1
|
Wang J, Chen D, Wei F, Deng J, Su J, Lin X, Wu S. Generation of Stable Cell Lines Expressing Akabane Virus N Protein and Insight into Its Function in Viral Replication. Pathogens 2023; 12:1058. [PMID: 37624018 PMCID: PMC10459709 DOI: 10.3390/pathogens12081058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/06/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Akabane virus (AKAV) is a world wide epidemic arbovirus belonging to the Bunyavirales order that predominantly infects livestock and causes severe congenital malformations. The nucleocapsid (N) protein of AKAV possesses multiple important functions in the virus life cycle, and it is an ideal choice for AKAV detection. In this study, we successfully constructed two stable BHK-21 cell lines (C8H2 and F7E5) that constitutively express the AKAV N protein using a lentivirus system combined with puromycin selection. RT-PCR analysis confirmed that the AKAV N gene was integrated into the BHK-21 cell genome and consistently transcribed. Indirect immunofluorescence (IFA) and Western blot (WB) assays proved that both C8H2 and F7E5 cells could react with the AKAV N protein mAb specifically, indicating potential applications in AKAV detection. Furthermore, we analyzed the growth kinetics of AKAV in the C8H2 and F7E5 cell lines and observed temporary inhibition of viral replication at 12, 24 and 36 h postinfection (hpi) compared to BHK-21 cells. Subsequent investigations suggested that the reduced viral replication was linked to the down-regulation of the viral mRNAs (Gc and RdRp). In summary, we have established materials for detecting AKAV and gained new insights into the function of the AKAV N protein.
Collapse
Affiliation(s)
- Jingjing Wang
- Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine, Beijing 100176, China; (J.W.); (D.C.); (F.W.); (J.D.); (X.L.)
| | - Dongjie Chen
- Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine, Beijing 100176, China; (J.W.); (D.C.); (F.W.); (J.D.); (X.L.)
| | - Fang Wei
- Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine, Beijing 100176, China; (J.W.); (D.C.); (F.W.); (J.D.); (X.L.)
| | - Junhua Deng
- Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine, Beijing 100176, China; (J.W.); (D.C.); (F.W.); (J.D.); (X.L.)
| | - Jia Su
- China Institute of Veterinary Drug Control, Beijing 100081, China;
| | - Xiangmei Lin
- Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine, Beijing 100176, China; (J.W.); (D.C.); (F.W.); (J.D.); (X.L.)
| | - Shaoqiang Wu
- Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine, Beijing 100176, China; (J.W.); (D.C.); (F.W.); (J.D.); (X.L.)
| |
Collapse
|
2
|
LaPointe A, Gale M, Kell AM. Orthohantavirus Replication in the Context of Innate Immunity. Viruses 2023; 15:1130. [PMID: 37243216 PMCID: PMC10220641 DOI: 10.3390/v15051130] [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: 04/12/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Orthohantaviruses are rodent-borne, negative-sense RNA viruses that are capable of causing severe vascular disease in humans. Over the course of viral evolution, these viruses have tailored their replication cycles in such a way as to avoid and/or antagonize host innate immune responses. In the rodent reservoir, this results in life long asymptomatic infections. However, in hosts other than its co-evolved reservoir, the mechanisms for subduing the innate immune response may be less efficient or absent, potentially leading to disease and/or viral clearance. In the case of human orthohantavirus infection, the interaction of the innate immune response with viral replication is thought to give rise to severe vascular disease. The orthohantavirus field has made significant advancements in understanding how these viruses replicate and interact with host innate immune responses since their identification by Dr. Ho Wang Lee and colleagues in 1976. Therefore, the purpose of this review, as part of this special issue dedicated to Dr. Lee, was to summarize the current knowledge of orthohantavirus replication, how viral replication activates innate immunity, and how the host antiviral response, in turn, impacts viral replication.
Collapse
Affiliation(s)
- Autumn LaPointe
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98109, USA
| | - Alison M. Kell
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| |
Collapse
|
3
|
Malet H, Williams HM, Cusack S, Rosenthal M. The mechanism of genome replication and transcription in bunyaviruses. PLoS Pathog 2023; 19:e1011060. [PMID: 36634042 PMCID: PMC9836281 DOI: 10.1371/journal.ppat.1011060] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Bunyaviruses are negative sense, single-strand RNA viruses that infect a wide range of vertebrate, invertebrate and plant hosts. WHO lists three bunyavirus diseases as priority diseases requiring urgent development of medical countermeasures highlighting their high epidemic potential. While the viral large (L) protein containing the RNA-dependent RNA polymerase is a key enzyme in the viral replication cycle and therefore a suitable drug target, our knowledge on the structure and activities of this multifunctional protein has, until recently, been very limited. However, in the last few years, facilitated by the technical advances in the field of cryogenic electron microscopy, many structures of bunyavirus L proteins have been solved. These structures significantly enhance our mechanistic understanding of bunyavirus genome replication and transcription processes and highlight differences and commonalities between the L proteins of different bunyavirus families. Here, we provide a review of our current understanding of genome replication and transcription in bunyaviruses with a focus on the viral L protein. Further, we compare within bunyaviruses and with the related influenza virus polymerase complex and highlight open questions.
Collapse
Affiliation(s)
- Hélène Malet
- University Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| | - Harry M. Williams
- Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
| | | | - Maria Rosenthal
- Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Discovery Research ScreeningPort, Hamburg, Germany
| |
Collapse
|
4
|
Tamiya K, Kobayashi S, Yoshii K, Kariwa H. Analysis of the relationship between replication of the Hokkaido genotype of Puumala orthohantavirus and autophagy. Virus Res 2022; 318:198830. [DOI: 10.1016/j.virusres.2022.198830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/02/2022] [Accepted: 05/27/2022] [Indexed: 11/29/2022]
|
5
|
Welke RW, Sperber HS, Bergmann R, Koikkarah A, Menke L, Sieben C, Krüger DH, Chiantia S, Herrmann A, Schwarzer R. Characterization of Hantavirus N Protein Intracellular Dynamics and Localization. Viruses 2022; 14:v14030457. [PMID: 35336863 PMCID: PMC8954124 DOI: 10.3390/v14030457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/02/2022] [Accepted: 02/18/2022] [Indexed: 02/07/2023] Open
Abstract
Hantaviruses are enveloped viruses that possess a tri-segmented, negative-sense RNA genome. The viral S-segment encodes the multifunctional nucleocapsid protein (N), which is involved in genome packaging, intracellular protein transport, immunoregulation, and several other crucial processes during hantavirus infection. In this study, we generated fluorescently tagged N protein constructs derived from Puumalavirus (PUUV), the dominant hantavirus species in Central, Northern, and Eastern Europe. We comprehensively characterized this protein in the rodent cell line CHO-K1, monitoring the dynamics of N protein complex formation and investigating co-localization with host proteins as well as the viral glycoproteins Gc and Gn. We observed formation of large, fibrillar PUUV N protein aggregates, rapidly coalescing from early punctate and spike-like assemblies. Moreover, we found significant spatial correlation of N with vimentin, actin, and P-bodies but not with microtubules. N constructs also co-localized with Gn and Gc albeit not as strongly as the glycoproteins associated with each other. Finally, we assessed oligomerization of N constructs, observing efficient and concentration-dependent multimerization, with complexes comprising more than 10 individual proteins.
Collapse
Affiliation(s)
- Robert-William Welke
- Department of Molecular Biophysics, Humboldt University, 10115 Berlin, Germany; (R.-W.W.); (R.B.); (A.H.)
| | - Hannah Sabeth Sperber
- Institute for Translational HIV Research, University Hospital Essen, 45147 Essen, Germany;
| | - Ronny Bergmann
- Department of Molecular Biophysics, Humboldt University, 10115 Berlin, Germany; (R.-W.W.); (R.B.); (A.H.)
| | - Amit Koikkarah
- Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany; (A.K.); (S.C.)
| | - Laura Menke
- Nanoscale Infection Biology Group, Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (L.M.); (C.S.)
| | - Christian Sieben
- Nanoscale Infection Biology Group, Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (L.M.); (C.S.)
- Institute for Genetics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Detlev H. Krüger
- Institut für Virologie, Charité–Universitätsmedizin Berlin, Gliedkörperschaft der Freien Universität Berlin und der Humboldt-Universität zu Berlin, 10117 Berlin, Germany;
| | - Salvatore Chiantia
- Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany; (A.K.); (S.C.)
| | - Andreas Herrmann
- Department of Molecular Biophysics, Humboldt University, 10115 Berlin, Germany; (R.-W.W.); (R.B.); (A.H.)
- Biophysikalische Chemie, Freie Universität, 14195 Berlin, Germany
| | - Roland Schwarzer
- Institute for Translational HIV Research, University Hospital Essen, 45147 Essen, Germany;
- Correspondence:
| |
Collapse
|
6
|
Guardado-Calvo P, Rey FA. The surface glycoproteins of hantaviruses. Curr Opin Virol 2021; 50:87-94. [PMID: 34418649 DOI: 10.1016/j.coviro.2021.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022]
Abstract
Hantaviruses are rodent-borne viruses distributed worldwide, transmitted through the air and with the ability to spread from person to person. They maintain a non-symptomatic persistent infection in their rodent hosts, but their spillover to humans produces a renal or pulmonary syndrome associated with high fatality rates. Hantavirus particles are lipid-enveloped and display a characteristic surface lattice built up of tetragonal spikes composed of two glycoproteins, Gn and Gc. The pleomorphism of these particles has hindered cryo-EM efforts to obtain detailed structural information and only by using a combination of X-ray crystallography and cryo-electron tomography it was possible to build an atomic model of the surface lattice. Here we review these structural efforts and the unanticipated evolutionary relations between hantaviruses and alphaviruses highlighted by these studies.
Collapse
Affiliation(s)
| | - Félix A Rey
- Institut Pasteur, Structural Virology Unit, and CNRS UMR 3569, Paris, France
| |
Collapse
|
7
|
Meier K, Thorkelsson SR, Quemin ERJ, Rosenthal M. Hantavirus Replication Cycle-An Updated Structural Virology Perspective. Viruses 2021; 13:1561. [PMID: 34452426 PMCID: PMC8402763 DOI: 10.3390/v13081561] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022] Open
Abstract
Hantaviruses infect a wide range of hosts including insectivores and rodents and can also cause zoonotic infections in humans, which can lead to severe disease with possible fatal outcomes. Hantavirus outbreaks are usually linked to the population dynamics of the host animals and their habitats being in close proximity to humans, which is becoming increasingly important in a globalized world. Currently there is neither an approved vaccine nor a specific and effective antiviral treatment available for use in humans. Hantaviruses belong to the order Bunyavirales with a tri-segmented negative-sense RNA genome. They encode only five viral proteins and replicate and transcribe their genome in the cytoplasm of infected cells. However, many details of the viral amplification cycle are still unknown. In recent years, structural biology methods such as cryo-electron tomography, cryo-electron microscopy, and crystallography have contributed essentially to our understanding of virus entry by membrane fusion as well as genome encapsidation by the nucleoprotein. In this review, we provide an update on the hantavirus replication cycle with a special focus on structural virology aspects.
Collapse
Affiliation(s)
- Kristina Meier
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
| | - Sigurdur R. Thorkelsson
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Emmanuelle R. J. Quemin
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Maria Rosenthal
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| |
Collapse
|
8
|
Detection of Envelope Glycoprotein Assembly from Old-World Hantaviruses in the Golgi Apparatus of Living Cells. J Virol 2021; 95:JVI.01238-20. [PMID: 33239451 PMCID: PMC7851546 DOI: 10.1128/jvi.01238-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hantaviruses are emerging pathogens that occasionally cause deadly outbreaks in the human population. While the structure of the viral envelope has been characterized with high precision, protein-protein interactions leading to the formation of new virions in infected cells are not fully understood yet. We use quantitative fluorescence microscopy (i.e., Number&Brightness analysis and fluorescence fluctuation spectroscopy) to monitor the interactions that lead to oligomeric spike complex formation in the physiological context of living cells. To this aim, we quantified protein-protein interactions for the glycoproteins Gn and Gc from Puumala and Hantaan orthohantaviruses in several cellular models. The oligomerization of each protein was analyzed in relation to subcellular localization, concentration, and the concentration of its interaction partner. Our results indicate that when expressed separately, Gn and Gc form respectively homo-tetrameric and homo-dimeric complexes, in a concentration-dependent manner. Site-directed mutations or deletion mutants showed the specificity of their homotypic interactions. When both glycoproteins were co-expressed, we observed in the Golgi apparatus clear indication of Gn-Gc interactions and the formation of Gn-Gc multimeric protein complexes of different sizes, while using various labeling schemes to minimize the influence of the fluorescent tags. Such large glycoprotein multimers may be identified as multiple Gn viral spikes interconnected via Gc-Gc contacts. This observation provides a possible first evidence for the initial assembly steps of the viral envelope, within this organelle, directly in living cells.IMPORTANCE In this work, we investigate protein-protein interactions that drive the assembly of the hantaviruses envelope. These emerging pathogens have the potential to cause deadly outbreaks in the human population. Therefore, it is important to improve our quantitative understanding of the viral assembly process in infected cells, from a molecular point of view. By applying advanced fluorescence microscopy methods, we monitored the formation of viral spike complexes in different cell types. Our data support a model for hantavirus assembly according to which viral spikes are formed via the clustering of hetero-dimers of the two viral glycoproteins Gn and Gc. Furthermore, the observation of large Gn-Gc hetero-multimers provide a possible first evidence for the initial assembly steps of the viral envelope, directly in the Golgi apparatus of living cells.
Collapse
|
9
|
Mittler E, Dieterle ME, Kleinfelter LM, Slough MM, Chandran K, Jangra RK. Hantavirus entry: Perspectives and recent advances. Adv Virus Res 2019; 104:185-224. [PMID: 31439149 DOI: 10.1016/bs.aivir.2019.07.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hantaviruses are important zoonotic pathogens of public health importance that are found on all continents except Antarctica and are associated with hemorrhagic fever with renal syndrome (HFRS) in the Old World and hantavirus pulmonary syndrome (HPS) in the New World. Despite the significant disease burden they cause, no FDA-approved specific therapeutics or vaccines exist against these lethal viruses. The lack of available interventions is largely due to an incomplete understanding of hantavirus pathogenesis and molecular mechanisms of virus replication, including cellular entry. Hantavirus Gn/Gc glycoproteins are the only viral proteins exposed on the surface of virions and are necessary and sufficient to orchestrate virus attachment and entry. In vitro studies have implicated integrins (β1-3), DAF/CD55, and gC1qR as candidate receptors that mediate viral attachment for both Old World and New World hantaviruses. Recently, protocadherin-1 (PCDH1) was demonstrated as a requirement for cellular attachment and entry of New World hantaviruses in vitro and lethal HPS in vivo, making it the first clade-specific host factor to be identified. Attachment of hantavirus particles to cellular receptors induces their internalization by clathrin-mediated, dynamin-independent, or macropinocytosis-like mechanisms, followed by particle trafficking to an endosomal compartment where the fusion of viral and endosomal membranes can occur. Following membrane fusion, which requires cholesterol and acid pH, viral nucleocapsids escape into the cytoplasm and launch genome replication. In this review, we discuss the current mechanistic understanding of hantavirus entry, highlight gaps in our existing knowledge, and suggest areas for future inquiry.
Collapse
Affiliation(s)
- Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maria Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lara M Kleinfelter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Megan M Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
10
|
Sperber HS, Welke RW, Petazzi RA, Bergmann R, Schade M, Shai Y, Chiantia S, Herrmann A, Schwarzer R. Self-association and subcellular localization of Puumala hantavirus envelope proteins. Sci Rep 2019; 9:707. [PMID: 30679542 PMCID: PMC6345964 DOI: 10.1038/s41598-018-36879-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/28/2018] [Indexed: 01/08/2023] Open
Abstract
Hantavirus assembly and budding are governed by the surface glycoproteins Gn and Gc. In this study, we investigated the glycoproteins of Puumala, the most abundant Hantavirus species in Europe, using fluorescently labeled wild-type constructs and cytoplasmic tail (CT) mutants. We analyzed their intracellular distribution, co-localization and oligomerization, applying comprehensive live, single-cell fluorescence techniques, including confocal microscopy, imaging flow cytometry, anisotropy imaging and Number&Brightness analysis. We demonstrate that Gc is significantly enriched in the Golgi apparatus in absence of other viral components, while Gn is mainly restricted to the endoplasmic reticulum (ER). Importantly, upon co-expression both glycoproteins were found in the Golgi apparatus. Furthermore, we show that an intact CT of Gc is necessary for efficient Golgi localization, while the CT of Gn influences protein stability. Finally, we found that Gn assembles into higher-order homo-oligomers, mainly dimers and tetramers, in the ER while Gc was present as mixture of monomers and dimers within the Golgi apparatus. Our findings suggest that PUUV Gc is the driving factor of the targeting of Gc and Gn to the Golgi region, while Gn possesses a significantly stronger self-association potential.
Collapse
Affiliation(s)
- Hannah Sabeth Sperber
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany.,Vitalant Research Institute, 270 Masonic Ave, San Francisco, CA, 94118, USA
| | - Robert-William Welke
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany
| | - Roberto Arturo Petazzi
- University of Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht-Str. 24-25, 14476, Potsdam, Germany
| | - Ronny Bergmann
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany
| | - Matthias Schade
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany
| | - Yechiel Shai
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Salvatore Chiantia
- University of Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht-Str. 24-25, 14476, Potsdam, Germany
| | - Andreas Herrmann
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany.
| | - Roland Schwarzer
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany. .,Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel. .,Gladstone Institute of Virology and Immunology, 1650 Owens Street, San Francisco, CA, 95158, USA.
| |
Collapse
|
11
|
Two Point Mutations in Old World Hantavirus Glycoproteins Afford the Generation of Highly Infectious Recombinant Vesicular Stomatitis Virus Vectors. mBio 2019; 10:mBio.02372-18. [PMID: 30622188 PMCID: PMC6325249 DOI: 10.1128/mbio.02372-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human hantavirus infections cause hantavirus pulmonary syndrome in the Americas and hemorrhagic fever with renal syndrome (HFRS) in Eurasia. No FDA-approved vaccines and therapeutics exist for these deadly viruses, and their development is limited by the requirement for high biocontainment. In this study, we identified and characterized key amino acid changes in the surface glycoproteins of HFRS-causing Hantaan virus that enhance their incorporation into recombinant vesicular stomatitis virus (rVSV) particles. The replication-competent rVSVs encoding Hantaan virus and Dobrava-Belgrade virus glycoproteins described in this work provide a powerful and facile system to study hantavirus entry under lower biocontainment and may have utility as hantavirus vaccines. Rodent-to-human transmission of hantaviruses is associated with severe disease. Currently, no FDA-approved, specific antivirals or vaccines are available, and the requirement for high biocontainment (biosafety level 3 [BSL-3]) laboratories limits hantavirus research. To study hantavirus entry in a BSL-2 laboratory, we set out to generate replication-competent, recombinant vesicular stomatitis viruses (rVSVs) bearing the Gn and Gc (Gn/Gc) entry glycoproteins. As previously reported, rVSVs bearing New World hantavirus Gn/Gc were readily rescued from cDNAs, but their counterparts bearing Gn/Gc from the Old World hantaviruses, Hantaan virus (HTNV) or Dobrava-Belgrade virus (DOBV), were refractory to rescue. However, serial passage of the rescued rVSV-HTNV Gn/Gc virus markedly increased its infectivity and capacity for cell-to-cell spread. This gain in viral fitness was associated with the acquisition of two point mutations: I532K in the cytoplasmic tail of Gn and S1094L in the membrane-proximal stem of Gc. Follow-up experiments with rVSVs and single-cycle VSV pseudotypes confirmed these results. Mechanistic studies revealed that both mutations were determinative and contributed to viral infectivity in a synergistic manner. Our findings indicate that the primary mode of action of these mutations is to relocalize HTNV Gn/Gc from the Golgi complex to the cell surface, thereby affording significantly enhanced Gn/Gc incorporation into budding VSV particles. Finally, I532K/S1094L mutations in DOBV Gn/Gc permitted the rescue of rVSV-DOBV Gn/Gc, demonstrating that incorporation of cognate mutations into other hantaviral Gn/Gc proteins could afford the generation of rVSVs that are otherwise challenging to rescue. The robust replication-competent rVSVs, bearing HTNV and DOBV Gn/Gc, reported herein may also have utility as vaccines.
Collapse
|
12
|
Hägele S, Müller A, Nusshag C, Reiser J, Zeier M, Krautkrämer E. Virus- and cell type-specific effects in orthohantavirus infection. Virus Res 2018; 260:102-113. [PMID: 30508604 DOI: 10.1016/j.virusres.2018.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/15/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023]
Abstract
Orthohantaviruses Hantaan (HTNV) and Puumala (PUUV) virus cause hemorrhagic fever with renal syndrome (HFRS), that is characterized by acute renal failure with often massive proteinuria and by morphological changes of the tubular and glomerular apparatus. Orthohantaviral N protein is found in renal cells and plays a key role in replication. However, the replication in human renal cells is not well characterized. Therefore, we examined the orthohantaviral infection in different human renal cells. Differences in localization of N protein, release of particles, and modulation of the actin cytoskeleton between both virus species are observed in human renal cells. A substantial portion of HTNV N protein demonstrates a filamentous pattern in addition to the typical punctate pattern. Release of HTNV depends on an intact actin and microtubule cytoskeleton. In contrast, PUUV N protein is generally localized in a punctate pattern and release of PUUV does not require an intact actin cytoskeleton. Infection of podocytes results in cytoskeletal rearrangements that are more pronounced for HTNV. Analyzing Vero E6 cells revealed differences compared to human renal cells. The pattern of N proteins is strictly punctate, release does not depend on an intact actin cytoskeleton and cytoskeletal rearrangements are not present. No virus-specific variations between HTNV and PUUV are observed in Vero E6 cells. Using human renal cells as cell culture model for orthohantavirus infection demonstrates virus-specific differences and orthohantavirus-induced cytoskeletal rearrangements that are not observed in Vero E6 cells. Therefore, the choice of an appropriate cell culture system is a prerequisite to study orthohantavirus pathogenicity.
Collapse
Affiliation(s)
- Stefan Hägele
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Alexander Müller
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University, Medical Center, Chicago, IL, USA
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Ellen Krautkrämer
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
13
|
Jiang DB, Zhang JP, Cheng LF, Zhang GW, Li Y, Li ZC, Lu ZH, Zhang ZX, Lu YC, Zheng LH, Zhang FL, Yang K. Hantavirus Gc induces long-term immune protection via LAMP-targeting DNA vaccine strategy. Antiviral Res 2018; 150:174-182. [PMID: 29273568 DOI: 10.1016/j.antiviral.2017.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/14/2017] [Accepted: 12/16/2017] [Indexed: 01/22/2023]
Abstract
Hemorrhagic fever with renal syndrome (HFRS) occurs widely throughout Eurasia. Unfortunately, there is no effective treatment, and prophylaxis remains the best option against the major pathogenic agent, hantaan virus (HTNV), which is an Old World hantavirus. However, the absence of cellular immune responses and immunological memory hampers acceptance of the current inactivated HFRS vaccine. Previous studies revealed that a lysosome-associated membrane protein 1 (LAMP1)-targeting strategy involving a DNA vaccine based on the HTNV glycoprotein Gn successfully conferred long-term immunity, and indicated that further research on Gc, another HTNV antigen, was warranted. Plasmids encoding Gc and lysosome-targeted Gc, designated pVAX-Gc and pVAX-LAMP/Gc, respectively, were constructed. Proteins of interest were identified by fluorescence microscopy following cell line transfection. Five groups of 20 female BALB/c mice were subjected to the following inoculations: inactivated HTNV vaccine, pVAX-LAMP/Gc, pVAX-Gc, and, as the negative controls, pVAX-LAMP or the blank vector pVAX1. Humoral and cellular immunity were assessed by enzyme-linked immunosorbent assays (ELISAs) and 15-mer peptide enzyme-linked immunospot (ELISpot) epitope mapping assays. Repeated immunization with pVAX-LAMP/Gc enhanced adaptive immune responses, as demonstrated by the specific and neutralizing antibody titers and increased IFN-γ production. The inactivated vaccine induced a comparable humoral reaction, but the negative controls only elicited insignificant responses. Using a mouse model of HTNV challenge, the in vivo protection conferred by the inactivated vaccine and Gc-based constructs (with/without LAMP recombination) was confirmed. Evidence of pan-epitope reactions highlighted the long-term cellular response to the LAMP-targeting strategy, and histological observations indicated the safety of the LAMP-targeting vaccines. The long-term protective immune responses induced by pVAX-LAMP/Gc may be due to the advantage afforded by lysosomal targeting after exogenous antigen processing initiation and major histocompatibility complex (MHC) class II antigen presentation trafficking. MHC II-restricted antigen recognition effectively primes HTNV-specific CD4+ T-cells, leading to the promotion of significant immune responses and immunological memory. An epitope-spreading phenomenon was observed, which mirrors the previous result from the Gn study, in which the dominant IFN-γ-responsive hot-spot epitopes were shared between HLA-II and H2d. Importantly, the pan-epitope reaction to Gc indicated that Gc should be with potential for use in further hantavirus DNA vaccine investigations.
Collapse
Affiliation(s)
- Dong-Bo Jiang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Jin-Peng Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Lin-Feng Cheng
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Guan-Wen Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Yun Li
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Zi-Chao Li
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Zhen-Hua Lu
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Zi-Xin Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Yu-Chen Lu
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Lian-He Zheng
- Department of Orthopedics, Tangdu Hospital, Xi'an, China.
| | - Fang-Lin Zhang
- Department of Microbiology, Fourth Military Medical University, Xi'an, China.
| | - Kun Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
14
|
Reuter M, Krüger DH. The nucleocapsid protein of hantaviruses: much more than a genome-wrapping protein. Virus Genes 2017; 54:5-16. [PMID: 29159494 DOI: 10.1007/s11262-017-1522-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/11/2017] [Indexed: 12/11/2022]
Abstract
The nucleocapsid (N) protein of hantaviruses represents an impressive example of a viral multifunctional protein. It encompasses properties as diverse as genome packaging, RNA chaperoning, intracellular protein transport, DNA degradation, intervention in host translation, and restricting host immune responses. These functions all rely on the capability of N to interact with RNA and other viral and cellular proteins. We have compiled data on the N protein of different hantavirus species together with information of the recently published three-dimensional structural data of the protein. The array of diverse functional activities accommodated in the hantaviral N protein goes far beyond to be a static structural protein and makes it an interesting target in the development of antiviral therapeutics.
Collapse
Affiliation(s)
- Monika Reuter
- Institute of Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Detlev H Krüger
- Institute of Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
15
|
Crimean-Congo hemorrhagic fever virus nucleocapsid protein has dual RNA binding modes. PLoS One 2017; 12:e0184935. [PMID: 28922369 PMCID: PMC5602631 DOI: 10.1371/journal.pone.0184935] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/01/2017] [Indexed: 12/17/2022] Open
Abstract
Crimean Congo hemorrhagic fever, a zoonotic viral disease, has high mortality rate in humans. There is currently no vaccine for Crimean Congo hemorrhagic fever virus (CCHFV) and chemical interventions are limited. The three negative sense genomic RNA segments of CCHFV are specifically encapsidated by the nucleocapsid protein into three ribonucleocapsids, which serve as templates for the viral RNA dependent RNA polymerase. Here we demonstrate that CCHFV nucleocapsid protein has two distinct binding modes for double and single strand RNA. In the double strand RNA binding mode, the nucleocapsid protein preferentially binds to the vRNA panhandle formed by the base pairing of complementary nucleotides at the 5’ and 3’ termini of viral genome. The CCHFV nucleocapsid protein does not have RNA helix unwinding activity and hence does not melt the duplex vRNA panhandle after binding. In the single strand RNA binding mode, the nucleocapsid protein does not discriminate between viral and non-viral RNA molecules. Binding of both vRNA panhandle and single strand RNA induce a conformational change in the nucleocapsid protein. Nucleocapsid protein remains in a unique conformational state due to simultaneously binding of structurally distinct vRNA panhandle and single strand RNA substrates. Although the role of dual RNA binding modes in the virus replication cycle is unknown, their involvement in the packaging of viral genome and regulation of CCHFV replication in conjunction with RdRp and host derived RNA regulators is highly likely.
Collapse
|
16
|
Amroun A, Priet S, de Lamballerie X, Quérat G. Bunyaviridae RdRps: structure, motifs, and RNA synthesis machinery. Crit Rev Microbiol 2017; 43:753-778. [PMID: 28418734 DOI: 10.1080/1040841x.2017.1307805] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bunyaviridae family is the largest and most diverse family of RNA viruses. It has more than 350 members divided into five genera: Orthobunyavirus, Phlebovirus, Nairovirus, Hantavirus, and Tospovirus. They are present in the five continents, causing recurrent epidemics, epizootics, and considerable agricultural loss. The genome of bunyaviruses is divided into three segments of negative single-stranded RNA according to their relative size: L (Large), M (Medium) and S (Small) segment. Bunyaviridae RNA-dependent RNA polymerase (RdRp) is encoded by the L segment, and is in charge of the replication and transcription of the viral RNA in the cytoplasm of the infected cell. Viral RdRps share a characteristic right hand-like structure with three subdomains: finger, palm, and thumb subdomains that define the formation of the catalytic cavity. In addition to the N-terminal endonuclease domain, eight conserved motifs (A-H) have been identified in the RdRp of Bunyaviridae. In this review, we have summarized the recent insights from the structural and functional studies of RdRp to understand the roles of different motifs shared by RdRps, the mechanism of viral RNA replication, genome segment packaging by the nucleoprotein, cap-snatching, mRNA transcription, and other RNA mechanisms of bunyaviruses.
Collapse
Affiliation(s)
- Abdennour Amroun
- a Faculté de Médecine , UMR "Emergence des Pathologies Virales" (Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP), Fondation IHU Méditerranée Infection, APHM Public Hospitals of Marseille , Marseille , France
| | - Stéphane Priet
- a Faculté de Médecine , UMR "Emergence des Pathologies Virales" (Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP), Fondation IHU Méditerranée Infection, APHM Public Hospitals of Marseille , Marseille , France
| | - Xavier de Lamballerie
- a Faculté de Médecine , UMR "Emergence des Pathologies Virales" (Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP), Fondation IHU Méditerranée Infection, APHM Public Hospitals of Marseille , Marseille , France
| | - Gilles Quérat
- a Faculté de Médecine , UMR "Emergence des Pathologies Virales" (Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP), Fondation IHU Méditerranée Infection, APHM Public Hospitals of Marseille , Marseille , France
| |
Collapse
|
17
|
Adouchief S, Smura T, Vapalahti O, Hepojoki J. Mapping of human B-cell epitopes of Sindbis virus. J Gen Virol 2016; 97:2243-2254. [PMID: 27339177 DOI: 10.1099/jgv.0.000531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mosquito-transmitted Sindbis virus (SINV) causes fever, skin lesions and musculoskeletal symptoms if transmitted to man. SINV is the prototype virus of genus Alphavirus, which includes other arthritogenic viruses such as chikungunya virus (CHIKV) and Ross River virus (RRV) that cause large epidemics with a considerable public health burden. Until now the human B-cell epitopes have been studied for CHIKV and RRV, but not for SINV. To identify the B-cell epitopes in SINV-infection, we synthetised a library of linear 18-mer peptides covering the structural polyprotein of SINV, and probed it with SINV IgG-positive and IgG-negative serum pools. By comparing the binding profiles of the pools, we identified 15 peptides that were strongly reactive only with the SINV IgG-positive pools. We then utilized alanine scanning and individual (n=22) patient sera to further narrow the number of common B-cell epitopes to six. These epitopes locate to the capsid, E2, E1 and to a region in PE2 (uncleaved E3-E2), which may only be present in immature virions. By sequence comparison, we observed that one of the capsid protein epitopes shares six identical amino acids with macrophage migration inhibitory factor (MIF) receptor, which is linked to inflammatory diseases and to molecular pathology of alphaviral arthritides. Our results add to the current understanding on SINV disease and raise questions of a potential role of uncleaved PE2 and the MIF receptor (CD74) mimotope in human SINV infection.
Collapse
Affiliation(s)
- Samuel Adouchief
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Teemu Smura
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.,Department of Virology and Immunology, Hospital District of Helsinki and Uusimaa (HUSLAB), Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Abstract
Hantaviruses are emerging zoonotic pathogens that belong to the Bunyaviridae family. They have been classified as category A pathogens by CDC (centers for disease control and prevention). Hantaviruses pose a serious threat to human health because their infection causes two highly fatal diseases, hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS). These pathogens are transmitted to humans through aerosolized excreta of their infected rodent hosts. Hantaviruses have a tripartite-segmented negative-sense RNA genome. The three genomic RNA segments, S, M, and L, encode a nucleocapsid protein (N), a precursor glycoprotein that is processed into two envelope glycoproteins (Gn and Gc) and the viral RNA-dependent RNA polymerase (RdRp), respectively. N protein is the major structural component of the virus, its main function is to protect and encapsidate the three genomic RNAs forming three viral ribonucleocapsids. Recent studies have proposed that N in conjunction with RdRp plays important roles in the transcription and replication of viral genome. In addition, N preferentially facilitates the translation of viral mRNA in cells. Glycoproteins, Gn and Gc, play major roles in viral attachment and entry to the host cells, virulence, and assembly and packaging of new virions in infected cells. RdRp functions as RNA replicase and transcriptase to replicate and transcribe the viral RNA and is also thought to have endonuclease activity. Currently, no antiviral therapy or vaccine is available for the treatment of hantavirus-associated diseases. Understanding the molecular details of hantavirus life cycle will help in the identification of targets for antiviral therapeutics and in the design of potential antiviral drug for the treatment of HFRS and HCPS. Due to the alarming fatality of hantavirus diseases, development of an effective vaccine against hantaviruses is a necessity.
Collapse
|
19
|
Paneth Iheozor-Ejiofor R, Levanov L, Hepojoki J, Strandin T, Lundkvist Å, Plyusnin A, Vapalahti O. Vaccinia virus-free rescue of fluorescent replication-defective vesicular stomatitis virus and pseudotyping with Puumala virus glycoproteins for use in neutralization tests. J Gen Virol 2016; 97:1052-1059. [PMID: 26916544 DOI: 10.1099/jgv.0.000437] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Puumala virus (PUUV) grows slowly in cell culture. To study antigenic properties of PUUV, an amenable method for their expression would be beneficial. To achieve this, a replication-defective recombinant vesicular stomatitis virus, rVSVΔG*EGFP, was rescued using BSRT7/5 and encephalomyocarditis virus (EMCV) internal ribosomal entry site (IRES)-enabled rescue plasmids. Using these particles, pseudotypes bearing PUUV Sotkamo strain glycoproteins were produced, with titres in the range 105-108, and were used in pseudotype focus reduction neutralization tests (pFRNTs) with neutralizing monoclonal antibodies and patient sera. The results were compared with those from orthodox focus reduction neutralization tests (oFRNTs) using native PUUV with the same samples and showed a strong positive correlation (rs = 0.82) between the methods. While developing the system we identified three amino acids which were mutated in the Vero E6 cell culture adapted PUUV prototype Sotkamo strain sequence, and changing these residues was critical for expression and neutralizing antibody binding of PUUV glycoproteins.
Collapse
Affiliation(s)
| | - Lev Levanov
- Department of Virology, Medicum, Helsinki, Finland
| | | | | | - Åke Lundkvist
- Department of Medical Biochemistry and Microbiology, Microbiology-Immunology, Uppsala University, Sweden
| | - Alexander Plyusnin
- Department of Virology, Medicum, Helsinki, Finland.,Department of Medical Biochemistry and Microbiology, Microbiology-Immunology, Uppsala University, Sweden
| | - Olli Vapalahti
- Department of Virology, Medicum, Helsinki, Finland.,Department of Virology and Immunology, HUSLAB, Helsinki University Hospital, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
Zivcec M, Metcalfe MG, Albariño CG, Guerrero LW, Pegan SD, Spiropoulou CF, Bergeron É. Assessment of Inhibitors of Pathogenic Crimean-Congo Hemorrhagic Fever Virus Strains Using Virus-Like Particles. PLoS Negl Trop Dis 2015; 9:e0004259. [PMID: 26625182 PMCID: PMC4666410 DOI: 10.1371/journal.pntd.0004259] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/03/2015] [Indexed: 11/18/2022] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is an often lethal, acute inflammatory illness that affects a large geographic area. The disease is caused by infection with CCHF virus (CCHFV), a nairovirus from the Bunyaviridae family. Basic research on CCHFV has been severely hampered by biosafety requirements and lack of available strains and molecular tools. We report the development of a CCHF transcription- and entry-competent virus-like particle (tecVLP) system that can be used to study cell entry and viral transcription/replication over a broad dynamic range (~4 orders of magnitude). The tecVLPs are morphologically similar to authentic CCHFV. Incubation of immortalized and primary human cells with tecVLPs results in a strong reporter signal that is sensitive to treatment with neutralizing monoclonal antibodies and by small molecule inhibitors of CCHFV. We used glycoproteins and minigenomes from divergent CCHFV strains to generate tecVLPs, and in doing so, we identified a monoclonal antibody that can prevent cell entry of tecVLPs containing glycoproteins from 3 pathogenic CCHFV strains. In addition, our data suggest that different glycoprotein moieties confer different cellular entry efficiencies, and that glycoproteins from the commonly used strain IbAr10200 have up to 100-fold lower ability to enter primary human cells compared to glycoproteins from pathogenic CCHFV strains.
Collapse
MESH Headings
- Antibodies, Monoclonal/metabolism
- Antibodies, Neutralizing/metabolism
- Antibodies, Viral/metabolism
- Drug Evaluation, Preclinical/methods
- Genes, Reporter
- Hemorrhagic Fever Virus, Crimean-Congo/genetics
- Hemorrhagic Fever Virus, Crimean-Congo/isolation & purification
- Hemorrhagic Fever Virus, Crimean-Congo/physiology
- Molecular Sequence Data
- Sequence Analysis, DNA
- Transcription, Genetic/drug effects
- Virion/genetics
- Virion/physiology
- Virion/ultrastructure
- Virus Internalization/drug effects
- Virus Replication/drug effects
Collapse
Affiliation(s)
- Marko Zivcec
- Viral Special Pathogens Branch, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Maureen G. Metcalfe
- Infectious Disease Pathology Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - César G. Albariño
- Viral Special Pathogens Branch, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lisa W. Guerrero
- Viral Special Pathogens Branch, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Scott D. Pegan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, United States of America
| | - Christina F. Spiropoulou
- Viral Special Pathogens Branch, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Éric Bergeron
- Viral Special Pathogens Branch, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
21
|
Abstract
In this chapter, we describe 73 zoonotic viruses that were isolated in Northern Eurasia and that belong to the different families of viruses with a single-stranded RNA (ssRNA) genome. The family includes viruses with a segmented negative-sense ssRNA genome (families Bunyaviridae and Orthomyxoviridae) and viruses with a positive-sense ssRNA genome (families Togaviridae and Flaviviridae). Among them are viruses associated with sporadic cases or outbreaks of human disease, such as hemorrhagic fever with renal syndrome (viruses of the genus Hantavirus), Crimean–Congo hemorrhagic fever (CCHFV, Nairovirus), California encephalitis (INKV, TAHV, and KHATV; Orthobunyavirus), sandfly fever (SFCV and SFNV, Phlebovirus), Tick-borne encephalitis (TBEV, Flavivirus), Omsk hemorrhagic fever (OHFV, Flavivirus), West Nile fever (WNV, Flavivirus), Sindbis fever (SINV, Alphavirus) Chikungunya fever (CHIKV, Alphavirus) and others. Other viruses described in the chapter can cause epizootics in wild or domestic animals: Geta virus (GETV, Alphavirus), Influenza A virus (Influenzavirus A), Bhanja virus (BHAV, Phlebovirus) and more. The chapter also discusses both ecological peculiarities that promote the circulation of these viruses in natural foci and factors influencing the occurrence of epidemic and epizootic outbreaks
Collapse
|
22
|
Mudhasani R, Kota KP, Retterer C, Tran JP, Whitehouse CA, Bavari S. High content image-based screening of a protease inhibitor library reveals compounds broadly active against Rift Valley fever virus and other highly pathogenic RNA viruses. PLoS Negl Trop Dis 2014; 8:e3095. [PMID: 25144302 PMCID: PMC4140764 DOI: 10.1371/journal.pntd.0003095] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/03/2014] [Indexed: 12/20/2022] Open
Abstract
High content image-based screening was developed as an approach to test a protease inhibitor small molecule library for antiviral activity against Rift Valley fever virus (RVFV) and to determine their mechanism of action. RVFV is the causative agent of severe disease of humans and animals throughout Africa and the Arabian Peninsula. Of the 849 compounds screened, 34 compounds exhibited ≥50% inhibition against RVFV. All of the hit compounds could be classified into 4 distinct groups based on their unique chemical backbone. Some of the compounds also showed broad antiviral activity against several highly pathogenic RNA viruses including Ebola, Marburg, Venezuela equine encephalitis, and Lassa viruses. Four hit compounds (C795-0925, D011-2120, F694-1532 and G202-0362), which were most active against RVFV and showed broad-spectrum antiviral activity, were selected for further evaluation for their cytotoxicity, dose response profile, and mode of action using classical virological methods and high-content imaging analysis. Time-of-addition assays in RVFV infections suggested that D011-2120 and G202-0362 targeted virus egress, while C795-0925 and F694-1532 inhibited virus replication. We showed that D011-2120 exhibited its antiviral effects by blocking microtubule polymerization, thereby disrupting the Golgi complex and inhibiting viral trafficking to the plasma membrane during virus egress. While G202-0362 also affected virus egress, it appears to do so by a different mechanism, namely by blocking virus budding from the trans Golgi. F694-1532 inhibited viral replication, but also appeared to inhibit overall cellular gene expression. However, G202-0362 and C795-0925 did not alter any of the morphological features that we examined and thus may prove to be good candidates for antiviral drug development. Overall this work demonstrates that high-content image analysis can be used to screen chemical libraries for new antivirals and to determine their mechanism of action and any possible deleterious effects on host cellular biology. Rift Valley fever (RVF) is an arthropod-borne viral zoonosis that occurs in large parts of sub-Saharan and North Africa and in 2000 emerged outside the African continent for the first time, raising concerns that it could further expand its geographical range. The disease in humans can result in encephalitis or hemorrhagic fever and in ruminants often results in abortion in pregnant females. Due to the lack of a licensed and commercially available vaccine, efforts to discover effective antiviral drugs are underway. Drug discovery using high content image-based screening is an effective tool that has been successfully used to identify new drugs. In this study, we developed an image-based assay to identify compounds active against RVF virus and other highly pathogenic human viruses. We demonstrated the usefulness of our image-based high content assay in identifying potential RVF antivirals by screening a small subset of chemical compounds for inhibition of RVF virus in a human cell line (HeLa) and partially characterized their mechanism of action within infected cells. The methods we developed in this study will be useful in discovering new effective drugs to combat Rift Valley fever.
Collapse
Affiliation(s)
- Rajini Mudhasani
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Krishna P. Kota
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Cary Retterer
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Julie P. Tran
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Chris A. Whitehouse
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Sina Bavari
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
23
|
Antigenic properties of N protein of hantavirus. Viruses 2014; 6:3097-109. [PMID: 25123683 PMCID: PMC4147688 DOI: 10.3390/v6083097] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/21/2014] [Accepted: 07/21/2014] [Indexed: 01/08/2023] Open
Abstract
Hantavirus causes two important rodent-borne viral zoonoses, hemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus pulmonary syndrome (HPS) in North and South America. Twenty-four species that represent sero- and genotypes have been registered within the genus Hantavirus by the International Committee on Taxonomy of Viruses (ICTV). Among the viral proteins, nucleocapsid (N) protein possesses an immunodominant antigen. The antigenicitiy of N protein is conserved compared with that of envelope glycoproteins. Therefore, N protein has been used for serological diagnoses and seroepidemiological studies. An understanding of the antigenic properties of N protein is important for the interpretation of results from serological tests using N antigen. N protein consists of about 430 amino acids and possesses various epitopes. The N-terminal quarter of N protein bears linear and immunodominant epitopes. However, a serotype-specific and multimerization-dependent antigenic site was found in the C-terminal half of N protein. In this paper, the structure, function, and antigenicity of N protein are reviewed.
Collapse
|
24
|
Hepojoki J, Strandin T, Hetzel U, Sironen T, Klingström J, Sane J, Mäkelä S, Mustonen J, Meri S, Lundkvist Å, Vapalahti O, Lankinen H, Vaheri A. Acute hantavirus infection induces galectin-3-binding protein. J Gen Virol 2014; 95:2356-2364. [PMID: 25013204 DOI: 10.1099/vir.0.066837-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hantaviruses are zoonotic viruses that cause life-threatening diseases when transmitted to humans. Severe hantavirus infection is manifested by impairment of renal function, pulmonary oedema and capillary leakage. Both innate and adaptive immune responses contribute to the pathogenesis, but the underlying mechanisms are not fully understood. Here, we showed that galectin-3-binding protein (Gal-3BP) was upregulated as a result of hantavirus infection both in vitro and in vivo. Gal-3BP is a secreted glycoprotein found in human serum, and increased Gal-3BP levels have been reported in chronic viral infections and in several types of cancer. Our in vitro experiments showed that, whilst Vero E6 cells (an African green monkey kidney cell line) constitutively expressed and secreted Gal-3BP, this protein was detected in primary human cells only as a result of hantavirus infection. Analysis of Gal-3BP levels in serum samples of cynomolgus macaques infected experimentally with hantavirus indicated that hantavirus infection induced Gal-3BP also in vivo. Finally, analysis of plasma samples collected from patients hospitalized because of acute hantavirus infection showed higher Gal-3BP levels during the acute than the convalescent phase. Furthermore, the Gal-3BP levels in patients with haemorrhagic fever with renal syndrome correlated with increased complement activation and with clinical variables reflecting the severity of acute hantavirus infection.
Collapse
Affiliation(s)
- Jussi Hepojoki
- Department of Virology, Peptide and Protein Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Tomas Strandin
- Department of Virology, Peptide and Protein Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Udo Hetzel
- Veterinary Pathology, Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Finland
| | - Tarja Sironen
- Department of Virology, Peptide and Protein Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska, University Hospital Huddinge, Stockholm, Sweden
| | - Jussi Sane
- Department of Virology, Peptide and Protein Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Satu Mäkelä
- School of Medicine, University of Tampere, Tampere, Finland.,Department of Internal Medicine, Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Jukka Mustonen
- School of Medicine, University of Tampere, Tampere, Finland.,Department of Internal Medicine, Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Åke Lundkvist
- Swedish Institute for Communicable Disease Control, Solna, Sweden
| | - Olli Vapalahti
- Department of Virology and Immunology, HUSLAB, Hospital District of Helsinki and Uusimaa, Finland.,Veterinary Pathology, Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Finland.,Department of Virology, Peptide and Protein Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Hilkka Lankinen
- Department of Virology, Peptide and Protein Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Antti Vaheri
- Department of Virology and Immunology, HUSLAB, Hospital District of Helsinki and Uusimaa, Finland.,Department of Virology, Peptide and Protein Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Kuivanen S, Hepojoki J, Vene S, Vaheri A, Vapalahti O. Identification of linear human B-cell epitopes of tick-borne encephalitis virus. Virol J 2014; 11:115. [PMID: 24946852 PMCID: PMC4078944 DOI: 10.1186/1743-422x-11-115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/13/2014] [Indexed: 12/30/2022] Open
Abstract
Background Tick-borne encephalitis (TBE) is a central nervous system infection transmitted to humans by ticks. The causative agent, tick-borne encephalitis virus (TBEV), belongs to the genus Flavivirus (family Flaviviridae), which includes globally important arthropod-borne viruses, such as dengue, Yellow fever, Japanese encephalitis and West Nile viruses. Flaviviruses are highly cross-reactive in serological tests that are currently based on viral envelope proteins. The envelope (E) protein is the major antigenic determinant and it is known to induce neutralizing antibody responses. Methods We synthesized the full-length TBEV proteome as overlapping synthetic 18-mer peptides to find dominant linear IgG epitopes. To distinguish natural TBEV infections from responses to TBE immunization or other flavivirus infections, the peptides were probed with sera of patients infected with TBEV, West Nile virus (WNV) or dengue virus (DENV), sera from TBE vaccinees and negative control sera by SPOT array technique. Results We identified novel linear TBEV IgG epitopes in the E protein and in the nonstructural protein 5 (NS5). Conclusions In this study, we screened TBEV structural and nonstructural proteins to find linear epitopes specific for TBEV. We found 11 such epitopes and characterized specifically two of them to be potential for differential diagnostics. This is the first report of identifying dominant linear human B-cell epitopes of the whole TBEV genome. The identified peptide epitopes have potential as antigens for diagnosing TBEV and to serologically distinguish flavivirus infections from each other.
Collapse
Affiliation(s)
- Suvi Kuivanen
- Department of Virology, Haartman Institute, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | | | | | | | | |
Collapse
|
26
|
Hantavirus Gn and Gc envelope glycoproteins: key structural units for virus cell entry and virus assembly. Viruses 2014; 6:1801-22. [PMID: 24755564 PMCID: PMC4014721 DOI: 10.3390/v6041801] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/20/2014] [Accepted: 03/31/2014] [Indexed: 01/24/2023] Open
Abstract
In recent years, ultrastructural studies of viral surface spikes from three different genera within the Bunyaviridae family have revealed a remarkable diversity in their spike organization. Despite this structural heterogeneity, in every case the spikes seem to be composed of heterodimers formed by Gn and Gc envelope glycoproteins. In this review, current knowledge of the Gn and Gc structures and their functions in virus cell entry and exit is summarized. During virus cell entry, the role of Gn and Gc in receptor binding has not yet been determined. Nevertheless, biochemical studies suggest that the subsequent virus-membrane fusion activity is accomplished by Gc. Further, a class II fusion protein conformation has been predicted for Gc of hantaviruses, and novel crystallographic data confirmed such a fold for the Rift Valley fever virus (RVFV) Gc protein. During virus cell exit, the assembly of different viral components seems to be established by interaction of Gn and Gc cytoplasmic tails (CT) with internal viral ribonucleocapsids. Moreover, recent findings show that hantavirus glycoproteins accomplish important roles during virus budding since they self-assemble into virus-like particles. Collectively, these novel insights provide essential information for gaining a more detailed understanding of Gn and Gc functions in the early and late steps of the hantavirus infection cycle.
Collapse
|
27
|
The role of viral genomic RNA and nucleocapsid protein in the autophagic clearance of hantavirus glycoprotein Gn. Virus Res 2014; 187:72-6. [PMID: 24412713 DOI: 10.1016/j.virusres.2013.12.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/18/2013] [Accepted: 12/24/2013] [Indexed: 01/24/2023]
Abstract
Hantaviruses have tri-segmented negative sense RNA genome. The viral M-segment RNA encodes a glycoprotein precursor (GPC), which is cleaved into two glycoprotein molecules Gn and Gc that form spikes on the viral envelope. We previously reported that Gn is degraded shortly after synthesis by the host autophagy machinery. However, Gn being an important integral component of the virion, must escape degradation during the packaging and assembly stage of virus replication cycle. The mechanism regulating the intrinsic steady-state levels of Gn during the course of virus replication cycle is not clear. We transfected cells with plasmids expressing viral S-segment RNA, nucleocapsid protein and glycoproteins Gn and Gc and monitored their expression levels over time. These studies revealed that accumulation of nucleocapsid protein, glycoprotein Gc and viral S-segment RNA helped to stabilize Gn. These observations suggest that initiation of virus assembly may help Gn to escape autophagic degradation by yet unknown mechanism.
Collapse
|
28
|
Hantavirus GnT elements mediate TRAF3 binding and inhibit RIG-I/TBK1-directed beta interferon transcription by blocking IRF3 phosphorylation. J Virol 2014; 88:2246-59. [PMID: 24390324 DOI: 10.1128/jvi.02647-13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hantaviruses successfully replicate in primary human endothelial cells by restricting the early induction of beta interferon (IFN-β) and interferon-stimulated genes (ISGs). Gn proteins from NY-1V, ANDV, and TULV, but not PHV, harbor elements in their 142-residue cytoplasmic tails (GnTs) that inhibit RIG-I/MAVS/TBK1-TRAF3-directed IFN-β induction. Here, we define GnT interactions and residues required to inhibit TRAF3-TBK1-directed IFN-β induction and IRF3 phosphorylation. We observed that GnTs bind TRAF3 via residues within the TRAF-N domain (residues 392 to 415) and that binding is independent of the MAVS-interactive TRAF-C domain (residues 415 to 568). We determined that GnT binding to TRAF3 is mediated by C-terminal degrons within NY-1V or ANDV GnTs and that mutations that add degrons to TULV or PHV GnTs confer TRAF3 binding. Further analysis of GnT domains revealed that TRAF3 binding is a discrete GnT function, independent of IFN regulation, and that residues 15 to 42 from the NY-1V GnT C terminus are required for inhibiting TBK1-directed IFN-β transcription. Mutagenesis of the NY-1V GnT revealed that altering tyrosine 627 (Y627A/S/F) abolished GnT regulation of RIG-I/TBK1-directed IRF3 phosphorylation and transcriptional responses of ISRE, κB, and IFN-β promoters. Moreover, GnTs from NY-1V, ANDV, and TULV, but not PHV, inhibited RIG-I-directed IRF3 phosphorylation. Collectively, these findings suggest a novel role for GnTs in regulating RIG-I/TBK1 pathway-directed IRF3 phosphorylation and IFN-β induction and define virulence determinants within GnTs that may permit the attenuation of pathogenic hantaviruses. IMPORTANCE These findings provide a mechanism for selected hantavirus GnT interactions to regulate RIG-I/TBK1 signaling responses required for IFN-β induction by inhibiting TBK1 phosphorylation of IRF3. These studies culminate in showing that a single GnT residue, Y627, is required for the NY-1V GnT to inhibit RIG-I/TBK1-directed IRF3 phosphorylation and IFN-β induction. These findings define a potential virulence determinant within the NY-1V GnT that may permit hantavirus attenuation.
Collapse
|
29
|
Abstract
ABSTRACT: Hantaviruses productively infect endothelial cells in their rodent reservoirs and humans, but the infection only causes disease in humans – hantavirus pulmonary syndrome and hemorrhagic fever with renal syndrome. Despite the enormous progress that has been made in understanding the pathogenesis and immune responses of hantavirus infection, there is a large gap in our molecular-based knowledge of hantaviral proteins in their structures, functions and the mechanisms that facilitate their entry, replication and assembly. Importantly, we know little about the specific viral determinants and viral protein–host interactions that drive differences noted in immune responses between the reservoir and humans. This review discusses our current understanding and future work needed for unraveling the biology of these viruses in their reservoirs and in humans.
Collapse
Affiliation(s)
- Ryan C McAllister
- Department of Pharmacology & Toxicology, University of Louisville, KY 40202, USA
- Center for Predictive Medicine for Biodefense & Emerging Infectious Diseases, KY, USA
| | - Colleen B Jonsson
- Department of Pharmacology & Toxicology, University of Louisville, KY 40202, USA
- Center for Predictive Medicine for Biodefense & Emerging Infectious Diseases, KY, USA
- Department of Microbiology and Immunology, University of Louisville, KY 40202, USA
- Departments of Microbiology & Immunology & Pharmacology & Toxicology, Center for Predictive Medicine for Biodefense & Emerging Infectious Diseases, University of Louisville, Clinical & Translational Research Building, 505 South Hancock Avenue, Louisville, KY 40202, USA
| |
Collapse
|
30
|
Hantavirus Gn and Gc glycoproteins self-assemble into virus-like particles. J Virol 2013; 88:2344-8. [PMID: 24335294 DOI: 10.1128/jvi.03118-13] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
How hantaviruses assemble and exit infected cells remains largely unknown. Here, we show that the expression of Andes (ANDV) and Puumala (PUUV) hantavirus Gn and Gc envelope glycoproteins lead to their self-assembly into virus-like particles (VLPs) which were released to cell supernatants. The viral nucleoprotein was not required for particle formation. Further, a Gc endodomain deletion mutant did not abrogate VLP formation. The VLPs were pleomorphic, exposed protrusions and reacted with patient sera.
Collapse
|
31
|
Carnec X, Ermonval M, Kreher F, Flamand M, Bouloy M. Role of the cytosolic tails of Rift Valley fever virus envelope glycoproteins in viral morphogenesis. Virology 2013; 448:1-14. [PMID: 24314631 DOI: 10.1016/j.virol.2013.09.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 08/05/2013] [Accepted: 09/25/2013] [Indexed: 11/30/2022]
Abstract
The correct folding, heterodimerization and trafficking of Gn/Gc envelope glycoproteins of Rift Valley fever virus, RVFV (Bunyaviridae and Phlebovirus genus) are essential for Golgi assembly and budding of viral particles. The Gn and Gc carboxy-terminus contain a Golgi targeting and an ER-retrieval signal, respectively. We generated RVFV-like particles with mutations in the cytosolic tails of Gn or Gc and identified regions important for release of infectious particles. The role of specific amino-acids in these regions was further investigated by creating recombinant mutant viruses by reverse-genetics. Residues outside the suspected Golgi targeting motif, i.e. the di-lysine K29-K30 motif and the N43, R44 and I46 residues of the Gn cytosolic domain, appeared important for Golgi localization and RNP packaging. Concerning the Gc tail, replacement of K2 or K3 in the di-lysine motif, had a drastic impact on Gn trafficking and induced an important organelle redistribution and cell remodeling, greatly affecting particle formation and release.
Collapse
Affiliation(s)
- Xavier Carnec
- Institut Pasteur, Unité de Génétique Moléculaire des Bunyavirus, 25 rue du Docteur Roux, 75015 Paris, France
| | | | | | | | | |
Collapse
|
32
|
Shimizu K, Yoshimatsu K, Koma T, Yasuda SP, Arikawa J. Role of nucleocapsid protein of hantaviruses in intracellular traffic of viral glycoproteins. Virus Res 2013; 178:349-56. [PMID: 24070985 DOI: 10.1016/j.virusres.2013.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/03/2023]
Abstract
To understand the role of nucleocapsid protein (NP) of hantaviruses in viral assembly, the effect of NP on intracellular traffic of viral glycoproteins Gn and Gc was investigated. Double staining of viral and host proteins in Hantaan virus (HTNV)-infected Vero E6 cells showed that Gn and Gc were localized to cis-Golgi, in which virus particles are thought to be formed. When HTNV Gn and Gc were expressed by a plasmid encoding glycoprotein precursor (GPC), which is posttranslationally cleaved into Gn and Gc, Gn was localized to cis-Golgi, whereas Gc showed diffuse distribution in the cytoplasm in 32.9% of Gc-positive cells. The ratio of the diffused Gc-positive cells was significantly decreased to 15.0% by co-expression of HTNV NP. Co-expression of HTNV GPC with NPs of other hantaviruses, such as Seoul virus, Puumala virus and Sin Nombre virus, also reduced the ratios of diffused Gc-positive cells to 13.5%, 25.2%, and 11.6%, respectively. Among amino- and carboxyl-terminally truncated HTNV NPs, NP75-429, NP116-429, NP1-333, NP1-233, and NP1-155 possessed activity to reduce the ratio of diffused Gc-positive cells, while NP155-429 and NP1-116 did not. NP30-429 has partial activity. These results indicate that amino acid region 116-155 of NP is important for the activity, although amino acid region 1-30 is partially related. Truncation of the HTNV Gc cytoplasmic tail caused an increase in diffused Gc-positive cells. In addition, the effect of coexpression of HTNV NP was weakened. These results suggest that HTNV NP has a role to promote Golgi localization of Gc through a mechanism possibly mediated by the Gc cytoplasmic tail.
Collapse
Affiliation(s)
- Kenta Shimizu
- Department of Microbiology, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | |
Collapse
|
33
|
Abstract
Hantaviruses are negative-sense single-stranded RNA viruses that infect many species of rodents, shrews, moles and bats. Infection in these reservoir hosts is almost asymptomatic, but some rodent-borne hantaviruses also infect humans, causing either haemorrhagic fever with renal syndrome (HFRS) or hantavirus cardiopulmonary syndrome (HCPS). In this Review, we discuss the basic molecular properties and cell biology of hantaviruses and offer an overview of virus-induced pathology, in particular vascular leakage and immunopathology.
Collapse
|
34
|
Epidemiology, molecular virology and diagnostics of Schmallenberg virus, an emerging orthobunyavirus in Europe. Vet Res 2013; 44:31. [PMID: 23675914 PMCID: PMC3663787 DOI: 10.1186/1297-9716-44-31] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 04/22/2013] [Indexed: 12/26/2022] Open
Abstract
After the unexpected emergence of Bluetongue virus serotype 8 (BTV-8) in northern Europe in 2006, another arbovirus, Schmallenberg virus (SBV), emerged in Europe in 2011 causing a new economically important disease in ruminants. The virus, belonging to the Orthobunyavirus genus in the Bunyaviridae family, was first detected in Germany, in The Netherlands and in Belgium in 2011 and soon after in the United Kingdom, France, Italy, Luxembourg, Spain, Denmark and Switzerland. This review describes the current knowledge on the emergence, epidemiology, clinical signs, molecular virology and diagnosis of SBV infection.
Collapse
|
35
|
Weissenhorn W, Poudevigne E, Effantin G, Bassereau P. How to get out: ssRNA enveloped viruses and membrane fission. Curr Opin Virol 2013; 3:159-67. [PMID: 23583788 PMCID: PMC7102784 DOI: 10.1016/j.coviro.2013.03.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 03/12/2013] [Accepted: 03/13/2013] [Indexed: 12/13/2022]
Abstract
Some but not all enveloped viruses recruit the ESCRT machinery for release. ESCRT-III assembles into spiral dome-like structures that together with VPS4 may catalyze membrane fission. ESCRT-independent influenza virus employs an amphipathic helix from M2 to catalyze fission. Glycoproteins from enveloped viruses such as flaviviridae assemble into an exterior protein coat-like structure, which may contribute to fission.
Enveloped viruses acquire their membrane from the host cell and accordingly need to separate their envelope from cellular membranes via membrane fission. Although some of the enveloped viruses recruit the endosomal sorting complex required for transport (ESCRT) to catalyze the final fission reaction, many enveloped viruses seem to bud in an ESCRT-independent manner. Here we describe the principles that govern membrane fission reactions in general and review progress in the understanding of ESCRT-mediated membrane fission. We relate ESCRT function to budding of single stranded RNA viruses and discuss alternative ways to mediate membrane fission that may govern ESCRT-independent budding.
Collapse
Affiliation(s)
- Winfried Weissenhorn
- Unit of Virus Host Cell Interactions (UVHCI), UMI 3265, Université Joseph Fourier-EMBL-CNRS, 6 rue Jules Horowitz, 38042 Grenoble Cedex 9, France.
| | | | | | | |
Collapse
|
36
|
Strandin T, Hepojoki J, Vaheri A. Cytoplasmic tails of bunyavirus Gn glycoproteins-Could they act as matrix protein surrogates? Virology 2013; 437:73-80. [PMID: 23357734 DOI: 10.1016/j.virol.2013.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 11/21/2012] [Accepted: 01/02/2013] [Indexed: 12/31/2022]
Abstract
Viruses of the family Bunyaviridae are negative-sense RNA viruses (NRVs). Unlike other NRVs bunyaviruses do not possess a matrix protein, which typically facilitates virus release from host cells and acts as an anchor between the viral membrane and its genetic core. Therefore the functions of matrix protein in bunyaviruses need to be executed by other viral proteins. In fact, the cytoplasmic tail of glycoprotein Gn (Gn-CT) of various bunyaviruses interacts with the genetic core (nucleocapsid protein and/or genomic RNA). In addition the Gn-CT of phleboviruses (a genus in the family Bunyaviridae) has been demonstrated to be essential for budding. This review brings together what is known on the role of various bunyavirus Gn-CTs in budding and assembly, and hypothesizes on their yet unrevealed functions in viral life cycle by comparing to the matrix proteins of NRVs.
Collapse
Affiliation(s)
- Tomas Strandin
- Department of Virology, Haartman Institute, P.O. Box 21, FI-00014, University of Helsinki, Finland.
| | | | | |
Collapse
|
37
|
Hantavirus regulation of type I interferon responses. Adv Virol 2012; 2012:524024. [PMID: 22924041 PMCID: PMC3423653 DOI: 10.1155/2012/524024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 05/18/2012] [Accepted: 07/04/2012] [Indexed: 11/22/2022] Open
Abstract
Hantaviruses primarily infect human endothelial cells (ECs) and cause two highly lethal human diseases. Early addition of Type I interferon (IFN) to ECs blocks hantavirus replication and thus for hantaviruses to be pathogenic they need to prevent early interferon induction. PHV replication is blocked in human ECs, but not inhibited in IFN deficient VeroE6 cells and consistent with this, infecting ECs with PHV results in the early induction of IFNβ and an array of interferon stimulated genes (ISGs). In contrast, ANDV, HTNV, NY-1V and TULV hantaviruses, inhibit early ISG induction and successfully replicate within human ECs. Hantavirus inhibition of IFN responses has been attributed to several viral proteins including regulation by the Gn proteins cytoplasmic tail (Gn-T). The Gn-T interferes with the formation of STING-TBK1-TRAF3 complexes required for IRF3 activation and IFN induction, while the PHV Gn-T fails to alter this complex or regulate IFN induction. These findings indicate that interfering with early IFN induction is necessary for hantaviruses to replicate in human ECs, and suggest that additional determinants are required for hantaviruses to be pathogenic. The mechanism by which Gn-Ts disrupt IFN signaling is likely to reveal potential therapeutic interventions and suggest protein targets for attenuating hantaviruses.
Collapse
|
38
|
Structure, function, and evolution of the Crimean-Congo hemorrhagic fever virus nucleocapsid protein. J Virol 2012; 86:10914-23. [PMID: 22875964 DOI: 10.1128/jvi.01555-12] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is an emerging tick-borne virus of the Bunyaviridae family that is responsible for a fatal human disease for which preventative or therapeutic measures do not exist. We solved the crystal structure of the CCHFV strain Baghdad-12 nucleocapsid protein (N), a potential therapeutic target, at a resolution of 2.1 Å. N comprises a large globular domain composed of both N- and C-terminal sequences, likely involved in RNA binding, and a protruding arm domain with a conserved DEVD caspase-3 cleavage site at its apex. Alignment of our structure with that of the recently reported N protein from strain YL04057 shows a close correspondence of all folds but significant transposition of the arm through a rotation of 180 degrees and a translation of 40 Å. These observations suggest a structural flexibility that may provide the basis for switching between alternative N protein conformations during important functions such as RNA binding and oligomerization. Our structure reveals surfaces likely involved in RNA binding and oligomerization, and functionally critical residues within these domains were identified using a minigenome system able to recapitulate CCHFV-specific RNA synthesis in cells. Caspase-3 cleaves the polypeptide chain at the exposed DEVD motif; however, the cleaved N protein remains an intact unit, likely due to the intimate association of N- and C-terminal fragments in the globular domain. Structural alignment with existing N proteins reveals that the closest CCHFV relative is not another bunyavirus but the arenavirus Lassa virus instead, suggesting that current segmented negative-strand RNA virus taxonomy may need revision.
Collapse
|
39
|
Kurolt IC, Paessler S, Markotić A. Resequencing of the Puumala virus strain Sotkamo from the WHO Arbovirus collection. Virus Genes 2012; 45:389-92. [PMID: 22798055 DOI: 10.1007/s11262-012-0780-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
Abstract
RNA viruses exhibit a high mutation rate as the RNA-dependent RNA polymerase lacks proofreading and repair capabilities. It is known that serial passaging on cell culture leads to virus adaptation. Puumala virus (PUUV) strain Sotkamo is the prototype virus for the low-pathogenic hantavirus Puumala, family Bunyaviridae. A full-length sequence of the strain Sotkamos tripartite genome was made available more than 15 years ago, after at least 15 passages on Vero E6 cells. A distinct sample from the sequenced strain, with unknown passage history, was then included in the WHO Arbovirus collection. The genome sequence of this included sample was determined in this study and exhibited over 99 % identity in comparison to the previously published sequence. A total of 23 nucleotide changes across all genome segments were found. The small segment had the highest nucleotide variance without changes on the protein level. Within the extraviral domain of the glycoproteins, the majority of non-synonymous mutations were detected, whereas the large segment is most conserved on the nucleotide level. It seems possible that the PUUV strain Sotkamo adapted differently to serial passaging on cell culture in two different laboratories. In addition, a distinct passage number could exhibit itself within the nucleotide differences.
Collapse
Affiliation(s)
- Ivan C Kurolt
- Research Department, University Hospital for Infectious Diseases "Dr. Fran Mihaljević", Mirogojska 8, 10000 Zagreb, Croatia.
| | | | | |
Collapse
|
40
|
Intraviral protein interactions of Chandipura virus. Arch Virol 2012; 157:1949-57. [PMID: 22763614 DOI: 10.1007/s00705-012-1389-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 05/15/2012] [Indexed: 10/28/2022]
Abstract
Chandipura virus (CHPV) is an emerging rhabdovirus responsible for several outbreaks of fatal encephalitis among children in India. The characteristic structure of the virus is a result of extensive and specific interplay among its five encoded proteins. The revelation of interactions among CHPV proteins can help in gaining insight into viral architecture and pathogenesis. In the current study, we carried out comprehensive yeast two-hybrid (Y2H) analysis to elucidate intraviral protein-protein interactions. All of the interactions identified by Y2H were assessed for reliability by GST pull-down and ELISA. A total of eight interactions were identified among four viral proteins. Five of these interactions are being reported for the first time for CHPV. Among these, the glycoprotein (G)-nucleocapsid (N) interaction could be considered novel, as this has not been reported for any members of the family Rhabdoviridae. This study provides a framework within which the roles of the identified protein interactions can be explored further for understanding the biology of this virus at the molecular level.
Collapse
|
41
|
Hussein ITM, Cheng E, Ganaie SS, Werle MJ, Sheema S, Haque A, Mir MA. Autophagic clearance of Sin Nombre hantavirus glycoprotein Gn promotes virus replication in cells. J Virol 2012; 86:7520-9. [PMID: 22553339 PMCID: PMC3416297 DOI: 10.1128/jvi.07204-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/24/2012] [Indexed: 11/20/2022] Open
Abstract
Hantavirus glycoprotein precursor (GPC) is posttranslationally cleaved into two glycoproteins, Gn and Gc. Cells transfected with plasmids expressing either GPC or both Gn and Gc revealed that Gn is posttranslationally degraded. Treatment of cells with the autophagy inhibitors 3-methyladenine, LY-294002, or Wortmanin rescued Gn degradation, suggesting that Gn is degraded by the host autophagy machinery. Confocal microscopic imaging showed that Gn is targeted to autophagosomes for degradation by an unknown mechanism. Examination of autophagy markers LC3-I and LC3-II demonstrated that both Gn expression and Sin Nombre hantavirus (SNV) infection induce autophagy in cells. To delineate whether induction of autophagy and clearance of Gn play a role in the virus replication cycle, we downregulated autophagy genes BCLN-1 and ATG7 using small interfering RNA (siRNA) and monitored virus replication over time. These studies revealed that inhibition of host autophagy machinery inhibits Sin Nombre virus replication in cells, suggesting that autophagic clearance of Gn is required for efficient virus replication. Our studies provide mechanistic insights into viral pathogenesis and reveal that SNV exploits the host autophagy machinery to decrease the intrinsic steady-state levels of an important viral component for efficient replication in host cells.
Collapse
Affiliation(s)
- Islam T M Hussein
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Hepojoki J, Strandin T, Lankinen H, Vaheri A. Hantavirus structure--molecular interactions behind the scene. J Gen Virol 2012; 93:1631-1644. [PMID: 22622328 DOI: 10.1099/vir.0.042218-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Viruses of the genus Hantavirus, carried and transmitted by rodents and insectivores, are the exception in the vector-borne virus family Bunyaviridae, since viruses of the other genera are transmitted via arthropods. The single-stranded, negative-sense, RNA genome of hantaviruses is trisegmented into small, medium and large (S, M and L) segments. The segments, respectively, encode three structural proteins: nucleocapsid (N) protein, two glycoproteins Gn and Gc and an RNA-dependent RNA-polymerase. The genome segments, encapsidated by the N protein to form ribonucleoproteins, are enclosed inside a lipid envelope that is decorated by spikes composed of Gn and Gc. The virion displays round or pleomorphic morphology with a diameter of roughly 120-160 nm depending on the detection method. This review focuses on the structural components of hantaviruses, their interactions, the mechanisms behind virion assembly and the interactions that maintain virion integrity. We attempt to summarize recent results on the virion structure and to suggest mechanisms on how the assembly is driven. We also compare hantaviruses to other bunyaviruses with known structure.
Collapse
Affiliation(s)
- Jussi Hepojoki
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Tomas Strandin
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Hilkka Lankinen
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| |
Collapse
|
43
|
Estrada DF, Conner M, Jeor SC, Guzman RND. The Structure of the Hantavirus Zinc Finger Domain is Conserved and Represents the Only Natively Folded Region of the Gn Cytoplasmic Tail. Front Microbiol 2011; 2:251. [PMID: 22203819 PMCID: PMC3243910 DOI: 10.3389/fmicb.2011.00251] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 11/27/2011] [Indexed: 11/25/2022] Open
Abstract
Hantaviruses, of the family Bunyaviridae, are present throughout the world and cause a variety of infections ranging from the asymptomatic to mild and severe hemorrhagic fevers. Hantaviruses are enveloped anti-sense RNA viruses that contain three genomic segments that encode for a nucleocapsid protein, two membrane glycoproteins (Gn and Gc), and an RNA polymerase. Recently, the pathogenicity of hantaviruses has been mapped to the carboxyl end of the 150 residue Gn cytoplasmic tail. The Gn tail has also been shown to play a role in binding the ribonucleoprotein (RNP), a step critical for virus assembly. In this study, we use NMR spectroscopy to compare the structure of a Gn tail zinc finger domain of both a pathogenic (Andes) and a non-pathogenic (Prospect Hill) hantavirus. We demonstrate that despite a stark difference in the virulence of both of these viruses, the structure of the Gn core zinc finger domain is largely conserved in both strains. We also use NMR backbone relaxation studies to demonstrate that the regions of the Andes virus Gn tail immediately outside the zinc finger domain, sites known to bind the RNP, are disordered and flexible, thus intimating that the zinc finger domain is the only structured region of the Gn tail. These structural observations provide further insight into the role of the Gn tail during viral assembly as well as its role in pathogenesis.
Collapse
Affiliation(s)
- D Fernando Estrada
- Department of Molecular Biosciences, University of Kansas Lawrence, KS, USA
| | | | | | | |
Collapse
|
44
|
Walter CT, Barr JN. Recent advances in the molecular and cellular biology of bunyaviruses. J Gen Virol 2011; 92:2467-2484. [PMID: 21865443 DOI: 10.1099/vir.0.035105-0] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The family Bunyaviridae of segmented, negative-stranded RNA viruses includes over 350 members that infect a bewildering variety of animals and plants. Many of these bunyaviruses are the causative agents of serious disease in their respective hosts, and are classified as emerging viruses because of their increased incidence in new populations and geographical locations throughout the world. Emerging bunyaviruses, such as Crimean-Congo hemorrhagic fever virus, tomato spotted wilt virus and Rift Valley fever virus, are currently attracting great interest due to migration of their arthropod vectors, a situation possibly linked to climate change. These and other examples of continued emergence suggest that bunyaviruses will probably continue to pose a sustained global threat to agricultural productivity, animal welfare and human health. The threat of emergence is particularly acute in light of the lack of effective preventative or therapeutic treatments for any of these viruses, making their study an important priority. This review presents recent advances in the understanding of the bunyavirus life cycle, including aspects of their molecular, cellular and structural biology. Whilst special emphasis is placed upon the emerging bunyaviruses, we also describe the extensive body of work involving model bunyaviruses, which have been the subject of major contributions to our overall understanding of this important group of viruses.
Collapse
Affiliation(s)
- Cheryl T Walter
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| | - John N Barr
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| |
Collapse
|
45
|
Strandin T, Hepojoki J, Wang H, Vaheri A, Lankinen H. The cytoplasmic tail of hantavirus Gn glycoprotein interacts with RNA. Virology 2011; 418:12-20. [PMID: 21807393 PMCID: PMC7172371 DOI: 10.1016/j.virol.2011.06.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 06/06/2011] [Accepted: 06/16/2011] [Indexed: 11/15/2022]
Abstract
We recently characterized the interaction between the intraviral domains of envelope glycoproteins (Gn and Gc) and ribonucleoprotein (RNP) of Puumala and Tula hantaviruses (genus Hantavirus, family Bunyaviridae). Herein we report a direct interaction between spike-forming glycoprotein and nucleic acid. We show that the envelope glycoprotein Gn of hantaviruses binds genomic RNA through its cytoplasmic tail (CT). The nucleic acid binding of Gn-CT is unspecific, as demonstrated by interactions with unrelated RNA and with single-stranded DNA. Peptide scan and protein deletions of Gn-CT mapped the nucleic acid binding to regions that overlap with the previously characterized N protein binding sites and demonstrated the carboxyl-terminal part of Gn-CT to be the most potent nucleic acid-binding site. We conclude that recognition of the RNP complex by the Gn-CT could be mediated by interactions with both genomic RNA and the N protein. This would provide the required selectivity for the genome packaging of hantaviruses.
Collapse
Affiliation(s)
- Tomas Strandin
- Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014, University of Helsinki, Finland.
| | | | | | | | | |
Collapse
|
46
|
Strandin T, Hepojoki J, Wang H, Vaheri A, Lankinen H. Inactivation of hantaviruses by N-ethylmaleimide preserves virion integrity. J Gen Virol 2011; 92:1189-1198. [DOI: 10.1099/vir.0.027896-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Thiol groups of cysteine residues are crucial for the infectivity of various enveloped viruses, but their role in the infectivity of viruses of the family Bunyaviridae has thus far not been studied. This report shows that thiol groups are essential to the infectivity of hantaviruses. Alkylation of the thiol functional groups using the membrane-permeable compound N-ethylmaleimide (NEM) and membrane-impermeable compound 5,5′-dithio-bis-(2-nitrobenzoic acid) (DTNB) showed NEM to be a highly effective inactivator of Puumala and Tula hantaviruses. The NEM-inactivated hantavirus maintained the buoyant density of the wild-type virus. Furthermore, the antigenicity of glycoproteins and the cell attachment capacity of virions were retained at NEM concentrations that totally abolished virus infectivity. These results signified preservation of virion integrity following inactivation with NEM, making chemically inactivated virions valuable research antigens. It was demonstrated with biotin-conjugated maleimide, a mechanistic analogue of NEM, that all the structural proteins of hantavirus were sensitive towards thiol alkylation. In contrast to hantaviruses, NEM did not abolish Uukuniemi phlebovirus infectivity to the same extent. This indicates differences in the use of free thiols in virus entry among members of the family Bunyaviridae.
Collapse
Affiliation(s)
- Tomas Strandin
- Department of Virology, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
- Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
- Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
| | - Hao Wang
- Department of Virology, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
| | - Hilkka Lankinen
- Department of Virology, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
- Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014 University of Helsinki, Finland
| |
Collapse
|
47
|
Estrada DF, De Guzman RN. Structural characterization of the Crimean-Congo hemorrhagic fever virus Gn tail provides insight into virus assembly. J Biol Chem 2011; 286:21678-86. [PMID: 21507948 DOI: 10.1074/jbc.m110.216515] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The RNA virus that causes the Crimean Congo Hemorrhagic Fever (CCHF) is a tick-borne pathogen of the Nairovirus genus, family Bunyaviridae. Unlike many zoonotic viruses that are only passed between animals and humans, the CCHF virus can also be transmitted from human to human with an overall mortality rate approaching 30%. Currently, there are no atomic structures for any CCHF virus proteins or for any Nairovirus proteins. A critical component of the virus is the envelope Gn glycoprotein, which contains a C-terminal cytoplasmic tail. In other Bunyaviridae viruses, the Gn tail has been implicated in host-pathogen interaction and viral assembly. Here we report the NMR structure of the CCHF virus Gn cytoplasmic tail, residues 729-805. The structure contains a pair of tightly arranged dual ββα zinc fingers similar to those found in the Hantavirus genus, with which it shares about 12% sequence identity. Unlike Hantavirus zinc fingers, however, the CCHF virus zinc fingers bind viral RNA and contain contiguous clusters of conserved surface electrostatics. Our results provide insight into a likely role of the CCHF virus Gn zinc fingers in Nairovirus assembly.
Collapse
Affiliation(s)
- D Fernando Estrada
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, USA
| | | |
Collapse
|
48
|
Klingström J, Ahlm C. Hantavirus protein interactions regulate cellular functions and signaling responses. Expert Rev Anti Infect Ther 2011; 9:33-47. [PMID: 21171876 DOI: 10.1586/eri.10.157] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rodent-borne pathogenic hantaviruses cause two severe and often lethal zoonotic diseases: hemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus cardiopulmonary syndrome (HCPS) in the Americas. Currently, no US FDA-approved therapeutics or vaccines are available for HFRS/HCPS. Infections with hantaviruses are not lytic, and it is currently not known exactly why infections in humans cause disease. A better understanding of how hantaviruses interfere with normal cell functions and activation of innate and adaptive immune responses might provide clues to future development of specific treatment and/or vaccines against hantavirus infection. In this article, the current knowledge regarding immune responses observed in patients, hantavirus interference with cellular proteins and signaling pathways, and possible approaches in the development of therapeutics are discussed.
Collapse
Affiliation(s)
- Jonas Klingström
- Centre for Microbiological Preparedness, Swedish Institute for Infectious Disease Control, Solna, Sweden.
| | | |
Collapse
|
49
|
The C-terminal 42 residues of the Tula virus Gn protein regulate interferon induction. J Virol 2011; 85:4752-60. [PMID: 21367904 DOI: 10.1128/jvi.01945-10] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Hantaviruses primarily infect the endothelial cell lining of capillaries and cause two vascular permeability-based diseases. The ability of pathogenic hantaviruses to regulate the early induction of interferon determines whether hantaviruses replicate in endothelial cells. Tula virus (TULV) and Prospect Hill virus (PHV) are hantaviruses which infect human endothelial cells but fail to cause human disease. PHV is unable to inhibit early interferon (IFN) responses and fails to replicate within human endothelial cells. However, TULV replicates successfully in human endothelial cells, suggesting that TULV is capable of regulating cellular IFN responses. We observed a >300-fold reduction in the IFN-stimulated genes (ISGs) MxA and ISG56 following TULV versus PHV infection of endothelial cells 1 day postinfection. Similar to results with pathogenic hantaviruses, expressing the TULV Gn protein cytoplasmic tail (Gn-T) blocked RIG-I- and TBK1-directed transcription from IFN-stimulated response elements (ISREs) and IFN-β promoters (>90%) but not transcription directed by constitutively active IFN regulatory factor-3 (IRF3). In contrast, expressing the PHV Gn-T had no effect on TBK1-induced transcriptional responses. Analysis of Gn-T truncations demonstrated that the C-terminal 42 residues of the Gn-T (Gn-T-C42) from TULV, but not PHV, inhibited IFN induction >70%. These findings demonstrate that the TULV Gn-T inhibits IFN- and ISRE-directed responses upstream of IRF3 at the level of the TBK1 complex and further define a 42-residue domain of the TULV Gn-T that inhibits IFN induction. In contrast to pathogenic hantavirus Gn-Ts, the TULV Gn-T lacks a C-terminal degron domain and failed to bind tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3), a TBK1 complex component required for IRF3 activation. These findings indicate that the nonpathogenic TULV Gn-T regulates IFN induction but accomplishes this via unique interactions with cellular TBK1 complexes. These findings fundamentally distinguish nonpathogenic hantaviruses, PHV and TULV, and demonstrate that IFN regulation alone is insufficient for hantaviruses to cause disease. Yet regulating the early IFN response is necessary for hantaviruses to replicate within human endothelial cells and to be pathogenic. Thus, in addition to IFN regulation, hantaviruses contain discrete virulence determinants which permit them to be human pathogens.
Collapse
|
50
|
Li J, Zhang Q, Wang T, Li C, Liang M, Li D. Tracking hantavirus nucleocapsid protein using intracellular antibodies. Virol J 2010; 7:339. [PMID: 21092325 PMCID: PMC3002308 DOI: 10.1186/1743-422x-7-339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 11/24/2010] [Indexed: 11/30/2022] Open
Abstract
Background Hantavirus nucleocapsid (N) protein is a multifunctional viral macromolecule involved in multiple stages of the viral replication cycle. The intracellular trafficking of N protein during virus assembly remains unclear. Methods We used N protein-specific intracellular expressed antibodies to track the localization and distribution of Hantaan virus and Seoul virus N protein. The N protein-specific antibody single-chain variable antibody fragments (scFvs), which bind an N-terminal linear epitope (L13F3) and C-terminal conformational domain (H34), were intracellularly expressed in the endoplasmic reticulum (ER) by fusion of the SEKDEL retention signal peptide at the carboxyl terminus, and in the cytoplasm (Cyto) by deletion of the ER membrane target signal peptide. Stable Vero-E6 cell lines expressing intracellular scFvs were either infected with hantavirus or transfected with an N protein expression plasmid; virus replication and N protein intracellular localization were determined. Result N protein co-localized with scFvs in the ER and cytoplasm with or without viral membrane glycoproteins. Hantavirus replication was inhibited in both the scFvs-ER- and scFvs-Cyto-expressing stable cell lines. Conclusion N protein may be expressed in the ER retention signal peptide of KDEL circulating region (ER/cis-Golgi) without the assistance of G protein, and so expression of N protein in both the cytoplasm and within the ER/cis-Golgi plays an important role in virus replication.
Collapse
Affiliation(s)
- Jiandong Li
- State Key Laboratory, Molecular Virology and Genetic Engineering, Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing 102206, PR China
| | | | | | | | | | | |
Collapse
|