectious enterovirus progeny and provides new insights into the poorly understood process of morphogenesis. Enteroviruses contain many significant human pathogens, including poliovirus, enterovirus 71, coxsackieviruses and rhinoviruses. Most enterovirus infections subside mild or asymptomatically, but may also result in severe morbidity and mortality. Here, we report on the mechanism of antiviral action of a small molecule, TP219, as an inhibitor of enterovirus morphogenesis. Morphogenesis represents an important stage at the end of the virus replication cycle and requires multiple steps, of which some are only poorly understood. Better understanding of this process holds much potential to facilitate the development of new therapies to combat enterovirus infections. We demonstrate that TP219 rapidly depletes intracellular glutathione (GSH) by covalently binding free GSH resulting in the inhibition of virus morphogenesis without affecting viral RNA replication. We discovered that GSH directly interacts with viral capsid precursors and mature virions and that this interaction is required for the formation of an assembly intermediate (pentameric particles) and consequently infectious progeny. Remarkably, enteroviruses that were capable of replicating in the absence of GSH contained a surface-exposed methionine at the protomeric interface. We propose that GSH is an essential and stabilizing host factor during morphogenesis and that this stabilization is a prerequisite for a functional encapsidation of progeny viral RNA.
Collapse
Affiliation(s)
- Hendrik Jan Thibaut
- Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Lonneke van der Linden
- Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
- Department Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Ping Jiang
- Department of Molecular Genetics and Microbiology, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Bert Thys
- Department of Pharmaceutical Biotechnology & Molecular Biology, Vrije Universiteit Brussel, Brussel, Belgium
| | | | - Leire Aguado
- Instituto de Química Médica (IQM-CSIC), Madrid, Spain
| | - Bart Rombaut
- Department of Pharmaceutical Biotechnology & Molecular Biology, Vrije Universiteit Brussel, Brussel, Belgium
| | - Eckard Wimmer
- Department of Molecular Genetics and Microbiology, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Aniko Paul
- Department of Molecular Genetics and Microbiology, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | | | - Frank J. M. van Kuppeveld
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Johan Neyts
- Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
15
|
Quantification of the dynamics of enterovirus 71 infection by experimental-mathematical investigation. J Virol 2012; 87:701-5. [PMID: 23097444 DOI: 10.1128/jvi.01453-12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterovirus 71 (EV71) is the causative agent of hand-foot-and-mouth disease and can trigger neurological disorders. EV71 outbreaks are a major public health concern in Asia-Pacific countries. By performing experimental-mathematical investigation, we demonstrate here that viral productivity and transmissibility but not viral cytotoxicity are drastically different among EV71 strains and can be associated with their epidemiological backgrounds. This is the first report demonstrating the dynamics of nonenveloped virus replication in cell culture using mathematical modeling.
Collapse
|
16
|
de Vries JS, Andriotis VME, Wu AJ, Rathjen JP. Tomato Pto encodes a functional N-myristoylation motif that is required for signal transduction in Nicotiana benthamiana. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2006; 45:31-45. [PMID: 16367952 DOI: 10.1111/j.1365-313x.2005.02590.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Pto kinase of tomato (Lycopersicon esculentum) confers resistance to bacterial speck disease caused by Pseudomonas syringae pv. tomato expressing avrPto or avrPtoB. Pto interacts directly with these type-III secreted effectors, leading to induction of defence responses including the hypersensitive response (HR). Signalling by Pto requires the nucleotide-binding site-leucine-rich repeat (NBS-LRR) protein Prf. Little is known of how Pto is controlled prior to or during stimulation, although kinase activity is required for Avr-dependent activation. Here we demonstrate a role for the N-terminus in signalling by Pto. N-terminal residues outside the kinase domain were required for induction of the HR in Nicotiana benthamiana. The N-terminus also contributed to both AvrPto-binding and phosphorylation abilities. Pto residues 1-10 comprise a consensus motif for covalent attachment of myristate, a hydrophobic 14-carbon saturated fatty acid, to the Gly-2 residue. Several lines of evidence indicate that this motif is important for Pto function. A heterologous N-myristoylation motif complemented N-terminal deletion mutants of Pto for Prf-dependent signalling. Signalling by wild-type and mutant forms of Pto was strictly dependent on the Gly-2 residue. The N-myristoylation motif of Pto complemented the cognate motif of AvrPto for avirulence function and membrane association. Furthermore, Pto was myristoylated in vivo dependent on the presence of Gly-2. The subcellular localization of Pto was independent of N-myristoylation, indicating that N-myristoylation is required for some function other than membrane affinity. Consistent with this idea, AvrPtoB was also found to be a soluble protein. The data indicate an important role(s) for the myristoylated N-terminus in Pto signalling.
Collapse
Affiliation(s)
- Jeroen S de Vries
- The Sainsbury Laboratory, John Innes Centre, Colney Lane, Norwich, NR4 7UH, UK
| | | | | | | |
Collapse
|
17
|
Shiomi H, Urasawa T, Urasawa S, Kobayashi N, Abe S, Taniguchi K. Isolation and characterisation of poliovirus mutants resistant to heating at 50°c for 30 min. J Med Virol 2004; 74:484-91. [PMID: 15368512 DOI: 10.1002/jmv.20202] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Poliovirus is heat-labile; on heating at 50 degrees Celsius for 30 min its infectivity decreases drastically and its antigenicity reverts from N to H. However, mutants resistant to heating at 50 degrees Celsius for 30 min from the Sabin 1 and 2 viruses were isolated by repeating the process of incubation of the virus stock at 50 degrees Celsius for 30 min and multiplication of the remaining virus in a cell culture. The isolated mutants were stable genetically, and maintained the rct and d markers of the parent virus. On electron microscopical examination, the mutants were observed to retain the intact morphology after being heated at 50 degrees Celsius for 30 min, while the parent virus was converted to empty particles devoid of RNA under the same conditions. On determination of the nucleotide sequence of the P1 region, a single nucleotide sequence substitution was detected at nucleotide no. 2741, resulting in an amino acid change from valine to alanine at the 87th position of VP1. This amino acid might be associated with the heat-resistance of the mutants. Furthermore, it was found that the thermostable mutants obtained in this study, which are resistant to "high" temperature (50 degrees Celsius) for a short time (30 min), were not stable against heating at the ambient temperature (37 degrees Celsius) for a long time (5 or 7 days). This suggests that the inactivation at high temperature for a short time and that at ambient temperature for a long time involve different mechanisms.
Collapse
Affiliation(s)
- Hiroshi Shiomi
- Department of Hygiene, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
18
|
Tosteson MT, Wang H, Naumov A, Chow M. Poliovirus binding to its receptor in lipid bilayers results in particle-specific, temperature-sensitive channels. J Gen Virol 2004; 85:1581-1589. [PMID: 15166442 DOI: 10.1099/vir.0.19745-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Poliovirus (PV) infection starts with binding to its receptor (PVR), followed by a receptor-aided, temperature-sensitive conformational change of the infectious particle (sedimenting at 160S) to a particle which sediments at 135S. Reported in this communication is the successful incorporation into lipid bilayers of two forms of the receptor: the full-length human receptor and a modified clone in which the extracellular domains of the receptor were fused to a glycosylphosphatidylinositol tail. Addition of virus (160S) to receptor-containing bilayers leads to channel formation, whereas no channels were observed when the receptor-modified viral particle (135S) was added. Increasing the temperature from 21 to 31 degrees C led to a 10-fold increase in the magnitude of the single channel conductance, which can be interpreted as a conformational change in the channel structure. A mutant PV with an amino acid change in VP4 (one of the coat proteins) which is defective in genome uncoating failed to produce channels, suggesting that VP4 might be involved in the channel architecture. These studies provide the first electrophysiological characterization of the interactions between poliovirus and its receptor incorporated into a lipid bilayer membrane. Furthermore, they form the foundation for future studies aiming at defining the molecular architecture of the virus-receptor complex.
Collapse
Affiliation(s)
- Magdalena T Tosteson
- Department of Cell Biology, Laboratory for Membrane Transport, Harvard Medical School, One Kendall Square, Building 600, Third Floor, Cambridge, MA 02139, USA
| | - Hong Wang
- Department of Microbiology and Immunology, University of Arkansas, Medical School, Little Rock, AR 72205, USA
| | - Anatoli Naumov
- Department of Microbiology and Immunology, University of Arkansas, Medical School, Little Rock, AR 72205, USA
| | - Marie Chow
- Department of Microbiology and Immunology, University of Arkansas, Medical School, Little Rock, AR 72205, USA
| |
Collapse
|
19
|
Abstract
Assembly of the human immunodeficiency virus type 1 (HIV-1) first occurs on the plasma membrane of host cells where binding is driven by strong electrostatic interactions between the N-terminal matrix (MA) domain of the structural precursor polyprotein, Gag, and the membrane. MA is also myristylated, but the exact role this modification plays is not clear. In this study, we compared the protein oligomerization and membrane binding properties of Myr(+) and Myr(-) Gag(MA) expressed in COS-1 cells. Sedimentation studies in solution showed that both the myristylated Gag precursor and the mature MA product were detected in larger complexes than their unmyristylated counterparts, and the myristylated MA protein bound liposomes with approximately 3-fold greater affinity than unmyristylated MA. Aromatic residues near the N-terminal region of the MA protein were more accessible to chymotrypsin in the unmyristylated form and, consistent with this, an epitope in the N-terminal region was more exposed. Moreover, the cyclophilin binding site in the CA domain downstream of MA was more accessible in the unmyristylated Gag protein, while the Tsg101 binding site in the C-terminal region was equally available in the unmyristylated and myristylated Gag proteins. Taken together, our results suggest that myristylation promotes assembly by inducing conformational changes and facilitating MA multimerization. This observation offers a novel role for myristylation.
Collapse
Affiliation(s)
- Fadila Bouamr
- Department of Molecular Genetics & Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | | | | |
Collapse
|
20
|
Fry EE, Knowles NJ, Newman JWI, Wilsden G, Rao Z, King AMQ, Stuart DI. Crystal structure of Swine vesicular disease virus and implications for host adaptation. J Virol 2003; 77:5475-86. [PMID: 12692248 PMCID: PMC153985 DOI: 10.1128/jvi.77.9.5475-5486.2003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Swine vesicular disease virus (SVDV) is an Enterovirus of the family Picornaviridae that causes symptoms indistinguishable from those of foot-and-mouth disease virus. Phylogenetic studies suggest that it is a recently evolved genetic sublineage of the important human pathogen coxsackievirus B5 (CBV5), and in agreement with this, it has been shown to utilize the coxsackie and adenovirus receptor (CAR) for cell entry. The 3.0-A crystal structure of strain UK/27/72 SVDV (highly virulent) reveals the expected similarity in core structure to those of other picornaviruses, showing most similarity to the closest available structure to CBV5, that of coxsackievirus B3 (CBV3). Features that help to cement together and rigidify the protein subunits are extended in this virus, perhaps explaining its extreme tolerance of environmental factors. Using the large number of capsid sequences available for both SVDV and CBV5, we have mapped the amino acid substitutions that may have occurred during the supposed adaptation of SVDV to a new host onto the structure of SVDV and a model of the SVDV/CAR complex generated by reference to the cryo-electron microscopy-visualized complex of CBV3 and CAR. The changes fall into three clusters as follows: one lines the fivefold pore, a second maps to the CAR-binding site and partially overlaps the site for decay accelerating factor (DAF) to bind to echovirus 7 (ECHO7), and the third lies close to the fivefold axis, where the low-density lipoprotein receptor binds to the minor group of rhinoviruses. Later changes in SVDV (post-1971) map to the first two clusters and may, by optimizing recognition of a pig CAR and/or DAF homologue, have improved the adaptation of the virus to pigs.
Collapse
Affiliation(s)
- Elizabeth E Fry
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford OX3 7BN, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
21
|
Danthi P, Tosteson M, Li QH, Chow M. Genome delivery and ion channel properties are altered in VP4 mutants of poliovirus. J Virol 2003; 77:5266-74. [PMID: 12692228 PMCID: PMC153979 DOI: 10.1128/jvi.77.9.5266-5274.2003] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
During entry into host cells, poliovirus undergoes a receptor-mediated conformational transition to form 135S particles with irreversible exposure of VP4 capsid sequences and VP1 N termini. To understand the role of VP4 during virus entry, the fate of VP4 during infection by site-specific mutants at threonine-28 of VP4 (4028T) was compared with that of the parental Mahoney type 1 virus. Three virus mutants were studied: the entry-defective, nonviable mutant 4028T.G and the viable mutants 4028T.S and 4028T.V, in which residue threonine-28 was changed to glycine, serine, and valine, respectively. We show that mutant and wild-type (WT) VP4 proteins are localized to cellular membranes after the 135S conformational transition. Both WT and viable 4028T mutant particles interact with lipid bilayers to form ion channels, whereas the entry-defective 4028T.G particles do not. In addition, the electrical properties of the channels induced by the mutant viruses are different from each other and from those of WT Mahoney and Sabin type 3 viruses. Finally, uncoating and/or cytoplasmic delivery of the viral genome is altered in the 4028T mutants: the 4028T.G lethal mutant does not release its genome into the cytoplasm, and genome delivery is slower during infection by mutant 4028T.V 135S particles than by mutant 4028T.S or WT 135S particles. The distinctive electrical characteristics of the different 4028T mutant channels indicate that VP4 sequences might form part of the channel structure. The different entry phenotypes of these VP4 mutants suggest that the ion channels may be related to VP4's role during genome uncoating and/or delivery.
Collapse
Affiliation(s)
- Pranav Danthi
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock 72205, USA
| | | | | | | |
Collapse
|
22
|
Mannová P, Liebl D, Krauzewicz N, Fejtová A, Štokrová J, Palková Z, Griffin BE, Forstová J. Analysis of mouse polyomavirus mutants with lesions in the minor capsid proteins. J Gen Virol 2002; 83:2309-2319. [PMID: 12185287 DOI: 10.1099/0022-1317-83-9-2309] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Polyomavirus mutants E, Q and H, expressing non-myristylated VP2, were generated by replacing the N-terminal glycine residue with glutamic acid, glutamine or histidine, respectively. Viruses mutated in either VP2 or VP3 translation initiation codons were also prepared. All mutated genomes, when transfected into murine host cells, gave rise to viral particles. Infectivity of VP2- and VP3- viruses, as measured by the number of cells expressing viral antigens, was dramatically diminished, indicative of defects in the early stages of infection. In contrast, the absence of a myristyl moiety on VP2 did not substantially affect the early steps of virus infection. No differences in numbers of cells expressing early or late viral antigens were observed between wild-type (wt) and E or Q myr- viruses during the course of a life cycle. Furthermore, no delay in virus DNA replication was detected. However, when cells were left for longer in culture, the number of infected cells, measured by typical virus bursts, was much lower when mutant rather than wt genomes were used. In situ, cell fractionation studies revealed differences in the interaction of viral particles with host cell structures. The infectivity of mutants was affected not only by loss of the myristyl group on VP2, but also, and to a greater extent, by alterations of the N-terminal amino acid composition.
Collapse
Affiliation(s)
- Petra Mannová
- Department of Genetics and Microbiology, Charles University in Prague, Viničná 5, 128 44 Prague 2, Czech Republic1
| | - David Liebl
- Department of Genetics and Microbiology, Charles University in Prague, Viničná 5, 128 44 Prague 2, Czech Republic1
| | - Nina Krauzewicz
- Department of Virology, Royal Postgraduate Medical School, Hammersmith Hospital, London W12 0NN, UK2
| | - Anna Fejtová
- Department of Genetics and Microbiology, Charles University in Prague, Viničná 5, 128 44 Prague 2, Czech Republic1
| | - Jitka Štokrová
- Institute of Molecular Genetics, Czech Academy of Sciences, Flemingovo n. 2, 166 37 Prague 6, Czech Republic3
| | - Zdena Palková
- Department of Genetics and Microbiology, Charles University in Prague, Viničná 5, 128 44 Prague 2, Czech Republic1
| | - Beverly E Griffin
- Department of Virology, Royal Postgraduate Medical School, Hammersmith Hospital, London W12 0NN, UK2
| | - Jitka Forstová
- Department of Genetics and Microbiology, Charles University in Prague, Viničná 5, 128 44 Prague 2, Czech Republic1
| |
Collapse
|
23
|
Slifka MK, Pagarigan R, Mena I, Feuer R, Whitton JL. Using recombinant coxsackievirus B3 to evaluate the induction and protective efficacy of CD8+ T cells during picornavirus infection. J Virol 2001; 75:2377-87. [PMID: 11160741 PMCID: PMC114821 DOI: 10.1128/jvi.75.5.2377-2387.2001] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Coxsackievirus B3 (CVB3) is a common human pathogen that has been associated with serious diseases including myocarditis and pancreatitis. To better understand the effect of cytotoxic T-lymphocyte (CTL) responses in controlling CVB3 infection, we have inserted well-characterized CTL epitopes into the CVB3 genome. Constructs were made by placing the epitope of interest upstream of the open reading frame encoding the CVB3 polyprotein, separated by a poly-glycine linker and an artificial 3Cpro/3CDpro cleavage site. This strategy results in the foreign protein being translated at the amino- terminus of the viral polyprotein, from which it is cleaved prior to viral assembly. In this study, we cloned major histocompatibility complex class I-restricted CTL epitopes from lymphocytic choriomeningitis virus (LCMV) into recombinant CVB3 (rCVB3). In vitro, rCVB3 growth kinetics showed a 1- to 2-h lag period before exponential growth was initiated, and peak titers were approximately 1 log unit lower than for wild-type virus. rCVB3 replicated to high titers in vivo and caused severe pancreatitis but minimal myocarditis. Despite the high virus titers, rCVB3 infection of naive mice failed to induce a strong CD8+ T-cell response to the encoded epitope; this has implications for the proposed role of "cross-priming" during virus infection and for the utility of recombinant picornaviruses as vaccine vectors. In contrast, rCVB3 infection of LCMV-immune mice resulted in direct ex vivo cytotoxic activity against target cells coated with the epitope peptide, demonstrating that the rCVB3-encoded LCMV-specific epitope was expressed and presented in vivo. The preexisting CD8+ memory T cells could limit rCVB replication; compared to naive mice, infection of LCMV-immune mice with rCVB3 resulted in approximately 50-fold-lower virus titers in the heart and approximately 6-fold-lower virus titers in the pancreas. Although the inserted CTL epitope was retained by rCVB3 through several passages in tissue culture, it was lost in an organ-specific manner in vivo; a substantial proportion of viruses from the pancreas retained the insert, compared to only 0 to 1.8% of myocardial viruses. Together, these results show that expression of heterologous viral proteins by recombinant CVB3 provides a useful model for determining the mechanisms underlying the immune response to this viral pathogen.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral
- CD8-Positive T-Lymphocytes/immunology
- Coxsackievirus Infections/immunology
- Coxsackievirus Infections/virology
- Enterovirus B, Human/genetics
- Enterovirus B, Human/growth & development
- Enterovirus B, Human/immunology
- Enterovirus B, Human/pathogenicity
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Glycoproteins/immunology
- Glycoproteins/metabolism
- Humans
- Immunologic Memory
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Molecular Sequence Data
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Recombination, Genetic
- Transfection
- Viral Proteins
- Virus Replication
Collapse
Affiliation(s)
- M K Slifka
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
24
|
Zhang Z, Greene B, Thuman-Commike PA, Jakana J, Prevelige PE, King J, Chiu W. Visualization of the maturation transition in bacteriophage P22 by electron cryomicroscopy. J Mol Biol 2000; 297:615-26. [PMID: 10731416 DOI: 10.1006/jmbi.2000.3601] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Large-scale conformational transitions are involved in the life-cycle of many types of virus. The dsDNA phages, herpesviruses, and adenoviruses must undergo a maturation transition in the course of DNA packaging to convert a scaffolding-containing precursor capsid to the DNA-containing mature virion. This conformational transition converts the procapsid, which is smaller, rounder, and displays a distinctive skewing of the hexameric capsomeres, to the mature virion, which is larger and more angular, with regular hexons. We have used electron cryomicroscopy and image reconstruction to obtain 15 A structures of both bacteriophage P22 procapsids and mature phage. The maturation transition from the procapsid to the phage results in several changes in both the conformations of the individual coat protein subunits and the interactions between neighboring subunits. The most extensive conformational transformation among these is the outward movement of the trimer clusters present at all strict and local 3-fold axes on the procapsid inner surface. As the trimer tips are the sites of scaffolding binding, this helps to explain the role of scaffolding protein in regulating assembly and maturation. We also observe DNA within the capsid packed in a manner consistent with the spool model. These structures allow us to suggest how the binding interactions of scaffolding and DNA with the coat shell may act to control the packaging of the DNA into the expanding procapsids.
Collapse
Affiliation(s)
- Z Zhang
- Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Probst C, Jecht M, Gauss-Müller V. Intrinsic signals for the assembly of hepatitis A virus particles. Role of structural proteins VP4 and 2A. J Biol Chem 1999; 274:4527-31. [PMID: 9988685 DOI: 10.1074/jbc.274.8.4527] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Capsid assembly is the final event of virus replication, and its understanding is pivotal for the design of empty capsid-based recombinant vaccines and drug delivery systems. Although the capsid structure of several members of the picornavirus family has been elucidated, little is known about the structural elements governing the assembly process that is tightly associated with proteolytic processing of the viral polyprotein. Among the picornaviruses, hepatitis A virus (HAV) is unique in that it contains VP1-2A as a structural component and the small structural protein VP4, which argues for an assembly pathway different from that proposed for other picornaviruses. Using a recombinant system we show here that proteolytic processing of the HAV capsid proteins' precursor P1-2A is independent of the terminal domains 2A and VP4 of the substrate. However, both terminal domains play distinct roles in the assembly of viral particles. 2A as part of P1-2A is a primary signal for the assembly of pentameric structures which only further aggregate to empty viral capsids when VP4 is present as the N terminus of the precursor. Particle formation in the hepatovirus genus is thus regulated by two intrinsic signals that are distinct from those described for other picornaviruses.
Collapse
Affiliation(s)
- C Probst
- Institute of Medical Microbiology and Hygiene, Medical University of Lübeck, 23538 Lübeck, Germany
| | | | | |
Collapse
|
26
|
Abstract
We have been exploring the mechanism of action of 5-(3,4-dichlorophenyl) methylhydantoin (hydantoin), an antiviral drug that inhibits the replication of poliovirus in culture. By varying the time of drug addition to infected cells, we found that the drug acts at a stage which is late in the replication cycle and subsequent to the step inhibited by guanidine. Furthermore, we detected normal levels of full-length plus-strand virion RNA in hydantoin-treated cultures. A new assembly intermediate in addition to the expected assembly intermediates was detected in drug-treated cultures. This intermediate has properties consistent with that of a packaging intermediate. Drug-resistant mutants were readily isolated. Sequence analysis of three independent drug-resistant mutants identified amino acid substitutions in the 2C coding region. Reconstruction by site-directed mutagenesis confirmed that these single mutations were sufficient to confer drug resistance. Taken together, these data suggest that the poliovirus 2C region is involved in virus encapsidation and that hydantoin inhibits this stage of replication.
Collapse
Affiliation(s)
- L M Vance
- Infectious Diseases Research, Lilly Research Laboratories, Indianapolis, Indiana 46285, USA
| | | | | | | |
Collapse
|
27
|
Conte MR, Klikova M, Hunter E, Ruml T, Matthews S. The three-dimensional solution structure of the matrix protein from the type D retrovirus, the Mason-Pfizer monkey virus, and implications for the morphology of retroviral assembly. EMBO J 1997; 16:5819-26. [PMID: 9312040 PMCID: PMC1170213 DOI: 10.1093/emboj/16.19.5819] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The Mason-Pfizer monkey virus (M-PMV) is the prototype of the type D retroviruses. In type B and D retroviruses, the Gag protein pre-assembles before association with the membrane, whereas in type C retroviruses (lentiviruses, BLV/HTLV group) Gag is targeted efficiently to the plasma membrane, where the particle formation occurs. The N-terminal domain of Gag, the matrix protein (MA), plays a critical role in determining this morphogenic difference. We have determined the three-dimensional solution structure of the M-PMV MA by heteronuclear nuclear magnetic resonance. The protein contains four alpha-helices that are structurally similar to the known type C MA structures. This similarity implies possible common assembly units and membrane-binding mechanisms for type C and B/D retroviruses. In addition to this, the interpretation of mutagenesis data has enabled us to identify, for the first time, the structural basis of a putative intracellular targeting motif.
Collapse
Affiliation(s)
- M R Conte
- Department of Biochemistry, Imperial College of Science, Technology and Medicine, University of London, London SW7 2AY, UK
| | | | | | | | | |
Collapse
|
28
|
Tang S, van Rij R, Silvera D, Andino R. Toward a poliovirus-based simian immunodeficiency virus vaccine: correlation between genetic stability and immunogenicity. J Virol 1997; 71:7841-50. [PMID: 9311872 PMCID: PMC192139 DOI: 10.1128/jvi.71.10.7841-7850.1997] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recombinant polioviruses expressing foreign antigens may provide a convenient vaccine vector to engender mucosal immunity. Replication-competent chimeric viruses can be constructed by fusing foreign antigenic sequences to several positions within the poliovirus polyprotein. Artificial cleavage sites ensure appropriate proteolytic processing of the recombinant polyprotein, yielding mature and functional viral proteins. To study the effect of the position of insertion, two different recombinant polioviruses were examined. A small amino-terminus insertion delayed virus maturation and produced a thermosensitive particle. In contrast, insertion at the junction between the P1 and P2 regions yielded a chimeric poliovirus that replicated like the wild type. Eight different chimeras were constructed by inserting simian immunodeficiency virus (SIV) sequences at the P1/P2 junction. All recombinant viruses replicated with near-wild-type efficiency in tissue culture cells and expressed high levels of the SIV antigens. One of the inserted fragments corresponding to gp41 envelope protein was N-glycosylated but was not secreted. Inserted sequences were only partially retained after few rounds of replication in HeLa cells. This problem could be remedied to some extent by altering the sequences flanking the insertion point. Reducing the homology of the direct repeats by 37% decrease the propensity of the recombinant viruses to delete the insert. To determine the immunogenic potential of the recombinants, mice susceptible to poliovirus infection were inoculated intraperitoneally. The antibody titers elicited against Gag p17 depended on the viral doses and the number of inoculations. In addition, recombinants which display higher genetic stability were more effective in inducing an immune response against the SIV antigens, and inoculation with a mix of recombinants carrying different SIV antigens (a cocktail of recombinants) elicited humoral responses against each of the individual SIV sequences.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/biosynthesis
- Antigens, Viral/immunology
- Base Sequence
- Cloning, Molecular
- Escherichia coli
- HeLa Cells
- Humans
- Mice
- Mice, Transgenic
- Molecular Sequence Data
- Mutagenesis, Insertional
- Poliovirus
- Poliovirus Vaccine, Inactivated
- Polymerase Chain Reaction
- Receptors, Virus/biosynthesis
- Receptors, Virus/genetics
- Receptors, Virus/physiology
- Recombinant Fusion Proteins/biosynthesis
- Recombination, Genetic
- Repetitive Sequences, Nucleic Acid
- Restriction Mapping
- SAIDS Vaccines
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
- Simian Immunodeficiency Virus/immunology
- Simian Immunodeficiency Virus/physiology
- Vaccines, Synthetic
- Viral Plaque Assay
- Viral Proteins/biosynthesis
- Virion/immunology
- Virion/physiology
Collapse
Affiliation(s)
- S Tang
- Department of Microbiology and Immunology, University of California, San Francisco 94143-0414, USA
| | | | | | | |
Collapse
|
29
|
Affiliation(s)
- J K Muckelbauer
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
30
|
Lentz KN, Smith AD, Geisler SC, Cox S, Buontempo P, Skelton A, DeMartino J, Rozhon E, Schwartz J, Girijavallabhan V, O'Connell J, Arnold E. Structure of poliovirus type 2 Lansing complexed with antiviral agent SCH48973: comparison of the structural and biological properties of three poliovirus serotypes. Structure 1997; 5:961-78. [PMID: 9261087 DOI: 10.1016/s0969-2126(97)00249-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Polioviruses are human pathogens and the causative agents of poliomyelitis. Polioviruses are icosahedral single-stranded RNA viruses, which belong to the picornavirus family, and occur as three distinct serotypes. All three serotypes of poliovirus can infect primates, but only type 2 can infect mice. The crystal structures of a type 1 and a type 3 poliovirus are already known. Structural studies of poliovirus type 2 Lansing (PV2L) were initiated to try to enhance our understanding of the differences in host range specificity, antigenicity and receptor binding among the three serotypes of poliovirus. RESULTS The crystal structure of the mouse neurovirulent PV2L complexed with a potent antiviral agent, SCH48973, was determined at 2.9 A resolution. Structural differences among the three poliovirus serotypes occur primarily in the loop regions of the viral coat proteins (VPs), most notably in the loops of VP1 that cluster near the fivefold axes of the capsid, where the BC loop of PV2L is disordered. Unlike other known structures of enteroviruses, the entire polypeptide chain of PV2L VP4 is visible in the electron density and RNA bases are observed stacking with conserved aromatic residues (Tyr4020 and Phe4046) of VP4. The broad-spectrum antiviral agent SCH48973 is observed binding in a pocket within the beta-barrel of VP1, in approximately the same location that natural 'pocket factors' bind to polioviruses. SCH48973 forms predominantly hydrophobic interactions with the pocket residues. CONCLUSIONS Some of the conformational changes required for infectivity and involved in the control of capsid stability and neurovirulence in mice may occur in the vicinity of the fivefold axis of the poliovirus, where there are significant structural differences among the three poliovirus serotypes in the surface exposed loops of VP1 (BC, DE, and HI). A surface depression is located at the fivefold axis of PV2L that is not present in the other two poliovirus serotypes. The observed interaction of RNA with VP4 supports the observation that loss of VP4 ultimately leads to the loss of viral RNA. A model is proposed that suggests dual involvement of the virion fivefold and pseudo-threefold axes in receptor-mediated initiation of infection by picornaviruses.
Collapse
Affiliation(s)
- K N Lentz
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Tosteson MT, Chow M. Characterization of the ion channels formed by poliovirus in planar lipid membranes. J Virol 1997; 71:507-11. [PMID: 8985378 PMCID: PMC191079 DOI: 10.1128/jvi.71.1.507-511.1997] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The steps in poliovirus infection leading to viral entry and uncoating are not well understood. Current evidence suggests that the virus first binds to a plasma membrane-bound receptor present in viable cells, leading to a conformational rearrangement of the viral proteins such that the virus crosses the membrane and releases the genomic RNA. The studies described in this report were undertaken to determine if poliovirus (160S) as well as one of the subviral particles (135S) could interact with membranes lacking poliovirus receptors in an effort to begin to understand the process of uncoating of the virus. We report that both forms of viral particles, 160S and 135S, interact with lipid membranes and induce the formation of ion-permeable channels in a manner that does not require acid pH. The channels induced by the viral particles 160S have a voltage-dependent conductance which depends on the ionic composition of the medium. Our findings raise the possibility that viral entry into cells may be mediated by direct interaction of viral surface proteins with membrane lipids.
Collapse
Affiliation(s)
- M T Tosteson
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
32
|
Ansardi DC, Porter DC, Anderson MJ, Morrow CD. Poliovirus Assembly and Encapsidation of Genomic RNA. Adv Virus Res 1996. [DOI: 10.1016/s0065-3527(08)60069-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
33
|
Ramsingh AI, Caggana M, Ronstrom S. Genetic mapping of the determinants of plaque morphology of coxsackievirus B4. Arch Virol 1995; 140:2215-26. [PMID: 8572942 DOI: 10.1007/bf01323241] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The genetic determinants of plaque size of two variants of coxsackie-virus B4, CB4-P and CB4-V, were identified using a panel of recombinant, chimeric viruses. When grown in monkey kidney cells, CB4-V yielded large plaques with an average size of 1.0 cm while CB4-P yielded small plaques with an average size of 0.4 cm. Two genetic domains, the 5' untranslated region and the VP4 gene sequence, independently influenced plaque size. Recombinant viruses containing the CB4-P genetic background with point mutations in either the VP1 or VP2 coding sequences had small plaque phenotypes. However, two additional chimerics containing the CB4-P genetic background with either a point mutation in the VP4 sequence or four substitutions in the 5' untranslated region, had large plaque phenotypes. Plaque size correlated with growth kinetics under single-step conditions. Large-plaque variants replicated to higher titers than small-plaque variants. Comparison of the growth kinetics of the recombinant viruses revealed some differences in viral replication. These data suggest that both the 5' untranslated region and arg-16 of VP4 influence viral replication but at different stages of the replication cycle.
Collapse
Affiliation(s)
- A I Ramsingh
- Wadsworth Center for Laboratories and Research, New York State Department of Health, Albany, USA
| | | | | |
Collapse
|
34
|
Muckelbauer JK, Kremer M, Minor I, Diana G, Dutko FJ, Groarke J, Pevear DC, Rossmann MG. The structure of coxsackievirus B3 at 3.5 A resolution. Structure 1995; 3:653-67. [PMID: 8591043 DOI: 10.1016/s0969-2126(01)00201-5] [Citation(s) in RCA: 198] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Group B coxsackieviruses (CVBs) are etiologic agents of a number of human diseases that range in severity from asymptomatic to lethal infections. They are small, single-stranded RNA icosahedral viruses that belong to the enterovirus genus of the picornavirus family. Structural studies were initiated in light of the information available on the cellular receptors for this virus and to assist in the design of antiviral capsid-binding compounds for the CVBs. RESULTS The structure of coxsackievirus B3 (CVB3) has been solved to a resolution of 3.5 A. The beta-sandwich structure of the viral capsid proteins VP1, VP2 and VP3 is conserved between CVB3 and other picornaviruses. Structural differences between CVB3 and other enteroviruses and rhinoviruses are located primarily on the viral surface. The hydrophobic pocket of the VP1 beta-sandwich is occupied by a pocket factor, modeled as a C16 fatty acid. An additional study has shown that the pocket factor can be displaced by an antiviral compound. Myristate was observed covalently linked to the N terminus of VP4. Density consistent with the presence of ions was observed on the icosahedral threefold and fivefold axes. CONCLUSIONS The canyon and twofold depression, major surface depressions, are predicted to be the primary and secondary receptor-binding sites on CVB3, respectively. Neutralizing immunogenic sites are predicted to lie on the extreme surfaces of the capsid at sites that lack amino acid sequence conservation among the CVBs. The ions located on the icosahedral threefold and fivefold axes together with the pocket factor may contribute to the pH stability of the coxsackieviruses.
Collapse
Affiliation(s)
- J K Muckelbauer
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Chow M, Moscufo N. Myristoyl modification of viral proteins: assays to assess functional roles. Methods Enzymol 1995; 250:495-509. [PMID: 7651174 DOI: 10.1016/0076-6879(95)50093-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Characterization of the effects of myristoylation on poliovirus function has largely depended on the availability of methods previously characterized to study various aspects of virus biology. Those methods are described here to provide specific examples of how they may be used to analyze the myristoylation mutants and to illustrate general approaches. It is possible that the poliovirus protocols may be directly transferable with little or no modification to analyze other systems. However, it is more likely that the application of specific methods, which have already been developed and characterized for the systems of interest and which utilize the strengths and reagents unique to those experimental systems, may be more efficient and informative.
Collapse
Affiliation(s)
- M Chow
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock 72205, USA
| | | |
Collapse
|
36
|
Spearman P, Wang JJ, Vander Heyden N, Ratner L. Identification of human immunodeficiency virus type 1 Gag protein domains essential to membrane binding and particle assembly. J Virol 1994; 68:3232-42. [PMID: 8151785 PMCID: PMC236814 DOI: 10.1128/jvi.68.5.3232-3242.1994] [Citation(s) in RCA: 222] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Assembly of human immunodeficiency virus type 1 (HIV-1) particles occurs at the plasma membrane of infected cells. Myristylation of HIV-1 Gag precursor polyprotein Pr55Gag is required for stable membrane binding and for assembly of viral particles. We expressed a series of proteins representing major regions of the HIV-1 Gag protein both with and without an intact myristyl acceptor glycine and performed subcellular fractionation studies to identify additional regions critical for membrane binding. Myristylation-dependent binding of Pr55Gag was demonstrated by using the vaccinia virus/T7 hybrid system for protein expression. Domains within the matrix protein (MA) region downstream of the initial 15 amino acids were required for membrane binding which was resistant to a high salt concentration (1 M NaCl). A myristylated construct lacking most of the matrix protein did not associate with the plasma membrane but formed intracellular retrovirus-like particles. A nonmyristylated construct lacking most of the MA region also was demonstrated by electron microscopy to form intracellular particles. Retrovirus-like extracellular particles were produced with a Gag protein construct lacking all of p6 and most of the nucleocapsid region. These studies suggest that a domain within the MA region downstream from the myristylation site is required for transport of Gag polyprotein to the plasma membrane and that stable plasma membrane binding requires both myristic acid and a downstream MA domain. The carboxyl-terminal p6 region and most of the nucleocapsid region are not required for retrovirus-like particle formation.
Collapse
Affiliation(s)
- P Spearman
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | | | | | | |
Collapse
|
37
|
Chapman NM, Tu Z, Tracy S, Gauntt CJ. An infectious cDNA copy of the genome of a non-cardiovirulent coxsackievirus B3 strain: its complete sequence analysis and comparison to the genomes of cardiovirulent coxsackieviruses. Arch Virol 1994; 135:115-30. [PMID: 8198437 DOI: 10.1007/bf01309769] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The genome of the non-cardiovirulent coxsackievirus B3 (CVB3) strain CVB3/0 was cloned and sequenced to aid in the elucidation of the viral genetic basis for the CVB3 cardiovirulent phenotype. Reverse-transcribed sub-genomic complementary DNA (cDNA) fragments were enzymatically amplified using generic oligonucleotide primers and were assembled as a complete infectious genomic copy (pCVB3-0) downstream of the T7 RNA polymerase promoter. Positive-strand viral RNA transcribed from pCVB3-0 using T7 RNA polymerase and transfected into HeLa cells produced infectious virus (CVB3/0c). No differences in phenotype were observed comparing growth of CVB3/0c to the parental CVB3/0 in HeLa single-step growth curves, virus yields, or plaque size. When inoculated into C3H/HeJ mice, CVB3/0c achieved cardiac titers equivalent to the parental CVB3/0 and like the parental virus, demonstrated a non-cardiovirulent phenotype. The nucleotide sequence of the cloned CVB3/0 genome was determined and compared to the genomes of infectious cDNA clones of cardiovirulent CVB3 strains. Two consistent differences among nucleotides in non-translated regions and 8 amino acid differences relative to two well-characterized infectious cDNA copies of genomes from cardiovirulent CVB3 strains were identified.
Collapse
Affiliation(s)
- N M Chapman
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha
| | | | | | | |
Collapse
|
38
|
Lea S, Hernández J, Blakemore W, Brocchi E, Curry S, Domingo E, Fry E, Abu-Ghazaleh R, King A, Newman J. The structure and antigenicity of a type C foot-and-mouth disease virus. Structure 1994; 2:123-39. [PMID: 8081743 DOI: 10.1016/s0969-2126(00)00014-9] [Citation(s) in RCA: 204] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Picornaviruses are responsible for a wide range of mammalian diseases and, in common with other RNA viruses, show considerable antigenic variation. Foot-and-mouth disease viruses (FMDVs) constitute one genus of the picornavirus family and are classified into seven serotypes, each of which shows considerable intratypic variation. This antigenic variation leads to continuing difficulties in controlling the disease. To date the structure of only one serotype, O, has been reported. RESULTS The three-dimensional structure of a serotype C (isolate C-S8c1) FMDV, has been determined crystallographically at 3.5 A resolution. The main chain conformation of the virion is very similar to that of type O1 virus. The immunodominant G-H loop of VP1, the presumed site of cell attachment, is disordered in both types of virus indicating a functional role for flexibility of this region. There are significant changes in the structure of other antigenic loops and in some internal regions involved in protomer-protomer contacts, including the entire amino-terminal portion of VP2, described here for the first time for a picornavirus. Antigenic sites have been identified by genetic and peptide mapping methods, and located on the capsid. The data reveal a major new discontinuous antigenic site (site D) which is located near to the three-fold axis and involves residues of VP1, VP2 and VP3 which lie adjacent to each other on the capsid. CONCLUSION In FMDV type C, amino acid substitutions seen in mutants that are resistant to neutralization by monoclonal antibodies (MAbs) map to predominantly surface-oriented residues with solvent-accessible side-chains not involved in interactions with other amino acids, whereas residues which are accessible but not substituted are found to be more frequently involved in protein-protein interactions. This provides a molecular interpretation for the repeated isolation of the same amino acid substitutions in MAb-resistant variants, an observation frequently made with RNA viruses. This first comparison of two FMDV serotypes shows how subtle changes at antigenic sites are sufficient to cause large changes in antigenic specificity between serotypes.
Collapse
Affiliation(s)
- S Lea
- Lab. of Molecular Biophysics, University of Oxford, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Moscufo N, Yafal AG, Rogove A, Hogle J, Chow M. A mutation in VP4 defines a new step in the late stages of cell entry by poliovirus. J Virol 1993; 67:5075-8. [PMID: 8392631 PMCID: PMC237900 DOI: 10.1128/jvi.67.8.5075-5078.1993] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
During the entry of poliovirus into cells, a conformational transition occurs within the virion that is dependent upon its binding to the cell surface receptor. This conformational rearrangement generates an altered particle of 135S, results in the extrusion of capsid protein VP4 and the amino terminus of VP1 from the virion interior, and leads to the acquisition of membrane-binding properties by the 135S particle. Although the subsequent fate of VP4 is unknown, its apparent absence from purified 135S particles has long suggested that VP4 is not directly involved during virus entry. We report here the construction by site-specific mutagenesis of a nonviable VP4 mutant that upon transfection of the cDNA appears to form mature virus particles. These particles, upon interaction with the cellular receptor, undergo the 135S conformational transition but are defective at a subsequent stage in virus entry. The results demonstrate that the participation of VP4 is required during cell entry of poliovirus. In addition, these data indicate the existence of additional stages in the cell entry process beyond receptor binding and the transition to 135S particles. These post-135S stages must include the poorly understood processes by which nonenveloped viruses cross the cell membrane, uncoat, and deliver their genomes into the cytoplasm.
Collapse
Affiliation(s)
- N Moscufo
- Department of Biology, Massachusetts Institute of Technology, Cambridge 02139-4307
| | | | | | | | | |
Collapse
|
40
|
Harty RN, Caughman GB, Holden VR, O'Callaghan DJ. Characterization of the myristylated polypeptide encoded by the UL1 gene that is conserved in the genome of defective interfering particles of equine herpesvirus 1. J Virol 1993; 67:4122-32. [PMID: 8389920 PMCID: PMC237781 DOI: 10.1128/jvi.67.7.4122-4132.1993] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Equine herpesvirus 1 (EHV-1, Kentucky A strain) preparations enriched for defective interfering particles (DIPs) can readily establish persistent infection. The UL1 gene, which is conserved in the genome of DIPs that mediate persistent infection, maps between nucleotides 1418 and 2192 (258 amino acids) from the L (long) terminus. UL1 has no homology with any known gene encoded by herpes simplex virus type 1 but has limited homology to open reading frame 2 of varicella-zoster virus and the "circ" gene of bovine herpesvirus type 1. Previous work showed that the EHV-1 UL1 gene belongs to the early kinetic class and is transcribed as a 1.2-kb polyadenylated mRNA (R. N. Harty, R. R. Yalamanchili, and D. J. O'Callaghan, Virology 183:830-833, 1991). In this report, the UL1 protein was identified and characterized as a 33-kDa polypeptide in EHV-1-infected cells by using rabbit polyclonal antiserum raised against a TrpE-UL1 fusion protein (amino acids 7 to 258 of UL1) synthesized in Escherichia coli. Results from Western blot (immunoblot), immunoprecipitation, indirect immunofluorescence, and biochemical analyses indicated that the UL1 polypeptide (i) is more abundant in cells infected with DIP-enriched virus than in cells infected with standard EHV-1, (ii) is synthesized as early as 3 h postinfection (p.i.) in infection with standard virus or in infection with DIP-enriched virus preparations and increases in abundance up to 12 h p.i., (iii) appears to be associated with the rough endoplasmic reticulum-Golgi apparatus early in infection (3 to 4 h p.i.), while a diffuse cytoplasmic pattern of fluorescence is observed late in infection (7 to 8 h p.i.), (iv) is modified by myristic acid as it contains a consensus N-terminal myristylation site and is readily labeled with [3H]myristic acid, and (v) is associated with mature EHV-1 virions.
Collapse
Affiliation(s)
- R N Harty
- Department of Microbiology and Immunology, Louisiana State University Medical Center, Shreveport 71130-3932
| | | | | | | |
Collapse
|
41
|
Parenti M, Viganó MA, Newman CM, Milligan G, Magee AI. A novel N-terminal motif for palmitoylation of G-protein alpha subunits. Biochem J 1993; 291 ( Pt 2):349-53. [PMID: 8484716 PMCID: PMC1132531 DOI: 10.1042/bj2910349] [Citation(s) in RCA: 141] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have examined the post-translational processing of G alpha subunits expressed endogenously in rat PC12 and NG108-15 rat/mouse hybrid cells, and after transfection of cDNA expression constructs into COS cells. Thioester-linked palmitoylation of alpha o, alpha s, alpha q/alpha 11 and alpha 12 has been detected by metabolic labelling with [3H]palmitate and immunoprecipitation. Palmitoylation of alpha o occurs post-translationally in cells treated with protein-synthesis inhibitors, suggesting possible dynamic acylation. Palmitoylation of the C-terminal CAAX motif has been excluded. Site-directed mutagenesis of alpha o has been used to implicate the site of modification as a cysteine residue next to the N-terminal myristoylated glycine, in a novel protein-lipid modification motif Met-Gly-Cys. The non-palmitoylated alpha o mutant is still myristoylated but shows reduced membrane binding, suggesting that reversible palmitoylation may regulate G alpha localization and function.
Collapse
Affiliation(s)
- M Parenti
- Dipartimento di Farmacología, Milan, Italy
| | | | | | | | | |
Collapse
|
42
|
Simons J, Rogove A, Moscufo N, Reynolds C, Chow M. Efficient analysis of nonviable poliovirus capsid mutants. J Virol 1993; 67:1734-8. [PMID: 8382319 PMCID: PMC237553 DOI: 10.1128/jvi.67.3.1734-1738.1993] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nonviable poliovirus capsid mutants were studied by an efficient infection-transfection system. Phenotypically, nonviable poliovirus capsid mutants appear to segregate into three classes: those that form only protomers, those that can form pentamers, and one that can form completed virions.
Collapse
Affiliation(s)
- J Simons
- Department of Applied Biological Sciences, Massachusetts Institute of Technology, Cambridge 02139
| | | | | | | | | |
Collapse
|