51
|
Oyarzun P, Ellis JJ, Gonzalez-Galarza FF, Jones AR, Middleton D, Boden M, Kobe B. A bioinformatics tool for epitope-based vaccine design that accounts for human ethnic diversity: application to emerging infectious diseases. Vaccine 2015; 33:1267-73. [PMID: 25629524 DOI: 10.1016/j.vaccine.2015.01.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/11/2014] [Accepted: 01/14/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Peptide vaccination based on multiple T-cell epitopes can be used to target well-defined ethnic populations. Because the response to T-cell epitopes is restricted by HLA proteins, the HLA specificity of T-cell epitopes becomes a major consideration for epitope-based vaccine design. We have previously shown that CD4+ T-cell epitopes restricted by 95% of human MHC class II proteins can be predicted with high-specificity. METHODS We describe here the integration of epitope prediction with population coverage and epitope selection algorithms. The population coverage assessment makes use of the Allele Frequency Net Database. We present the computational platform Predivac-2.0 for HLA class II-restricted epitope-based vaccine design, which accounts comprehensively for human genetic diversity. RESULTS We validated the performance of the tool on the identification of promiscuous and immunodominant CD4+ T-cell epitopes from the human immunodeficiency virus (HIV) protein Gag. We further describe an application for epitope-based vaccine design in the context of emerging infectious diseases associated with Lassa, Nipah and Hendra viruses. Putative CD4+ T-cell epitopes were mapped on the surface glycoproteins of these pathogens and are good candidates to be experimentally tested, as they hold potential to provide cognate help in vaccination settings in their respective target populations. CONCLUSION Predivac-2.0 is a novel approach in epitope-based vaccine design, particularly suited to be applied to virus-related emerging infectious diseases, because the geographic distributions of the viruses are well defined and ethnic populations in need of vaccination can be determined ("ethnicity-oriented approach"). Predivac-2.0 is accessible through the website http://predivac.biosci.uq.edu.au/.
Collapse
Affiliation(s)
- Patricio Oyarzun
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia; Biotechnology Centre, Universidad San Sebastián, Concepción, Chile.
| | - Jonathan J Ellis
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia
| | | | - Andrew R Jones
- Institute of Integrative Biology, University of Liverpool, United Kingdom
| | - Derek Middleton
- Transplant Immunology Laboratory, Royal Liverpool University Hospital & School of Infection and Host Defence University of Liverpool, United Kingdom
| | - Mikael Boden
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia; School of Information Technology and Electrical Engineering, University of Queensland, Queensland 4072, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia.
| |
Collapse
|
52
|
Abstract
Hendra virus infection of horses occurred sporadically between 1994 and 2010 as a result of spill-over from the viral reservoir in Australian mainland flying-foxes, and occasional onward transmission to people also followed from exposure to affected horses. An unprecedented number of outbreaks were recorded in 2011 leading to heightened community concern. Release of an inactivated subunit vaccine for horses against Hendra virus represents the first commercially available product that is focused on mitigating the impact of a Biosafety Level 4 pathogen. Through preventing the development of acute Hendra virus disease in horses, vaccine use is also expected to reduce the risk of transmission of infection to people.
Collapse
Affiliation(s)
- Deborah Middleton
- Australian Animal Health Laboratory, CSIRO, PB 24, Geelong, Victoria 3220, Australia.
| |
Collapse
|
53
|
Sawatsky B, McAuley AJ, Holbrook MR, Bente DA. Comparative pathogenesis of Alkhumra hemorrhagic fever and Kyasanur forest disease viruses in a mouse model. PLoS Negl Trop Dis 2014; 8:e2934. [PMID: 24922308 PMCID: PMC4055546 DOI: 10.1371/journal.pntd.0002934] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 04/28/2014] [Indexed: 11/19/2022] Open
Abstract
Kyasanur Forest disease virus (KFDV) and Alkhumra hemorrhagic fever virus (AHFV) are genetically closely-related, tick-borne flaviviruses that cause severe, often fatal disease in humans. Flaviviruses in the tick-borne encephalitis (TBE) complex typically cause neurological disease in humans whereas patients infected with KFDV and AHFV predominately present with hemorrhagic fever. A small animal model for KFDV and AHFV to study the pathogenesis and evaluate countermeasures has been lacking mostly due to the need of a high biocontainment laboratory to work with the viruses. To evaluate the utility of an existing mouse model for tick-borne flavivirus pathogenesis, we performed serial sacrifice studies in BALB/c mice infected with either KFDV strain P9605 or AHFV strain Zaki-1. Strikingly, infection with KFDV was completely lethal in mice, while AHFV caused no clinical signs of disease and no animals succumbed to infection. KFDV and high levels of pro-inflammatory cytokines were detected in the brain at later time points, but no virus was found in visceral organs; conversely, AHFV Zaki-1 and elevated levels of cytokines were found in the visceral organs at earlier time points, but were not detected in the brain. While infection with either virus caused a generalized leukopenia, only AHFV Zaki-1 induced hematologic abnormalities in infected animals. Our data suggest that KFDV P9605 may have lost its ability to cause hemorrhagic disease as the result of multiple passages in suckling mouse brains. However, likely by virtue of fewer mouse passages, AHFV Zaki-1 has retained the ability to replicate in visceral organs, cause hematologic abnormalities, and induce pro-inflammatory cytokines without causing overt disease. Given these striking differences, the use of inbred mice and the virus passage history need to be carefully considered in the interpretation of animal studies using these viruses. Kyasanur Forest disease virus (KFDV) and Alkhumra hemorrhagic fever virus (AHFV) are tick-borne flaviviruses that cause severe hemorrhagic disease in humans. The pathogenesis of the disease is still not very well understood mostly due to the lack of suitable animal models. Despite sharing a high degree of genetic sequence similarity, KFDV replicates primarily in the brain and is uniformly lethal for BALB/c mice. In contrast, AHFV does not cause clinically overt signs in mice, replicates in the visceral organs, and induces pro-inflammatory cytokines and hematological changes. Given the striking differences in pathogenesis and tissue tropism, the use of inbred mice as well as the passage history of the virus needs to be carefully considered in the interpretation of animal studies using these viruses.
Collapse
Affiliation(s)
- Bevan Sawatsky
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Alexander J. McAuley
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michael R. Holbrook
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, Untied States of America
- National Institute of Allergy and Infectious Diseases, Integrated Research Facility, Frederick, Maryland, United States of America
| | - Dennis A. Bente
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
54
|
DeBuysscher BL, Scott D, Marzi A, Prescott J, Feldmann H. Single-dose live-attenuated Nipah virus vaccines confer complete protection by eliciting antibodies directed against surface glycoproteins. Vaccine 2014; 32:2637-44. [PMID: 24631094 DOI: 10.1016/j.vaccine.2014.02.087] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/12/2014] [Accepted: 02/26/2014] [Indexed: 11/24/2022]
Abstract
BACKGROUND Nipah virus (NiV), a zoonotic pathogen causing severe respiratory illness and encephalitis in humans, emerged in Malaysia in 1998 with subsequent outbreaks on an almost annual basis since 2001 in parts of the Indian subcontinent. The high case fatality rate, human-to-human transmission, wide-ranging reservoir distribution and lack of licensed intervention options are making NiV a serious regional and potential global public health problem. The objective of this study was to develop a fast-acting, single-dose NiV vaccine that could be implemented in a ring vaccination approach during outbreaks. METHODS In this study we have designed new live-attenuated vaccine vectors based on recombinant vesicular stomatitis viruses (rVSV) expressing NiV glycoproteins (G or F) or nucleoprotein (N) and evaluated their protective efficacy in Syrian hamsters, an established NiV animal disease model. We further characterized the humoral immune response to vaccination in hamsters using ELISA and neutralization assays and performed serum transfer studies. RESULTS Vaccination of Syrian hamsters with a single dose of the rVSV vaccine vectors resulted in strong humoral immune responses with neutralizing activities found only in those animals vaccinated with rVSV expressing NiV G or F proteins. Vaccinated animals with neutralizing antibody responses were completely protected from lethal NiV disease, whereas animals vaccinated with rVSV expressing NiV N showed only partial protection. Protection of NiV G or F vaccinated animals was conferred by antibodies, most likely the neutralizing fraction, as demonstrated by serum transfer studies. Protection of N-vaccinated hamsters was not antibody-dependent indicating a role of adaptive cellular responses for protection. CONCLUSIONS The rVSV vectors expressing Nipah virus G or F are prime candidates for new 'emergency vaccines' to be utilized for NiV outbreak management.
Collapse
Affiliation(s)
- Blair L DeBuysscher
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA; Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Dana Scott
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Joseph Prescott
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA.
| |
Collapse
|
55
|
Middleton D, Pallister J, Klein R, Feng YR, Haining J, Arkinstall R, Frazer L, Huang JA, Edwards N, Wareing M, Elhay M, Hashmi Z, Bingham J, Yamada M, Johnson D, White J, Foord A, Heine HG, Marsh GA, Broder CC, Wang LF. Hendra virus vaccine, a one health approach to protecting horse, human, and environmental health. Emerg Infect Dis 2014; 20:372-9. [PMID: 24572697 PMCID: PMC3944873 DOI: 10.3201/eid2003.131159] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In recent years, the emergence of several highly pathogenic zoonotic diseases in humans has led to a renewed emphasis on the interconnectedness of human, animal, and environmental health, otherwise known as One Health. For example, Hendra virus (HeV), a zoonotic paramyxovirus, was discovered in 1994, and since then, infections have occurred in 7 humans, each of whom had a strong epidemiologic link to similarly affected horses. As a consequence of these outbreaks, eradication of bat populations was discussed, despite their crucial environmental roles in pollination and reduction of the insect population. We describe the development and evaluation of a vaccine for horses with the potential for breaking the chain of HeV transmission from bats to horses to humans, thereby protecting horse, human, and environmental health. The HeV vaccine for horses is a key example of a One Health approach to the control of human disease.
Collapse
Affiliation(s)
| | | | - Reuben Klein
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Yan-Ru Feng
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Jessica Haining
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Rachel Arkinstall
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Leah Frazer
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Jin-An Huang
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Nigel Edwards
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Mark Wareing
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Martin Elhay
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Zia Hashmi
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - John Bingham
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Manabu Yamada
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Dayna Johnson
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - John White
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Adam Foord
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Hans G. Heine
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Glenn A. Marsh
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Christopher C. Broder
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| | - Lin-Fa Wang
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria, Australia (D. Middleton, J. Pallister, R. Klein, J. Haining, R. Arkinstall, L. Frazer, J. Bingham, D. Johnson, J. White, A. Foord, H.G. Heine, G.A. Marsh, L.-F. Wang)
- Uniformed Services University, Bethesda, Maryland, USA (Y.-R. Feng, C.C. Broder); Zoetis Research & Manufacturing Pty Ltd, Parkville, Victoria, Australia (J.-A. Huang, N. Edwards, M. Wareing, M. Elhay, Z. Hashmi)
- National Institute of Animal Health, Ibaraki, Japan (M. Yamada)
- Duke–NUS (Duke and the National University of Singapore) Graduate Medical School, Singapore (L.-F. Wang)
| |
Collapse
|
56
|
A recombinant Hendra virus G glycoprotein subunit vaccine protects nonhuman primates against Hendra virus challenge. J Virol 2014; 88:4624-31. [PMID: 24522928 DOI: 10.1128/jvi.00005-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Hendra virus (HeV) is a zoonotic emerging virus belonging to the family Paramyxoviridae. HeV causes severe and often fatal respiratory and/or neurologic disease in both animals and humans. Currently, there are no licensed vaccines or antiviral drugs approved for human use. A number of animal models have been developed for studying HeV infection, with the African green monkey (AGM) appearing to most faithfully reproduce the human disease. Here, we assessed the utility of a newly developed recombinant subunit vaccine based on the HeV attachment (G) glycoprotein in the AGM model. Four AGMs were vaccinated with two doses of the HeV vaccine (sGHeV) containing Alhydrogel, four AGMs received the sGHeV with Alhydrogel and CpG, and four control animals did not receive the sGHeV vaccine. Animals were challenged with a high dose of infectious HeV 21 days after the boost vaccination. None of the eight specifically vaccinated animals showed any evidence of clinical illness and survived the challenge. All four controls became severely ill with symptoms consistent with HeV infection, and three of the four animals succumbed 8 days after exposure. Success of the recombinant subunit vaccine in AGMs provides pivotal data in supporting its further preclinical development for potential human use. IMPORTANCE A Hendra virus attachment (G) glycoprotein subunit vaccine was tested in nonhuman primates to assess its ability to protect them from a lethal infection with Hendra virus. It was found that all vaccinated African green monkeys were completely protected against subsequent Hendra virus infection and disease. The success of this new subunit vaccine in nonhuman primates provides critical data in support of its further development for future human use.
Collapse
|
57
|
Nieto K, Salvetti A. AAV Vectors Vaccines Against Infectious Diseases. Front Immunol 2014; 5:5. [PMID: 24478774 PMCID: PMC3896988 DOI: 10.3389/fimmu.2014.00005] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 01/07/2014] [Indexed: 12/12/2022] Open
Abstract
Since their discovery as a tool for gene transfer, vectors derived from the adeno-associated virus (AAV) have been used for gene therapy applications and attracted scientist to this field for their exceptional properties of efficiency of in vivo gene transfer and the level and duration of transgene expression. For many years, AAVs have been considered as low immunogenic vectors due to their ability to induce long-term expression of non-self-proteins in contrast to what has been observed with other viral vectors, such as adenovirus, for which strong immune responses against the same transgene products were documented. The perceived low immunogenicity likely explains why the use of AAV vectors for vaccination was not seriously considered before the early 2000s. Indeed, while analyses conducted using a variety of transgenes and animal species slowly changed the vision of immunological properties of AAVs, an increasing number of studies were also performed in the field of vaccination. Even if the comparison with other modes of vaccination was not systemically performed, the analyses conducted so far in the field of active immunotherapy strongly suggest that AAVs possess some interesting features to be used as tools to produce an efficient and sustained antibody response. In addition, recent studies also highlighted the potential of AAVs for passive immunotherapy. This review summarizes the main studies conducted to evaluate the potential of AAV vectors for vaccination against infectious agents and discusses their advantages and drawbacks. Altogether, the variety of studies conducted in this field contributes to the understanding of the immunological properties of this versatile virus and to the definition of its possible future applications.
Collapse
Affiliation(s)
- Karen Nieto
- Tumor Immunology Program (D030), German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Anna Salvetti
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon , Lyon , France ; LabEx Ecofect, Université de Lyon , Lyon , France
| |
Collapse
|
58
|
Raksha S, Tan WS, Hamid M, Ramanan RN, Tey BT. A Single-Step Purification of the Glycoprotein of Nipah Virus Produced in Insect Cells using an Anion Exchange Chromatography Method. SEP SCI TECHNOL 2014. [DOI: 10.1080/01496395.2013.838265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
59
|
Mire CE, Versteeg KM, Cross RW, Agans KN, Fenton KA, Whitt MA, Geisbert TW. Single injection recombinant vesicular stomatitis virus vaccines protect ferrets against lethal Nipah virus disease. Virol J 2013; 10:353. [PMID: 24330654 PMCID: PMC3878732 DOI: 10.1186/1743-422x-10-353] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/03/2013] [Indexed: 11/10/2022] Open
Abstract
Background Nipah virus (NiV) is a highly pathogenic zoonotic agent in the family Paramyxoviridae that is maintained in nature by bats. Outbreaks have occurred in Malaysia, Singapore, India, and Bangladesh and have been associated with 40 to 75% case fatality rates. There are currently no vaccines or postexposure treatments licensed for combating human NiV infection. Methods and results Four groups of ferrets received a single vaccination with different recombinant vesicular stomatitis virus vectors expressing: Group 1, control with no glycoprotein; Group 2, the NiV fusion protein (F); Group 3, the NiV attachment protein (G); and Group 4, a combination of the NiV F and G proteins. Animals were challenged intranasally with NiV 28 days after vaccination. Control ferrets in Group 1 showed characteristic clinical signs of NiV disease including respiratory distress, neurological disorders, viral load in blood and tissues, and gross lesions and antigen in target tissues; all animals in this group succumbed to infection by day 8. Importantly, all specifically vaccinated ferrets in Groups 2-4 showed no evidence of clinical illness and survived challenged. All animals in these groups developed anti-NiV F and/or G IgG and neutralizing antibody titers. While NiV RNA was detected in blood at day 6 post challenge in animals from Groups 2-4, the levels were orders of magnitude lower than animals from control Group 1. Conclusions These data show protective efficacy against NiV in a relevant model of human infection. Further development of this technology has the potential to yield effective single injection vaccines for NiV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd,, Galveston, TX, USA.
| |
Collapse
|
60
|
Lo MK, Bird BH, Chattopadhyay A, Drew CP, Martin BE, Coleman JD, Rose JK, Nichol ST, Spiropoulou CF. Single-dose replication-defective VSV-based Nipah virus vaccines provide protection from lethal challenge in Syrian hamsters. Antiviral Res 2013; 101:26-9. [PMID: 24184127 DOI: 10.1016/j.antiviral.2013.10.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/18/2013] [Accepted: 10/22/2013] [Indexed: 01/02/2023]
Abstract
Nipah virus (NiV) continues to cause outbreaks of fatal human encephalitis due to spillover from its bat reservoir. We determined that a single dose of replication-defective vesicular stomatitis virus (VSV)-based vaccine vectors expressing either the NiV fusion (F) or attachment (G) glycoproteins protected hamsters from over 1000 times LD50 NiV challenge. This highly effective single-dose protection coupled with an enhanced safety profile makes these candidates ideal for potential use in livestock and humans.
Collapse
Affiliation(s)
- Michael K Lo
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States.
| | - Brian H Bird
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Anasuya Chattopadhyay
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Clifton P Drew
- Infectious Disease Pathology Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Brock E Martin
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joann D Coleman
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - John K Rose
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States.
| |
Collapse
|
61
|
Hazelton B, Ba Alawi F, Kok J, Dwyer DE. Hendra virus: a one health tale of flying foxes, horses and humans. Future Microbiol 2013; 8:461-74. [PMID: 23534359 DOI: 10.2217/fmb.13.19] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hendra virus, a member of the family Paramyxoviridae, was first recognized following a devastating outbreak in Queensland, Australia, in 1994. The naturally acquired symptomatic infection, characterized by a rapidly progressive illness involving the respiratory system and/or CNS, has so far only been recognized in horses and humans. However, there is potential for other species to be infected, with significant consequences for animal and human health. Prevention of infection involves efforts to interrupt the bat-to-horse and horse-to-human transmission interfaces. Education and infection-control efforts remain the key to reducing risk of transmission, particularly as no effective antiviral treatment is currently available. The recent release of an equine Hendra G glycoprotein subunit vaccine is an exciting advance that offers the opportunity to curb the recent increase in equine transmission events occurring in endemic coastal regions of Australia and thereby reduce the risk of infection in humans.
Collapse
Affiliation(s)
- Briony Hazelton
- Centre for Infectious Diseases & Microbiology Laboratory Services, Institute of Clinical Pathology & Medical Research, Westmead Hospital, Westmead, New South Wales 2145, Australia.
| | | | | | | |
Collapse
|
62
|
Pallister JA, Klein R, Arkinstall R, Haining J, Long F, White JR, Payne J, Feng YR, Wang LF, Broder CC, Middleton D. Vaccination of ferrets with a recombinant G glycoprotein subunit vaccine provides protection against Nipah virus disease for over 12 months. Virol J 2013; 10:237. [PMID: 23867060 PMCID: PMC3718761 DOI: 10.1186/1743-422x-10-237] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/11/2013] [Indexed: 11/17/2022] Open
Abstract
Background Nipah virus (NiV) is a zoonotic virus belonging to the henipavirus genus in the family Paramyxoviridae. Since NiV was first identified in 1999, outbreaks have continued to occur in humans in Bangladesh and India on an almost annual basis with case fatality rates reported between 40% and 100%. Methods Ferrets were vaccinated with 4, 20 or 100 μg HeVsG formulated with the human use approved adjuvant, CpG, in a prime-boost regime. One half of the ferrets were exposed to NiV at 20 days post boost vaccination and the other at 434 days post vaccination. The presence of virus or viral genome was assessed in ferret fluids and tissues using real-time PCR, virus isolation, histopathology, and immunohistochemistry; serology was also carried out. Non-immunised ferrets were also exposed to virus to confirm the pathogenicity of the inoculum. Results Ferrets exposed to Nipah virus 20 days post vaccination remained clinically healthy. Virus or viral genome was not detected in any tissues or fluids of the vaccinated ferrets; lesions and antigen were not identified on immunohistological examination of tissues; and there was no increase in antibody titre during the observation period, consistent with failure of virus replication. Of the ferrets challenged 434 days post vaccination, all five remained well throughout the study period; viral genome – but not virus - was recovered from nasal secretions of one ferret given 20 μg HeVsG and bronchial lymph nodes of the other. There was no increase in antibody titre during the observation period, consistent with lack of stimulation of a humoral memory response. Conclusions We have previously shown that ferrets vaccinated with 4, 20 or 100 μg HeVsG formulated with CpG adjuvant, which is currently in several human clinical trials, were protected from HeV disease. Here we show, under similar conditions of use, that the vaccine also provides protection against NiV-induced disease. Such protection persists for at least 12 months post-vaccination, with data supporting only localised and self-limiting virus replication in 2 of 5 animals. These results augur well for acceptability of the vaccine to industry.
Collapse
Affiliation(s)
- Jackie A Pallister
- CSIRO Livestock Industries, Australian Animal Health Laboratory, 5 Portarlington Road, Geelong, VIC 3220, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Broder CC, Xu K, Nikolov DB, Zhu Z, Dimitrov DS, Middleton D, Pallister J, Geisbert TW, Bossart KN, Wang LF. A treatment for and vaccine against the deadly Hendra and Nipah viruses. Antiviral Res 2013; 100:8-13. [PMID: 23838047 DOI: 10.1016/j.antiviral.2013.06.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 12/29/2022]
Abstract
Hendra virus and Nipah virus are bat-borne paramyxoviruses that are the prototypic members of the genus Henipavirus. The henipaviruses emerged in the 1990s, spilling over from their natural bat hosts and causing serious disease outbreaks in humans and livestock. Hendra virus emerged in Australia and since 1994 there have been 7 human infections with 4 case fatalities. Nipah virus first appeared in Malaysia and subsequent outbreaks have occurred in Bangladesh and India. In total, there have been an estimated 582 human cases of Nipah virus and of these, 54% were fatal. Their broad species tropism and ability to cause fatal respiratory and/or neurologic disease in humans and animals make them important transboundary biological threats. Recent experimental findings in animals have demonstrated that a human monoclonal antibody targeting the viral G glycoprotein is an effective post-exposure treatment against Hendra and Nipah virus infection. In addition, a subunit vaccine based on the G glycoprotein of Hendra virus affords protection against Hendra and Nipah virus challenge. The vaccine has been developed for use in horses in Australia and is the first vaccine against a Biosafety Level-4 (BSL-4) agent to be licensed and commercially deployed. Together, these advances offer viable approaches to address Hendra and Nipah virus infection of livestock and people.
Collapse
Affiliation(s)
- Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Dhondt KP, Horvat B. Henipavirus infections: lessons from animal models. Pathogens 2013; 2:264-87. [PMID: 25437037 PMCID: PMC4235719 DOI: 10.3390/pathogens2020264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 04/02/2013] [Accepted: 04/04/2013] [Indexed: 11/16/2022] Open
Abstract
The Henipavirus genus contains two highly lethal viruses, the Hendra and Nipah viruses and one, recently discovered, apparently nonpathogenic member; Cedar virus. These three, negative-sense single-stranded RNA viruses, are hosted by fruit bats and use EphrinB2 receptors for entry into cells. The Hendra and Nipah viruses are zoonotic pathogens that emerged in the middle of 90s and have caused severe, and often fatal, neurologic and/or respiratory diseases in both humans and different animals; including spillover into equine and porcine species. Development of relevant models is critical for a better understanding of viral pathogenesis, generating new diagnostic tools, and assessing anti-viral therapeutics and vaccines. This review summarizes available data on several animal models where natural and/or experimental infection has been demonstrated; including pteroid bats, horses, pigs, cats, hamsters, guinea pigs, ferrets, and nonhuman primates. It recapitulates the principal features of viral pathogenesis in these animals and current knowledge on anti-viral immune responses. Lastly it describes the recently characterized murine animal model, which provides the possibility to use numerous and powerful tools available for mice to further decipher henipaviruses immunopathogenesis, prophylaxis, and treatment. The utility of different models to analyze important aspects of henipaviruses-induced disease in humans, potential routes of transmission, and therapeutic approaches are equally discussed.
Collapse
Affiliation(s)
- Kévin P Dhondt
- International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon 1, 21 Avenue T. Garnier, Lyon 69007, France.
| | - Branka Horvat
- International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon 1, 21 Avenue T. Garnier, Lyon 69007, France.
| |
Collapse
|
65
|
Yoneda M, Georges-Courbot MC, Ikeda F, Ishii M, Nagata N, Jacquot F, Raoul H, Sato H, Kai C. Recombinant measles virus vaccine expressing the Nipah virus glycoprotein protects against lethal Nipah virus challenge. PLoS One 2013; 8:e58414. [PMID: 23516477 PMCID: PMC3597623 DOI: 10.1371/journal.pone.0058414] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 02/04/2013] [Indexed: 11/21/2022] Open
Abstract
Nipah virus (NiV) is a member of the genus Henipavirus, which emerged in Malaysia in 1998. In pigs, infection resulted in a predominantly non-lethal respiratory disease; however, infection in humans resulted in over 100 deaths. Nipah virus has continued to re-emerge in Bangladesh and India, and person-to-person transmission appeared in the outbreak. Although a number of NiV vaccine studies have been reported, there are currently no vaccines or treatments licensed for human use. In this study, we have developed a recombinant measles virus (rMV) vaccine expressing NiV envelope glycoproteins (rMV-HL-G and rMV-Ed-G). Vaccinated hamsters were completely protected against NiV challenge, while the mortality of unvaccinated control hamsters was 90%. We trialed our vaccine in a non-human primate model, African green monkeys. Upon intraperitoneal infection with NiV, monkeys showed several clinical signs of disease including severe depression, reduced ability to move and decreased food ingestion and died at 7 days post infection (dpi). Intranasal and oral inoculation induced similar clinical illness in monkeys, evident around 9 dpi, and resulted in a moribund stage around 14 dpi. Two monkeys immunized subcutaneously with rMV-Ed-G showed no clinical illness prior to euthanasia after challenge with NiV. Viral RNA was not detected in any organ samples collected from vaccinated monkeys, and no pathological changes were found upon histopathological examination. From our findings, we propose that rMV-NiV-G is an appropriate NiV vaccine candidate for use in humans.
Collapse
Affiliation(s)
- Misako Yoneda
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- * E-mail: (MY); (CK)
| | | | - Fusako Ikeda
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Miho Ishii
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Noriyo Nagata
- National Institute of Infectious Diseases, Department of Pathology Tokyo, Japan
| | - Frederic Jacquot
- Institut National de la Sante et de la Recherche Médicale, Laboratory P4 INSERM Jean Mérieux, Lyon, France
| | - Hervé Raoul
- Institut National de la Sante et de la Recherche Médicale, Laboratory P4 INSERM Jean Mérieux, Lyon, France
| | - Hiroki Sato
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Chieko Kai
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- * E-mail: (MY); (CK)
| |
Collapse
|
66
|
Abstract
Nipah (NiV) and Hendra (HeV) viruses are the deadliest human pathogens within the Paramyxoviridae family, which include human and animal pathogens of global biomedical importance. NiV and HeV infections cause respiratory and encephalitic illness with high mortality rates in humans. Henipaviruses (HNV) are the only Paramyxoviruses classified as biosafety level 4 (BSL4) pathogens due to their extreme pathogenicity, potential for bioterrorism, and lack of licensed vaccines and therapeutics. HNV use ephrin-B2 and ephrin-B3, highly conserved proteins, as viral entry receptors. This likely accounts for their unusually broad species tropism, and also provides opportunities to study how receptor usage, cellular tropism, and end-organ pathology relates to the pathobiology of HNV infections. The clinical and pathologic manifestations of NiV and HeV virus infections are reviewed in the chapters by Wong et al. and Geisbert et al. in this issue. Here, we will review the biology of the HNV receptors, and how receptor usage relates to HNV cell tropism in vitro and in vivo.
Collapse
Affiliation(s)
- Olivier Pernet
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1489, USA
| | | | | |
Collapse
|
67
|
Abstract
Highly pathogenic Nipah virus (NiV) infections are transmitted via airway secretions and urine, commonly via the respiratory route. Epithelial surfaces represent important replication sites in both primary and systemic infection phases. NiV entry and spread from polarized epithelial cells therefore determine virus entry and dissemination within a new host and influence virus shedding via mucosal surfaces in the respiratory and urinary tract. To date, there is no knowledge regarding the entry and exit sites of NiV in polarized epithelial cells. In this report, we show for the first time that NiV can infect polarized kidney epithelial cells (MDCK) from both cell surfaces, while virus release is primarily restricted to the apical plasma membrane. Substantial amounts of basolateral infectivity were detected only after infection with high virus doses, at time points when the integrity of the cell monolayer was largely disrupted as a result of cell-to-cell fusion. Confocal immunofluorescence analyses of envelope protein distribution at early and late infection stages suggested that apical virus budding is determined by the polarized sorting of the NiV matrix protein, M. Studies with stably M-expressing and with monensin-treated cells furthermore demonstrated that M protein transport is independent from the glycoproteins, implying that the M protein possesses an intrinsic apical targeting signal.
Collapse
|
68
|
Abstract
As the threat of exposure to emerging and reemerging viruses within a naive population increases, it is vital that the basic mechanisms of pathogenesis and immune response be thoroughly investigated. By using animal models in this endeavor, the response to viruses can be studied in a more natural context to identify novel drug targets, and assess the efficacy and safety of new products. This is especially true in the advent of the Food and Drug Administration's animal rule. Although no one animal model is able to recapitulate all the aspects of human disease, understanding the current limitations allows for a more targeted experimental design. Important facets to be considered before an animal study are the route of challenge, species of animals, biomarkers of disease, and a humane endpoint. This chapter covers the current animal models for medically important human viruses, and demonstrates where the gaps in knowledge exist.
Collapse
|
69
|
Clayton BA, Middleton D, Bergfeld J, Haining J, Arkinstall R, Wang L, Marsh GA. Transmission routes for nipah virus from Malaysia and Bangladesh. Emerg Infect Dis 2012; 18:1983-93. [PMID: 23171621 PMCID: PMC3557903 DOI: 10.3201/eid1812.120875] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Human infections with Nipah virus in Malaysia and Bangladesh are associated with markedly different patterns of transmission and pathogenicity. To compare the 2 strains, we conducted an in vivo study in which 2 groups of ferrets were oronasally exposed to either the Malaysia or Bangladesh strain of Nipah virus. Viral shedding and tissue tropism were compared between the 2 groups. Over the course of infection, significantly higher levels of viral RNA were recovered from oral secretions of ferrets infected with the Bangladesh strain. Higher levels of oral shedding of the Bangladesh strain of Nipah virus might be a key factor in onward transmission in outbreaks among humans.
Collapse
Affiliation(s)
- Bronwyn A Clayton
- Commonwealth Scientifi c and Industrial Research Organisation Livestock Industries, Geelong, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
70
|
Ploquin A, Szécsi J, Mathieu C, Guillaume V, Barateau V, Ong KC, Wong KT, Cosset FL, Horvat B, Salvetti A. Protection against henipavirus infection by use of recombinant adeno-associated virus-vector vaccines. J Infect Dis 2012; 207:469-78. [PMID: 23175762 PMCID: PMC7107322 DOI: 10.1093/infdis/jis699] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Nipah virus (NiV) and Hendra virus (HeV) are closely related, recently emerged paramyxoviruses that are capable of causing considerable morbidity and mortality in several mammalian species, including humans. Henipavirus-specific vaccines are still commercially unavailable, and development of novel antiviral strategies to prevent lethal infections due to henipaviruses is highly desirable. Here we describe the development of adeno-associated virus (AAV) vaccines expressing the NiV G protein. Characterization of these vaccines in mice demonstrated that a single intramuscular AAV injection was sufficient to induce a potent and long-lasting antibody response. Translational studies in hamsters further demonstrated that all vaccinated animals were protected against lethal challenge with NiV. In addition, this vaccine induced a cross-protective immune response that was able to protect 50% of the animals against a challenge by HeV. This study presents a new efficient vaccination strategy against henipaviruses and opens novel perspectives on the use of AAV vectors as vaccines against emergent diseases.
Collapse
Affiliation(s)
- Aurélie Ploquin
- INSERM U758, 2Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Lo MK, Peeples ME, Bellini WJ, Nichol ST, Rota PA, Spiropoulou CF. Distinct and overlapping roles of Nipah virus P gene products in modulating the human endothelial cell antiviral response. PLoS One 2012; 7:e47790. [PMID: 23094089 PMCID: PMC3477106 DOI: 10.1371/journal.pone.0047790] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 09/17/2012] [Indexed: 12/15/2022] Open
Abstract
Nipah virus (NiV) is a highly pathogenic zoonotic paramyxovirus that causes fatal encephalitis in up to 75% of infected humans. Like other paramyxoviruses, NiV employs co-transcriptional mRNA editing during transcription of the phosphoprotein (P) gene to generate additional mRNAs encoding the V and W proteins. The C protein is translated from the P mRNA, but in an alternative reading frame. There is evidence from both in vitro and in vivo studies to show that the P gene products play a role in NiV pathogenesis. We have developed a reverse genetic system to dissect the individual roles of the NiV P gene products in limiting the antiviral response in primary human microvascular lung endothelial cells, which represent important targets in human NiV infection. By characterizing growth curves and early antiviral responses against a number of recombinant NiVs with genetic modifications altering expression of the proteins encoded by the P gene, we observed that multiple elements encoded by the P gene have both distinct and overlapping roles in modulating virus replication as well as in limiting expression of antiviral mediators such as IFN-β, CXCL10, and CCL5. Our findings corroborate observations from in vivo hamster infection studies, and provide molecular insights into the attenuation and the histopathology observed in hamsters infected with C, V, and W-deficient NiVs. The results of this study also provide an opportunity to verify the results of earlier artificial plasmid expression studies in the context of authentic viral infection.
Collapse
Affiliation(s)
- Michael K Lo
- Centers for Disease Control & Prevention, Viral Special Pathogens Branch, Atlanta, Georgia, United States of America.
| | | | | | | | | | | |
Collapse
|
72
|
Prescott J, de Wit E, Feldmann H, Munster VJ. The immune response to Nipah virus infection. Arch Virol 2012; 157:1635-41. [PMID: 22669317 PMCID: PMC3432143 DOI: 10.1007/s00705-012-1352-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 04/20/2012] [Indexed: 02/07/2023]
Abstract
Nipah virus has recently emerged as a zoonotic agent that is highly pathogenic in humans. Outbreaks have occurred regularly over the last two decades in South and Southeast Asia, where mortality rates reach as high as 100 %. The natural reservoir of Nipah virus has been identified as bats from the Pteropus family, where infection is largely asymptomatic. Human disease is characterized by both respiratory and encephalitic components, and thus far, no effective vaccine or intervention strategies are available. Little is know about how the immune response of either the reservoir host or incidental hosts responds to infection, and how this immune response is either inadequate or might contribute to disease in the dead-end host. Experimental vaccines strategies have given us some insight into the immunological requirements for protection. This review summarizes our current understanding of the immune response to Nipah virus infection and emphasizes the need for further research.
Collapse
Affiliation(s)
- Joseph Prescott
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | | | | | | |
Collapse
|
73
|
Jiang S, Lu L, Liu Q, Xu W, Du L. Receptor-binding domains of spike proteins of emerging or re-emerging viruses as targets for development of antiviral vaccines. Emerg Microbes Infect 2012; 1:e13. [PMID: 26038424 PMCID: PMC3630917 DOI: 10.1038/emi.2012.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 03/10/2012] [Accepted: 03/12/2012] [Indexed: 12/13/2022]
Abstract
A number of emerging and re-emerging viruses have caused epidemics or pandemics of infectious diseases leading to major devastations throughout human history. Therefore, developing effective and safe vaccines against these viruses is clearly important for the protection of at-risk populations. Our previous studies have shown that the receptor-binding domain (RBD) in the spike protein of severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV) is a key target for the development of SARS vaccines. In this review, we highlight some key advances in the development of antiviral vaccines targeting the RBDs of spike proteins of emerging and re-emerging viruses, using SARS-CoV, influenza virus, Hendra virus (HeV) and Nipah virus (NiV) as examples.
Collapse
Affiliation(s)
- Shibo Jiang
- MOE/MOH Key Laboratory of Medical Molecular Virology, Shanghai Medical College and Institute of Medical Microbiology, Fudan University , Shanghai 200032, China ; Lindsley F. Kimball Research Institute, New York Blood Center , New York, NY 10065, USA
| | - Lu Lu
- MOE/MOH Key Laboratory of Medical Molecular Virology, Shanghai Medical College and Institute of Medical Microbiology, Fudan University , Shanghai 200032, China
| | - Qi Liu
- MOE/MOH Key Laboratory of Medical Molecular Virology, Shanghai Medical College and Institute of Medical Microbiology, Fudan University , Shanghai 200032, China ; Department of Medical Microbiology and Immunology, School of Basic Medicine, Dali University , Dali 671000, China
| | - Wei Xu
- MOE/MOH Key Laboratory of Medical Molecular Virology, Shanghai Medical College and Institute of Medical Microbiology, Fudan University , Shanghai 200032, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center , New York, NY 10065, USA
| |
Collapse
|
74
|
Marsh GA, de Jong C, Barr JA, Tachedjian M, Smith C, Middleton D, Yu M, Todd S, Foord AJ, Haring V, Payne J, Robinson R, Broz I, Crameri G, Field HE, Wang LF. Cedar virus: a novel Henipavirus isolated from Australian bats. PLoS Pathog 2012; 8:e1002836. [PMID: 22879820 PMCID: PMC3410871 DOI: 10.1371/journal.ppat.1002836] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 06/19/2012] [Indexed: 12/13/2022] Open
Abstract
The genus Henipavirus in the family Paramyxoviridae contains two viruses, Hendra virus (HeV) and Nipah virus (NiV) for which pteropid bats act as the main natural reservoir. Each virus also causes serious and commonly lethal infection of people as well as various species of domestic animals, however little is known about the associated mechanisms of pathogenesis. Here, we report the isolation and characterization of a new paramyxovirus from pteropid bats, Cedar virus (CedPV), which shares significant features with the known henipaviruses. The genome size (18,162 nt) and organization of CedPV is very similar to that of HeV and NiV; its nucleocapsid protein displays antigenic cross-reactivity with henipaviruses; and it uses the same receptor molecule (ephrin- B2) for entry during infection. Preliminary challenge studies with CedPV in ferrets and guinea pigs, both susceptible to infection and disease with known henipaviruses, confirmed virus replication and production of neutralizing antibodies although clinical disease was not observed. In this context, it is interesting to note that the major genetic difference between CedPV and HeV or NiV lies within the coding strategy of the P gene, which is known to play an important role in evading the host innate immune system. Unlike HeV, NiV, and almost all known paramyxoviruses, the CedPV P gene lacks both RNA editing and also the coding capacity for the highly conserved V protein. Preliminary study indicated that CedPV infection of human cells induces a more robust IFN-β response than HeV. Hendra and Nipah viruses are 2 highly pathogenic paramyxoviruses that have emerged from bats within the last two decades. Both are capable of causing fatal disease in both humans and many mammal species. Serological and molecular evidence for henipa-like viruses have been reported from numerous locations including Asia and Africa, however, until now no successful isolation of these viruses have been reported. This paper reports the isolation of a novel paramyxovirus, named Cedar virus, from fruit bats in Australia. Full genome sequencing of this virus suggests a close relationship with the henipaviruses. Antibodies to Cedar virus were shown to cross react with, but not cross neutralize Hendra or Nipah virus. Despite this close relationship, when Cedar virus was tested in experimental challenge models in ferrets and guinea pigs, we identified virus replication and generation of neutralizing antibodies, but no clinical disease was observed. As such, this virus provides a useful reference for future reverse genetics experiments to determine the molecular basis of the pathogenicity of the henipaviruses.
Collapse
Affiliation(s)
- Glenn A. Marsh
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Carol de Jong
- Queensland Centre for Emerging Infectious Diseases, Biosecurity Queensland, Coopers Plain, Australia
| | - Jennifer A. Barr
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Mary Tachedjian
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Craig Smith
- Queensland Centre for Emerging Infectious Diseases, Biosecurity Queensland, Coopers Plain, Australia
| | - Deborah Middleton
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Meng Yu
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Shawn Todd
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Adam J. Foord
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Volker Haring
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Jean Payne
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Rachel Robinson
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Ivano Broz
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Gary Crameri
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Hume E. Field
- Queensland Centre for Emerging Infectious Diseases, Biosecurity Queensland, Coopers Plain, Australia
- * E-mail: (HEF); (LFW)
| | - Lin-Fa Wang
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- * E-mail: (HEF); (LFW)
| |
Collapse
|
75
|
Gutiérrez AH, Spero DM, Gay C, Zimic M, De Groot AS. New vaccines needed for pathogens infecting animals and humans: One Health. Hum Vaccin Immunother 2012; 8:971-8. [PMID: 22485046 DOI: 10.4161/hv.20202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The field of "One Health" encourages researchers to collaborate across a wide range of disciplines to improve health at the animal-human-ecosystems interface. One Health recognizes the potential of emerging infectious diseases to impact public health and global food security, and the need for a multidisciplinary approach to counteract the effect of these diseases. Vaccinologists are also beginning to engage in research related to One Health, recognizing that preventing transmission of emerging infectious diseases at the animal-human interface is critically important for protecting the world population from epizootics and pandemics. In this synopsis of recent work in the One Health field, we describe some emerging One Health pathogens, discuss the importance of One Health to food safety and biodefense, propose strategies for improving One Health including the development of new vaccines and new vaccine design approaches, and close with a brief discussion of the opportunities and risks related to One Health vaccine research.
Collapse
Affiliation(s)
- Andres H Gutiérrez
- Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | | | | | | | | |
Collapse
|
76
|
Kong D, Wen Z, Su H, Ge J, Chen W, Wang X, Wu C, Yang C, Chen H, Bu Z. Newcastle disease virus-vectored Nipah encephalitis vaccines induce B and T cell responses in mice and long-lasting neutralizing antibodies in pigs. Virology 2012; 432:327-35. [PMID: 22726244 DOI: 10.1016/j.virol.2012.06.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 04/13/2012] [Accepted: 06/01/2012] [Indexed: 11/29/2022]
Abstract
Nipah virus (NiV), a member of the Paramyxoviridae family, causes deadly encephalitis in humans and huge economic losses to the pig industry. Here, we generated recombinant avirulent Newcastle disease virus (NDV) LaSota strains expressing the NiV G and F proteins respectively (designated as rLa-NiVG and rLa-NiVF), and evaluated their immunogenicity in mice and pigs. Both rLa-NiVG and rLa-NiVF displayed growth properties similar to those of LaSota virus in chicken eggs. Co-infection of rLa-NiVG and rLa-NiVF caused marked syncytia formation, while intracerebral co-inoculation of these viruses in mice showed they were safe in at least one mammalian species. Animal immunization studies showed rLa-NiVG and rLa-NiVF induced NiV neutralizing antibody responses in mice and pigs, and F protein-specific CD8+ T cell responses in mice. Most importantly, rLa-NiVG and rLa-NiVF administered alone or together, induced a long-lasting neutralizing antibody response in pigs. Recombinant rLa-NiVG/F thus appear to be promising NiV vaccine candidates for pigs and potentially humans.
Collapse
Affiliation(s)
- Dongni Kong
- State Key Laboratory of Veterinary Biotechnology and Animal Influenza Laboratory of the Ministry of Agriculture, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Rockx B, Winegar R, Freiberg AN. Recent progress in henipavirus research: molecular biology, genetic diversity, animal models. Antiviral Res 2012; 95:135-49. [PMID: 22643730 DOI: 10.1016/j.antiviral.2012.05.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 05/08/2012] [Accepted: 05/19/2012] [Indexed: 12/17/2022]
Abstract
Nipah and Hendra virus are members of a newly identified genus of emerging paramyxoviruses, the henipaviruses. Both viruses have the ability to cause severe pulmonary infection and severe acute encephalitis. Following their discovery in the 1990s, outbreaks caused by these zoonotic paramyxoviruses have been associated with high public health and especially economic threat potential. Currently, only geographic groupings in Asia and Australia have been described for the henipaviruses. However, while few viral isolates are available and more detailed characterization is necessary, there has been recent evidence that divergent henipaviruses might be present on the African continent. This review endeavours to capture recent advances in the field of henipavirus research, with a focus on genome structure and replication mechanisms, reservoir hosts, genetic diversity, pathogenesis and animal models.
Collapse
Affiliation(s)
- Barry Rockx
- Departments of Microbiology and Immunology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, United States.
| | | | | |
Collapse
|
78
|
Pessi A, Langella A, Capitò E, Ghezzi S, Vicenzi E, Poli G, Ketas T, Mathieu C, Cortese R, Horvat B, Moscona A, Porotto M. A general strategy to endow natural fusion-protein-derived peptides with potent antiviral activity. PLoS One 2012; 7:e36833. [PMID: 22666328 PMCID: PMC3353973 DOI: 10.1371/journal.pone.0036833] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 04/07/2012] [Indexed: 01/26/2023] Open
Abstract
Fusion between the viral and target cell membranes is an obligatory step for the infectivity of all enveloped virus, and blocking this process is a clinically validated therapeutic strategy. Viral fusion is driven by specialized proteins which, although specific to each virus, act through a common mechanism, the formation of a complex between two heptad repeat (HR) regions. The HR regions are initially separated in an intermediate termed “prehairpin”, which bridges the viral and cell membranes, and then fold onto each other to form a 6-helical bundle (6HB), driving the two membranes to fuse. HR-derived peptides can inhibit viral infectivity by binding to the prehairpin intermediate and preventing its transition to the 6HB. The antiviral activity of HR-derived peptides differs considerably among enveloped viruses. For weak inhibitors, potency can be increased by peptide engineering strategies, but sequence-specific optimization is time-consuming. In seeking ways to increase potency without changing the native sequence, we previously reported that attachment to the HR peptide of a cholesterol group (”cholesterol-tagging”) dramatically increases its antiviral potency, and simultaneously increases its half-life in vivo. We show here that antiviral potency may be increased by combining cholesterol-tagging with dimerization of the HR-derived sequence, using as examples human parainfluenza virus, Nipah virus, and HIV-1. Together, cholesterol-tagging and dimerization may represent strategies to boost HR peptide potency to levels that in some cases may be compatible with in vivo use, possibly contributing to emergency responses to outbreaks of existing or novel viruses.
Collapse
Affiliation(s)
| | | | | | - Silvia Ghezzi
- Viral Pathogens and Biosafety, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogens and Biosafety, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Guido Poli
- AIDS Immunopathogenesis Units, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Thomas Ketas
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Cyrille Mathieu
- INSERM, Ecole Normale Supérieure de Lyon, Lyon, France
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
| | | | - Branka Horvat
- INSERM, Ecole Normale Supérieure de Lyon, Lyon, France
- IFR128 BioSciences Lyon-Gerland Lyon-Sud, University of Lyon, Lyon, France
| | - Anne Moscona
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Matteo Porotto
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail: (AP); (MP)
| |
Collapse
|
79
|
Marsh GA, Haining J, Hancock TJ, Robinson R, Foord AJ, Barr JA, Riddell S, Heine HG, White JR, Crameri G, Field HE, Wang LF, Middleton D. Experimental infection of horses with Hendra virus/Australia/horse/2008/Redlands. Emerg Infect Dis 2012; 17:2232-8. [PMID: 22172152 PMCID: PMC3311212 DOI: 10.3201/eid1712.111162] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Early consideration of HeV and institution of infection control are critical for reducing human risk. Hendra virus (HeV) is a highly pathogenic zoonotic paramyxovirus harbored by Australian flying foxes with sporadic spillovers directly to horses. Although the mode and critical control points of HeV spillover to horses from flying foxes, and the risk for transmission from infected horses to other horses and humans, are poorly understood, we successfully established systemic HeV disease in 3 horses exposed to Hendra virus/Australia/Horse/2008/Redlands by the oronasal route, a plausible route for natural infection. In 2 of the 3 animals, HeV RNA was detected continually in nasal swabs from as early as 2 days postexposure, indicating that systemic spread of the virus may be preceded by local viral replication in the nasal cavity or nasopharynx. Our data suggest that a critical factor for reducing HeV exposure risk to humans includes early consideration of HeV in the differential diagnosis and institution of appropriate infection control procedures.
Collapse
Affiliation(s)
- Glenn A Marsh
- Commonwealth Scientific and Industrial Research Organization Livestock Industries, Geelong, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
The henipaviruses, Hendra virus (HeV), and Nipah virus (NiV), are enigmatic emerging pathogens that causes severe and often fatal neurologic and/or respiratory disease in both animals and humans. Amongst people, case fatality rates range between 40 and 75% and there are no vaccines or treatments approved for human use. A number of species of animals including guinea pigs, hamsters, cats, ferrets, pigs, and African green monkeys have been employed as animal models of human henipavirus infection. Here, we review the development of animal models for henipavirus infection, discuss the pathology and pathogenesis of these models, and assess the utility of each model to recapitulate important aspects of henipavirus-mediated disease seen in humans.
Collapse
|
81
|
Broder CC. Henipavirus outbreaks to antivirals: the current status of potential therapeutics. Curr Opin Virol 2012; 2:176-87. [PMID: 22482714 DOI: 10.1016/j.coviro.2012.02.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 02/22/2012] [Accepted: 02/25/2012] [Indexed: 12/29/2022]
Abstract
The henipaviruses, Hendra virus and Nipah virus, are classic examples of recently emerged viral zoonoses. In a relatively short time since their discoveries in the mid and late 1990s, respectively, a great deal of new information has been accumulated detailing their biology and certain unique characteristics. Their broad species tropism and abilities to cause severe and often fatal respiratory and/or neurologic disease in both animals and humans has sparked considerable interest in developing effective antiviral strategies to prevent or treat henipavirus infection and disease. Here, recent findings on the few most advanced henipavirus countermeasures are summarized and discussed.
Collapse
Affiliation(s)
- Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, United States.
| |
Collapse
|
82
|
Rapid screening for entry inhibitors of highly pathogenic viruses under low-level biocontainment. PLoS One 2012; 7:e30538. [PMID: 22396728 PMCID: PMC3292545 DOI: 10.1371/journal.pone.0030538] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 12/18/2011] [Indexed: 01/17/2023] Open
Abstract
Emerging viruses including Nipah, Hendra, Lujo, and Junin viruses have enormous potential to spread rapidly. Nipah virus, after emerging as a zoonosis, has also evolved the capacity for human-to-human transmission. Most of the diseases caused by these pathogens are untreatable and require high biocontainment conditions. Universal methods for rapidly identifying and screening candidate antivirals are urgently needed. We have developed a modular antiviral platform strategy that relies on simple bioinformatic and genetic information about each pathogen. Central to this platform is the use of envelope glycoprotein cDNAs to establish multi-cycle replication systems under BSL2 conditions for viral pathogens that normally require BSL3 and BSL4 facilities. We generated monoclonal antibodies against Nipah G by cDNA immunization in rats, and we showed that these antibodies neutralize both Nipah and Hendra live viruses. We then used these effective Henipavirus inhibitors to validate our screening strategy. Our proposed strategy should contribute to the response capability for emerging infectious diseases, providing a way to initiate antiviral development immediately upon identifying novel viruses.
Collapse
|
83
|
Snary EL, Ramnial V, Breed AC, Stephenson B, Field HE, Fooks AR. Qualitative release assessment to estimate the likelihood of henipavirus entering the United Kingdom. PLoS One 2012; 7:e27918. [PMID: 22328916 PMCID: PMC3273481 DOI: 10.1371/journal.pone.0027918] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 10/27/2011] [Indexed: 11/25/2022] Open
Abstract
The genus Henipavirus includes Hendra virus (HeV) and Nipah virus (NiV), for which fruit bats (particularly those of the genus Pteropus) are considered to be the wildlife reservoir. The recognition of henipaviruses occurring across a wider geographic and host range suggests the possibility of the virus entering the United Kingdom (UK). To estimate the likelihood of henipaviruses entering the UK, a qualitative release assessment was undertaken. To facilitate the release assessment, the world was divided into four zones according to location of outbreaks of henipaviruses, isolation of henipaviruses, proximity to other countries where incidents of henipaviruses have occurred and the distribution of Pteropus spp. fruit bats. From this release assessment, the key findings are that the importation of fruit from Zone 1 and 2 and bat bushmeat from Zone 1 each have a Low annual probability of release of henipaviruses into the UK. Similarly, the importation of bat meat from Zone 2, horses and companion animals from Zone 1 and people travelling from Zone 1 and entering the UK was estimated to pose a Very Low probability of release. The annual probability of release for all other release routes was assessed to be Negligible. It is recommended that the release assessment be periodically re-assessed to reflect changes in knowledge and circumstances over time.
Collapse
Affiliation(s)
- Emma L Snary
- Centre for Epidemiology and Risk Analysis, Animal Health and Veterinary Laboratories Agency, Addlestone, Surrey, United Kingdom.
| | | | | | | | | | | |
Collapse
|
84
|
Broder CC, Geisbert TW, Xu K, Nikolov DB, Wang LF, Middleton D, Pallister J, Bossart KN. Immunization strategies against henipaviruses. Curr Top Microbiol Immunol 2012; 359:197-223. [PMID: 22481140 PMCID: PMC4465348 DOI: 10.1007/82_2012_213] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hendra virus and Nipah virus are recently discovered and closely related emerging viruses that now comprise the genus henipavirus within the sub-family Paramyxoviridae and are distinguished by their broad species tropism and in addition to bats can infect and cause fatal disease in a wide variety of mammalian hosts including humans. The high mortality associated with human and animal henipavirus infections has highlighted the importance and necessity of developing effective immunization strategies. The development of suitable animal models of henipavirus infection and pathogenesis has been critical for testing the efficacy of potential therapeutic approaches. Several henipavirus challenge models have been used and recent successes in both active and passive immunization strategies against henipaviruses have been reported which have all targeted the viral envelope glycoproteins.
Collapse
Affiliation(s)
- Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - Thomas W. Geisbert
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Kai Xu
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Dimitar B. Nikolov
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Lin-Fa Wang
- CSIRO Livestock Industries, Australian Animal Health Laboratory, 5 Portarlington Road, Geelong, VIC 3220, Australia
| | - Deborah Middleton
- CSIRO Livestock Industries, Australian Animal Health Laboratory, 5 Portarlington Road, Geelong, VIC 3220, Australia
| | - Jackie Pallister
- CSIRO Livestock Industries, Australian Animal Health Laboratory, 5 Portarlington Road, Geelong, VIC 3220, Australia
| | - Katharine N. Bossart
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA. National Emerging Infectious Diseases Laboratories Institute, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
85
|
Field H, Crameri G, Kung NYH, Wang LF. Ecological aspects of hendra virus. Curr Top Microbiol Immunol 2012; 359:11-23. [PMID: 22476530 DOI: 10.1007/82_2012_214] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Hendra virus, a novel and fatally zoonotic member of the family Paramyxoviridae, was first described in Australia in 1994. Periodic spillover from its natural host (fruit bats) results in catastrophic disease in horses and occasionally the subsequent infection of humans. Prior to 2011, 14 equine incidents involving seven human cases (four fatal) were recorded. The year 2011 saw a dramatic departure from the sporadic incidents of the previous 16 years, with a cluster of 18 incidents in a single 3-month period. The fundamental difference in 2011 was the total number of incidents, the geographic clustering, and the expanded geographic range. The 2011 cluster more than doubled the total number of incidents previously reported, and poses the possibility of a new HeV infection paradigm. Epidemiologic evidence suggests that compelling additional host and/or environmental factors were at play.
Collapse
Affiliation(s)
- Hume Field
- Biosecurity Queensland, Department of Employment, Economic Development and Innovation, Brisbane, QLD, Australia.
| | | | | | | |
Collapse
|
86
|
Wang LF, Daniels P. Diagnosis of henipavirus infection: current capabilities and future directions. Curr Top Microbiol Immunol 2012; 359:179-96. [PMID: 22481141 DOI: 10.1007/82_2012_215] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the last major review on diagnosis of henipavirus infection about a decade ago, significant progress has been made in many different areas of test development, especially in the development of molecular tests using real-time PCR and many novel serological test platforms. In addition to provide an updated review of the current test capabilities, this review also identifies key future challenges in henipavirus diagnosis.
Collapse
Affiliation(s)
- Lin-Fa Wang
- CSRIO Livestock Industries, Australian Animal Health Laboratory, Geelong, VIC, Australia.
| | | |
Collapse
|
87
|
Colgrave ML, Snelling HJ, Shiell BJ, Feng YR, Chan YP, Bossart KN, Xu K, Nikolov DB, Broder CC, Michalski WP. Site occupancy and glycan compositional analysis of two soluble recombinant forms of the attachment glycoprotein of Hendra virus. Glycobiology 2011; 22:572-84. [PMID: 22171062 DOI: 10.1093/glycob/cwr180] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hendra virus (HeV) continues to cause morbidity and mortality in both humans and horses with a number of sporadic outbreaks. HeV has two structural membrane glycoproteins that mediate the infection of host cells: the attachment (G) and the fusion (F) glycoproteins that are essential for receptor binding and virion-host cell membrane fusion, respectively. N-linked glycosylation of viral envelope proteins are critical post-translation modifications that have been implicated in roles of structural integrity, virus replication and evasion of the host immune response. Deciphering the glycan composition and structure on these glycoproteins may assist in the development of glycan-targeted therapeutic intervention strategies. We examined the site occupancy and glycan composition of recombinant soluble G (sG) glycoproteins expressed in two different mammalian cell systems, transient human embryonic kidney 293 (HEK293) cells and vaccinia virus (VV)-HeLa cells, using a suite of biochemical and biophysical tools: electrophoresis, lectin binding and tandem mass spectrometry. The N-linked glycans of both VV and HEK293-derived sG glycoproteins carried predominantly mono- and disialylated complex-type N-glycans and a smaller population of high mannose-type glycans. All seven consensus sequences for N-linked glycosylation were definitively found to be occupied in the VV-derived protein, whereas only four sites were found and characterized in the HEK293-derived protein. We also report, for the first time, the existence of O-linked glycosylation sites in both proteins. The striking characteristic of both proteins was glycan heterogeneity in both N- and O-linked sites. The structural features of G protein glycosylation were also determined by X-ray crystallography and interactions with the ephrin-B2 receptor are discussed.
Collapse
Affiliation(s)
- Michelle L Colgrave
- Queensland Bioscience Precinct, CSIRO Livestock Industries, St Lucia, QLD 4067, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Halpin K, Hyatt AD, Fogarty R, Middleton D, Bingham J, Epstein JH, Rahman SA, Hughes T, Smith C, Field HE, Daszak P. Pteropid bats are confirmed as the reservoir hosts of henipaviruses: a comprehensive experimental study of virus transmission. Am J Trop Med Hyg 2011; 85:946-51. [PMID: 22049055 PMCID: PMC3205647 DOI: 10.4269/ajtmh.2011.10-0567] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 05/26/2011] [Indexed: 11/07/2022] Open
Abstract
Bats of the genus Pteropus have been identified as the reservoir hosts for the henipaviruses Hendra virus (HeV) and Nipah virus (NiV). The aim of these studies was to assess likely mechanisms for henipaviruses transmission from bats. In a series of experiments, Pteropus bats from Malaysia and Australia were inoculated with NiV and HeV, respectively, by natural routes of infection. Despite an intensive sampling strategy, no NiV was recovered from the Malaysian bats and HeV was reisolated from only one Australian bat; no disease was seen. These experiments suggest that opportunities for henipavirus transmission may be limited; therefore, the probability of a spillover event is low. For spillover to occur, a range of conditions and events must coincide. An alternate assessment framework is required if we are to fully understand how this reservoir host maintains and transmits not only these but all viruses with which it has been associated.
Collapse
Affiliation(s)
- Kim Halpin
- Commonwealth Scientific and Industrial Research Organization, Australian Animal Health Laboratory, Geelong, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Wong KT, Ong KC. Pathology of acute henipavirus infection in humans and animals. PATHOLOGY RESEARCH INTERNATIONAL 2011; 2011:567248. [PMID: 21961078 PMCID: PMC3180787 DOI: 10.4061/2011/567248] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 06/09/2011] [Indexed: 11/25/2022]
Abstract
Zoonoses as causes of human infections have been increasingly reported, and many of these are viruses that cause central nervous system infections. This paper focuses on the henipaviruses (family Paramyxoviridae, genus henipavirus) that have recently emerged to cause severe encephalitis and systemic infection in humans and animals in the Asia-Pacific region. The pathological features in the human infections comprise vasculopathy (vasculitis, endothelial multinucleated syncytia, thrombosis, etc.) and parenchymal cell infection in the central nervous system, lung, kidney, and other major organs. Most animals naturally or experimentally infected show more or less similar features confirming the dual pathogenetic mechanism of vasculopathy-associated microinfarction and direct extravascular parenchymal cell infection as causes of tissue injury. The most promising animal models include the hamster, ferret, squirrel monkey, and African green monkey. With increasing evidence of infection in the natural hosts, the pteropid bats and, hence, probable future outbreaks in many more countries, a greater awareness of henipavirus infection in both humans and animals is imperative.
Collapse
Affiliation(s)
- K. T. Wong
- Department of Pathology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - K. C. Ong
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
90
|
Ksiazek TG, Rota PA, Rollin PE. A review of Nipah and Hendra viruses with an historical aside. Virus Res 2011; 162:173-83. [PMID: 21963678 DOI: 10.1016/j.virusres.2011.09.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 09/17/2011] [Accepted: 09/18/2011] [Indexed: 11/29/2022]
Abstract
The emergence of Hendra and Nipah viruses in the 1990s has been followed by the further emergence of these viruses in the tropical Old World. The history and current knowledge of the disease, the viruses and their epidemiology is reviewed in this article. A historical aside summarizes the role that Dr. Brian W.J. Mahy played at critical junctures in the early stories of these viruses.
Collapse
Affiliation(s)
- Thomas G Ksiazek
- Galveston National Laboratory, Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0610, USA.
| | | | | |
Collapse
|
91
|
Pallister J, Middleton D, Wang LF, Klein R, Haining J, Robinson R, Yamada M, White J, Payne J, Feng YR, Chan YP, Broder CC. A recombinant Hendra virus G glycoprotein-based subunit vaccine protects ferrets from lethal Hendra virus challenge. Vaccine 2011; 29:5623-30. [PMID: 21689706 DOI: 10.1016/j.vaccine.2011.06.015] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 05/30/2011] [Accepted: 06/07/2011] [Indexed: 10/18/2022]
Abstract
The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are two deadly zoonotic viruses for which no vaccines or therapeutics have yet been approved for human or livestock use. In 14 outbreaks since 1994 HeV has been responsible for multiple fatalities in horses and humans, with all known human infections resulting from close contact with infected horses. A vaccine that prevents virus shedding in infected horses could interrupt the chain of transmission to humans and therefore prevent HeV disease in both. Here we characterise HeV infection in a ferret model and show that it closely mirrors the disease seen in humans and horses with induction of systemic vasculitis, including involvement of the pulmonary and central nervous systems. This model of HeV infection in the ferret was used to assess the immunogenicity and protective efficacy of a subunit vaccine based on a recombinant soluble version of the HeV attachment glycoprotein G (HeVsG), adjuvanted with CpG. We report that ferrets vaccinated with a 100 μg, 20 μg or 4 μg dose of HeVsG remained free of clinical signs of HeV infection following a challenge with 5000 TCID₅₀ of HeV. In addition, and of considerable importance, no evidence of virus or viral genome was detected in any tissues or body fluids in any ferret in the 100 and 20 μg groups, while genome was detected in the nasal washes only of one animal in the 4 μg group. Together, our findings indicate that 100 μg or 20 μg doses of HeVsG vaccine can completely prevent a productive HeV infection in the ferret, suggesting that vaccination to prevent the infection and shedding of HeV is possible.
Collapse
Affiliation(s)
- Jackie Pallister
- CSIRO Livestock Industries, Australian Animal Health Laboratory, 5 Portarlington Road, Geelong, VIC 3220, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Gerlier D. Emerging zoonotic viruses: new lessons on receptor and entry mechanisms. Curr Opin Virol 2011; 1:27-34. [PMID: 22440564 PMCID: PMC7102697 DOI: 10.1016/j.coviro.2011.05.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 05/20/2011] [Accepted: 05/24/2011] [Indexed: 11/27/2022]
Abstract
Viruses enter the host cell by binding cellular receptors that allow appropriate delivery of the viral genome. Although the horizontal propagation of viruses feeds the continuous emergence of novel pathogenic viruses, the genetic variation of cellular receptors can represent a challenging barrier. The SARS coronavirus, henipaviruses and filoviruses are zoonotic RNA viruses that use bats as their reservoir. Their lethality for man has fostered extensive research both on the cellular receptors they use and their entry pathways. These studies have allowed new insights into the diversity of the molecular mechanisms underlying both virus entry and pathogenesis.
Collapse
Affiliation(s)
- Denis Gerlier
- Human Virology, INSERM, U758, Ecole Normale Supérieure de Lyon, Lyon, F-69007, France.
| |
Collapse
|
93
|
Vigant F, Lee B. Hendra and nipah infection: pathology, models and potential therapies. Infect Disord Drug Targets 2011; 11:315-336. [PMID: 21488828 PMCID: PMC3253017 DOI: 10.2174/187152611795768097] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2009] [Accepted: 03/24/2010] [Indexed: 05/30/2023]
Abstract
The Paramyxoviridae family comprises of several genera that contain emerging or re-emerging threats for human and animal health with no real specific effective treatment available. Hendra and Nipah virus are members of a newly identified genus of emerging paramyxoviruses, Henipavirus. Since their discovery in the 1990s, henipaviruses outbreaks have been associated with high economic and public health threat potential. When compared to other paramyxoviruses, henipaviruses appear to have unique characteristics. Henipaviruses are zoonotic paramyxoviruses with a broader tropism than most other paramyxoviruses, and can cause severe acute encephalitis with unique features among viral encephalitides. There are currently no approved effective prophylactic or therapeutic treatments for henipavirus infections. Although ribavirin was empirically used and seemed beneficial during the biggest outbreak caused by one of these viruses, the Nipah virus, its efficacy is disputed in light of its lack of efficacy in several animal models of henipavirus infection. Nevertheless, because of its highly pathogenic nature, much effort has been spent in developing anti-henipavirus therapeutics. In this review we describe the unique features of henipavirus infections and the different strategies and animal models that have been developed so far in order to identify and test potential drugs to prevent or treat henipavirus infections. Some of these components have the potential to be broad-spectrum antivirals as they target effectors of viral pathogenecity common to other viruses. We will focus on small molecules or biologics, rather than vaccine strategies, that have been developed as anti-henipaviral therapeutics.
Collapse
Affiliation(s)
- Frederic Vigant
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA, USA 90095
| | - Benhur Lee
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA, USA 90095
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA 90095
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA 90095
| |
Collapse
|
94
|
Abstract
Nipah virus (NiV) is a recently emerged zoonotic paramyxovirus whose natural reservoirs are several species of Pteropus fruit bats. NiV provokes a widespread vasculitis often associated with severe encephalitis, with up to 75% mortality in humans. We have analyzed the pathogenesis of NiV infection, using human leukocyte cultures and the hamster animal model, which closely reproduces human NiV infection. We report that human lymphocytes and monocytes are not permissive for NiV and a low level of virus replication is detected only in dendritic cells. Interestingly, despite the absence of infection, lymphocytes could efficiently bind NiV and transfer infection to endothelial and Vero cells. This lymphocyte-mediated transinfection was inhibited after proteolytic digestion and neutralization by NiV-specific antibodies, suggesting that cells could transfer infectious virus to other permissive cells without the requirement for NiV internalization. In NiV-infected hamsters, leukocytes captured and carried NiV after intraperitoneal infection without themselves being productively infected. Such NiV-loaded mononuclear leukocytes transfer lethal NiV infection into naïve animals, demonstrating efficient virus transinfection in vivo. Altogether, these results reveal a remarkable capacity of NiV to hijack leukocytes as vehicles to transinfect host cells and spread the virus throughout the organism. This mode of virus transmission represents a rapid and potent method of NiV dissemination, which may contribute to its high pathogenicity.
Collapse
|
95
|
Walpita P, Barr J, Sherman M, Basler CF, Wang L. Vaccine potential of Nipah virus-like particles. PLoS One 2011; 6:e18437. [PMID: 21494680 PMCID: PMC3071823 DOI: 10.1371/journal.pone.0018437] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 03/07/2011] [Indexed: 11/19/2022] Open
Abstract
Nipah virus (NiV) was first recognized in 1998 in a zoonotic disease outbreak associated with highly lethal febrile encephalitis in humans and a predominantly respiratory disease in pigs. Periodic deadly outbreaks, documentation of person-to-person transmission, and the potential of this virus as an agent of agroterror reinforce the need for effective means of therapy and prevention. In this report, we describe the vaccine potential of NiV virus-like particles (NiV VLPs) composed of three NiV proteins G, F and M. Co-expression of these proteins under optimized conditions resulted in quantifiable amounts of VLPs with many virus-like/vaccine desirable properties including some not previously described for VLPs of any paramyxovirus: The particles were fusogenic, inducing syncytia formation; PCR array analysis showed NiV VLP-induced activation of innate immune defense pathways; the surface structure of NiV VLPs imaged by cryoelectron microscopy was dense, ordered, and repetitive, and consistent with similarly derived structure of paramyxovirus measles virus. The VLPs were composed of all the three viral proteins as designed, and their intracellular processing also appeared similar to NiV virions. The size, morphology and surface composition of the VLPs were consistent with the parental virus, and importantly, they retained their antigenic potential. Finally, these particles, formulated without adjuvant, were able to induce neutralizing antibody response in Balb/c mice. These findings indicate vaccine potential of these particles and will be the basis for undertaking future protective efficacy studies in animal models of NiV disease.
Collapse
Affiliation(s)
- Pramila Walpita
- Department of Microbiology, Center for Biodefense and Emerging Infectious Disease, University of Texas Medical Branch, Galveston, Texas, United States of America.
| | | | | | | | | |
Collapse
|
96
|
Interactions of human complement with virus particles containing the Nipah virus glycoproteins. J Virol 2011; 85:5940-8. [PMID: 21450814 DOI: 10.1128/jvi.00193-11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Complement is an innate immune response system that most animal viruses encounter during natural infections. We have tested the role of human complement in the neutralization of virus particles harboring the Nipah virus (NiV) glycoproteins. A luciferase-expressing vesicular stomatitis virus (VSV) pseudotype that contained the NiV fusion (F) and attachment (G) glycoproteins (NiVpp) showed dose- and time-dependent activation of human complement through the alternative pathway. In contrast to our findings with other paramyxoviruses, normal human serum (NHS) alone did not neutralize NiVpp infectivity in vitro, and electron microscopy demonstrated no significant deposition of complement component C3 on particles. This lack of NiVpp neutralization by NHS was not due to a global inhibition of complement pathways, since complement was found to significantly enhance neutralization by antibodies specific for the NiV F and G glycoproteins. Complement components C4 and C1q were necessary but not sufficient by themselves for the enhancement of antibody neutralization. Human complement also enhanced NiVpp neutralization by a soluble version of the NiV receptor EphrinB2, and this depended on components in the classical pathway. The ability of complement to enhance neutralization fell into one of two profiles: (i) anti-F monoclonal antibodies showed enhancement only at high and not low antibody concentrations, and (ii) anti-G monoclonal antibodies and EphrinB2 showed enhancement at both high and very low levels of antibody (e.g., 3.1 ng) or EphrinB2 (e.g., 2.5 ng). Together, these data establish the importance of human complement in the neutralization of particles containing the NiV glycoproteins and will help guide the design of more effective therapeutics that harness the potency of complement pathways.
Collapse
|
97
|
Complementing defective viruses that express separate paramyxovirus glycoproteins provide a new vaccine vector approach. J Virol 2010; 85:2004-11. [PMID: 21177820 DOI: 10.1128/jvi.01852-10] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Replication-defective vaccine vectors based on vesicular stomatitis virus (VSV) lacking its envelope glycoprotein gene (G) are highly effective in animal models. However, such ΔG vectors are difficult to grow because they require complementation with the VSV G protein. In addition, the complementing G protein induces neutralizing antibodies in animals and thus limits multiple vector applications. In the process of generating an experimental Nipah virus (a paramyxovirus) vaccine, we generated two defective VSVΔG vectors, each expressing one of the two Nipah virus (NiV) glycoproteins (G and F) that are both required for virus entry to host cells. These replication-defective VSV vectors were effective at generating NiV neutralizing antibody in mice. Most interestingly, we found that these two defective viruses could be grown together and passaged in tissue culture cells in the absence of VSV G complementation. This mixture of complementing defective viruses was also highly effective at generating NiV neutralizing antibody in animals. This novel approach to growing and producing a vaccine from two defective viruses could be generally applicable to vaccine production for other paramyxoviruses or for other viruses where the expression of at least two different proteins is required for viral entry. Such an approach minimizes biosafety concerns that could apply to single, replication-competent VSV recombinants expressing all proteins required for infection.
Collapse
|
98
|
Khetawat D, Broder CC. A functional henipavirus envelope glycoprotein pseudotyped lentivirus assay system. Virol J 2010; 7:312. [PMID: 21073718 PMCID: PMC2994542 DOI: 10.1186/1743-422x-7-312] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 11/12/2010] [Indexed: 12/12/2022] Open
Abstract
Background Hendra virus (HeV) and Nipah virus (NiV) are newly emerged zoonotic paramyxoviruses discovered during outbreaks in Queensland, Australia in 1994 and peninsular Malaysia in 1998/9 respectively and classified within the new Henipavirus genus. Both viruses can infect a broad range of mammalian species causing severe and often-lethal disease in humans and animals, and repeated outbreaks continue to occur. Extensive laboratory studies on the host cell infection stage of HeV and NiV and the roles of their envelope glycoproteins have been hampered by their highly pathogenic nature and restriction to biosafety level-4 (BSL-4) containment. To circumvent this problem, we have developed a henipavirus envelope glycoprotein pseudotyped lentivirus assay system using either a luciferase gene or green fluorescent protein (GFP) gene encoding human immunodeficiency virus type-1 (HIV-1) genome in conjunction with the HeV and NiV fusion (F) and attachment (G) glycoproteins. Results Functional retrovirus particles pseudotyped with henipavirus F and G glycoproteins displayed proper target cell tropism and entry and infection was dependent on the presence of the HeV and NiV receptors ephrinB2 or B3 on target cells. The functional specificity of the assay was confirmed by the lack of reporter-gene signals when particles bearing either only the F or only G glycoprotein were prepared and assayed. Virus entry could be specifically blocked when infection was carried out in the presence of a fusion inhibiting C-terminal heptad (HR-2) peptide, a well-characterized, cross-reactive, neutralizing human mAb specific for the henipavirus G glycoprotein, and soluble ephrinB2 and B3 receptors. In addition, the utility of the assay was also demonstrated by an examination of the influence of the cytoplasmic tail of F in its fusion activity and incorporation into pseudotyped virus particles by generating and testing a panel of truncation mutants of NiV and HeV F. Conclusions Together, these results demonstrate that a specific henipavirus entry assay has been developed using NiV or HeV F and G glycoprotein pseudotyped reporter-gene encoding retrovirus particles. This assay can be conducted safely under BSL-2 conditions and will be a useful tool for measuring henipavirus entry and studying F and G glycoprotein function in the context of virus entry, as well as in assaying and characterizing neutralizing antibodies and virus entry inhibitors.
Collapse
Affiliation(s)
- Dimple Khetawat
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland 20814, USA.
| | | |
Collapse
|
99
|
Defang GN, Khetawat D, Broder CC, Quinnan GV. Induction of neutralizing antibodies to Hendra and Nipah glycoproteins using a Venezuelan equine encephalitis virus in vivo expression system. Vaccine 2010; 29:212-20. [PMID: 21050901 DOI: 10.1016/j.vaccine.2010.10.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Revised: 10/13/2010] [Accepted: 10/20/2010] [Indexed: 10/18/2022]
Abstract
The emergence of Hendra Virus (HeV) and Nipah Virus (NiV) which can cause fatal infections in both animals and humans has triggered a search for an effective vaccine. Here, we have explored the potential for generating an effective humoral immune response to these zoonotic pathogens using an alphavirus-based vaccine platform. Groups of mice were immunized with Venezuelan equine encephalitis virus replicon particles (VRPs) encoding the attachment or fusion glycoproteins of either HeV or NiV. We demonstrate the induction of highly potent cross-reactive neutralizing antibodies to both viruses using this approach. Preliminary study suggested early enhancement in the antibody response with use of a modified version of VRP. Overall, these data suggest that the use of an alphavirus-derived vaccine platform might serve as a viable approach for the development of an effective vaccine against the henipaviruses.
Collapse
Affiliation(s)
- Gabriel N Defang
- Department of Preventive Medicine and Biometrics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | | | | | | |
Collapse
|
100
|
Inhibition of Nipah virus infection in vivo: targeting an early stage of paramyxovirus fusion activation during viral entry. PLoS Pathog 2010; 6:e1001168. [PMID: 21060819 PMCID: PMC2965769 DOI: 10.1371/journal.ppat.1001168] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 09/29/2010] [Indexed: 11/19/2022] Open
Abstract
In the paramyxovirus cell entry process, receptor binding triggers conformational changes in the fusion protein (F) leading to viral and cellular membrane fusion. Peptides derived from C-terminal heptad repeat (HRC) regions in F have been shown to inhibit fusion by preventing formation of the fusogenic six-helix bundle. We recently showed that the addition of a cholesterol group to HRC peptides active against Nipah virus targets these peptides to the membrane where fusion occurs, dramatically increasing their antiviral effect. In this work, we report that unlike the untagged HRC peptides, which bind to the postulated extended intermediate state bridging the viral and cell membranes, the cholesterol tagged HRC-derived peptides interact with F before the fusion peptide inserts into the target cell membrane, thus capturing an earlier stage in the F-activation process. Furthermore, we show that cholesterol tagging renders these peptides active in vivo: the cholesterol-tagged peptides cross the blood brain barrier, and effectively prevent and treat in an established animal model what would otherwise be fatal Nipah virus encephalitis. The in vivo efficacy of cholesterol-tagged peptides, and in particular their ability to penetrate the CNS, suggests that they are promising candidates for the prevention or therapy of infection by Nipah and other lethal paramyxoviruses.
Collapse
|