201
|
Sayama Y, Demetria C, Saito M, Azul RR, Taniguchi S, Fukushi S, Yoshikawa T, Iizuka I, Mizutani T, Kurane I, Malbas FF, Lupisan S, Catbagan DP, Animas SB, Morales RG, Lopez EL, Dazo KRC, Cruz MS, Olveda R, Saijo M, Oshitani H, Morikawa S. A seroepidemiologic study of Reston ebolavirus in swine in the Philippines. BMC Vet Res 2012; 8:82. [PMID: 22709971 PMCID: PMC3433389 DOI: 10.1186/1746-6148-8-82] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 06/12/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ebola viruses cause viral hemorrhagic fever in humans and non-human primates and are endemic in Africa. Reston ebolavirus (REBOV) has caused several epizootics in cynomolgus monkeys (Macaca fascicularis) but is not associated with any human disease. In late 2008, REBOV infections were identified in swine for the first time in the Philippines. METHODS A total of 215 swine sera collected at two REBOV-affected farms in 2008, in Pangasinan and Bulacan, were tested for the presence of REBOV-specific antibodies using multiple serodiagnosis systems. A total of 98 swine sera collected in a non-epizootic region, Tarlac, were also tested to clarify the prevalence of REBOV infection in the general swine population in the Philippines. RESULTS Some 70 % of swine sera at the affected farms were positive for REBOV antibodies in the multiple serodiagnosis systems. On the other hand, none of the swine sera collected in Tarlac showed positive reactions in any of the diagnosis systems. CONCLUSIONS The high prevalence of REBOV infection in swine in the affected farms in 2008 suggests that swine is susceptible for REBOV infection. The multiple serological assays used in the study are thought to be useful for future surveillance of REOBV infection in swine in the Philippines.
Collapse
Affiliation(s)
- Yusuke Sayama
- Department of Virology 1, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Deng MY, Millien M, Jacques-Simon R, Flanagan JK, Bracht AJ, Carrillo C, Barrette RW, Fabian A, Mohamed F, Moran K, Rowland J, Swenson SL, Jenkins-Moore M, Koster L, Thomsen BV, Mayr G, Pyburn D, Morales P, Shaw J, Burrage T, White W, McIntosh MT, Metwally S. Diagnosis of Porcine teschovirus encephalomyelitis in the Republic of Haiti. J Vet Diagn Invest 2012; 24:671-8. [DOI: 10.1177/1040638712445769] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Ming Y. Deng
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Max Millien
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Rodney Jacques-Simon
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - J. Keith Flanagan
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Alexa J. Bracht
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Consuelo Carrillo
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Roger W. Barrette
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Andrew Fabian
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Fawzi Mohamed
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Karen Moran
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Jessica Rowland
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Sabrina L. Swenson
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Melinda Jenkins-Moore
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Leo Koster
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Bruce V. Thomsen
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Gregory Mayr
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Dave Pyburn
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Paula Morales
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - John Shaw
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Thomas Burrage
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - William White
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Michael T. McIntosh
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| | - Samia Metwally
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National Veterinary Services Laboratories, Foreign Animal Disease Diagnostic Laboratory (Deng, Bracht, Carrillo, Barrette, Fabian, Mohamed, Moran, Rowland, Mayr, McIntosh, Metwally)
- Plum Island Animal Disease Center, Science and Technology Directorate of the Department of Homeland Security (Burrage, White)
- Greenport, NY; Animal Heath and Production, Ministry of Agriculture (Millien, Jacques-Simon), Institute of International Cooperation in Agriculture (Flanagan)
- Port-au-Prince, Haiti; USDA, APHIS, VS, National Veterinary Services Laboratories, Ames, IA (Swenson, Jenkins-Moore, Koster, Thomsen)
- USDA, APHIS, VS, Swine Health Program, Des Moines, IA (Pyburn)
| |
Collapse
|
203
|
Raut C, Yadav P, Towner J, Amman B, Erickson B, Cannon D, Sivaram A, Basu A, Nichol S, Mishra A, Mourya D. Isolation of a novel adenovirus from Rousettus leschenaultii bats from India. Intervirology 2012; 55:488-90. [PMID: 22572722 PMCID: PMC11932452 DOI: 10.1159/000337026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 02/02/2012] [Indexed: 11/19/2022] Open
Abstract
Surveillance work was initiated to study the presence of highly infectious diseases like Ebola-Reston, Marburg, Nipah and other possible viruses that are known to be found in the bat species and responsible for causing diseases in humans. A novel adenovirus was isolated from a common species of fruit bat (Rousettus leschenaultii) captured in Maharashtra State, India. Partial sequence analysis of the DNA polymerase gene shows this isolate to be a newly recognized member of the genus Mastadenovirus (family Adenoviridae), approximately 20% divergent at the nucleotide level from Japanese BatAdV, its closest known relative.
Collapse
Affiliation(s)
- C.G. Raut
- Microbial Containment Complex, National Institute of Virology, Pune, India
| | - P.D. Yadav
- Microbial Containment Complex, National Institute of Virology, Pune, India
| | - J.S. Towner
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Ga., USA
| | - B.R. Amman
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Ga., USA
| | - B.R. Erickson
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Ga., USA
| | - D.L. Cannon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Ga., USA
| | - A. Sivaram
- Microbial Containment Complex, National Institute of Virology, Pune, India
| | - A. Basu
- Microbial Containment Complex, National Institute of Virology, Pune, India
| | - S.T. Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Ga., USA
| | - A.C. Mishra
- Microbial Containment Complex, National Institute of Virology, Pune, India
| | - D.T. Mourya
- Microbial Containment Complex, National Institute of Virology, Pune, India
| |
Collapse
|
204
|
Olson SH, Reed P, Cameron KN, Ssebide BJ, Johnson CK, Morse SS, Karesh WB, Mazet JAK, Joly DO. Dead or alive: animal sampling during Ebola hemorrhagic fever outbreaks in humans. EMERGING HEALTH THREATS JOURNAL 2012; 5:EHTJ-5-9134. [PMID: 22558004 PMCID: PMC3342678 DOI: 10.3402/ehtj.v5i0.9134] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 03/11/2012] [Accepted: 03/12/2012] [Indexed: 11/17/2022]
Abstract
There are currently no widely accepted animal surveillance guidelines for human Ebola hemorrhagic fever (EHF) outbreak investigations to identify potential sources of Ebolavirus (EBOV) spillover into humans and other animals. Animal field surveillance during and following an outbreak has several purposes, from helping identify the specific animal source of a human case to guiding control activities by describing the spatial and temporal distribution of wild circulating EBOV, informing public health efforts, and contributing to broader EHF research questions. Since 1976, researchers have sampled over 10,000 individual vertebrates from areas associated with human EHF outbreaks and tested for EBOV or antibodies. Using field surveillance data associated with EHF outbreaks, this review provides guidance on animal sampling for resource-limited outbreak situations, target species, and in some cases which diagnostics should be prioritized to rapidly assess the presence of EBOV in animal reservoirs. In brief, EBOV detection was 32.7% (18/55) for carcasses (animals found dead) and 0.2% (13/5309) for live captured animals. Our review indicates that for the purposes of identifying potential sources of transmission from animals to humans and isolating suspected virus in an animal in outbreak situations, (1) surveillance of free-ranging non-human primate mortality and morbidity should be a priority, (2) any wildlife morbidity or mortality events should be investigated and may hold the most promise for locating virus or viral genome sequences, (3) surveillance of some bat species is worthwhile to isolate and detect evidence of exposure, and (4) morbidity, mortality, and serology studies of domestic animals should prioritize dogs and pigs and include testing for virus and previous exposure.
Collapse
Affiliation(s)
- Sarah H Olson
- Wildlife Health Program, Wildlife Conservation Society, Nanaimo, BC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
205
|
Yu X, Chen N, Wang L, Wu J, Zhou Z, Ni J, Li X, Zhai X, Shi J, Tian K. New genomic characteristics of highly pathogenic porcine reproductive and respiratory syndrome viruses do not lead to significant changes in pathogenicity. Vet Microbiol 2012; 158:291-9. [PMID: 22525010 DOI: 10.1016/j.vetmic.2012.02.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 02/16/2012] [Accepted: 02/23/2012] [Indexed: 11/25/2022]
Abstract
Highly pathogenic porcine reproductive and respiratory syndrome (HP-PRRS) initially emerged in China and currently prevails in other Asian countries as well, resulting in immense economic losses. HP-PRRS virus (HP-PRRSV) has undergone rapid evolution since its first recognition in 2006. To analyze the genomic and pathogenic characteristics of 2010 HP-PRRSV, we tested 919 clinical samples collected from China, Laos and Vietnam, sequenced 29 complete genomes of HP-PRRSV isolates, and determined the pathogenicity of seven HP-PRRS viruses isolated from 2006 to 2010. HP-PRRSV was detected from 45.2% (415/919) samples, while only 0.1% (1/919) was classical PRRSV, indicating that HP-PRRSV isolates with a unique discontinuous deletion of 30 amino acids (aa) in non-structural protein 2 (Nsp2) are still the predominant viruses. 2010 HP-PRRSV together with 2009 HP-PRRSV isolates form a new evolutionary branch based on phylogenetic analyses. The numbers of potential N-glycosylation sites are variable in major glycoprotein GP5 but are conserved in minor glycoproteins GP2, GP3 and GP4. Pathogenicity studies showed that HP-PRRS viruses isolated from 2006 to 2010 maintain similar level of high pathogenicity, which caused high fever (>41°C for at least four days), 100% morbidity, and 40-100% mortality in 4-10 weeks old pigs. Real time monitoring information from this study could help to understand the genetic and pathogenic evolution of HP-PRRSV and assist in the control of HP-PRRS in Asia.
Collapse
Affiliation(s)
- Xiuling Yu
- Veterinary Diagnostic Lab, China Animal Disease Control Center, No. 2 Yuanmingyuan West Rd., Haidian District, Beijing 100193, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Denner J, Tönjes RR. Infection barriers to successful xenotransplantation focusing on porcine endogenous retroviruses. Clin Microbiol Rev 2012; 25:318-43. [PMID: 22491774 PMCID: PMC3346299 DOI: 10.1128/cmr.05011-11] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Xenotransplantation may be a solution to overcome the shortage of organs for the treatment of patients with organ failure, but it may be associated with the transmission of porcine microorganisms and the development of xenozoonoses. Whereas most microorganisms may be eliminated by pathogen-free breeding of the donor animals, porcine endogenous retroviruses (PERVs) cannot be eliminated, since these are integrated into the genomes of all pigs. Human-tropic PERV-A and -B are present in all pigs and are able to infect human cells. Infection of ecotropic PERV-C is limited to pig cells. PERVs may adapt to host cells by varying the number of LTR-binding transcription factor binding sites. Like all retroviruses, they may induce tumors and/or immunodeficiencies. To date, all experimental, preclinical, and clinical xenotransplantations using pig cells, tissues, and organs have not shown transmission of PERV. Highly sensitive and specific methods have been developed to analyze the PERV status of donor pigs and to monitor recipients for PERV infection. Strategies have been developed to prevent PERV transmission, including selection of PERV-C-negative, low-producer pigs, generation of an effective vaccine, selection of effective antiretrovirals, and generation of animals transgenic for a PERV-specific short hairpin RNA inhibiting PERV expression by RNA interference.
Collapse
|
207
|
Abstract
Marburg and Ebola viruses cause a severe hemorrhagic disease in humans with high fatality rates. Early target cells of filoviruses are monocytes, macrophages, and dendritic cells. The infection spreads to the liver, spleen and later other organs by blood and lymph flow. A hallmark of filovirus infection is the depletion of non-infected lymphocytes; however, the molecular mechanisms leading to the observed bystander lymphocyte apoptosis are poorly understood. Also, there is limited knowledge about the fate of infected cells in filovirus disease. In this review we will explore what is known about the intracellular events leading to virus amplification and cell damage in filovirus infection. Furthermore, we will discuss how cellular dysfunction and cell death may correlate with disease pathogenesis.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; E-Mails: (J.O.); (R.B.C.)
- National Emerging Infectious Diseases Laboratories Institute, Boston University, 72 East Concord Street, Boston, MA 02118, USA
| | - Elena Ryabchikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Science, Pr. Lavrent’eva, 8, Novosibirsk 630090, Russian Federation; E-Mail:
| | - Ronald B. Corley
- Department of Microbiology, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; E-Mails: (J.O.); (R.B.C.)
- National Emerging Infectious Diseases Laboratories Institute, Boston University, 72 East Concord Street, Boston, MA 02118, USA
| | - Elke Mühlberger
- Department of Microbiology, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; E-Mails: (J.O.); (R.B.C.)
- National Emerging Infectious Diseases Laboratories Institute, Boston University, 72 East Concord Street, Boston, MA 02118, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-617-638-0336; Fax: +1-617-638-4286
| |
Collapse
|
208
|
An animal model that reflects human disease: the common marmoset (Callithrix jacchus). Curr Opin Virol 2012; 2:357-62. [PMID: 22709521 PMCID: PMC3378983 DOI: 10.1016/j.coviro.2012.02.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 02/01/2012] [Accepted: 02/03/2012] [Indexed: 11/23/2022]
Abstract
The common marmoset is a new world primate belonging to the Callitrichidae family weighing between 350 and 400 g. The marmoset has been shown to be an outstanding model for studying aging, reproduction, neuroscience, toxicology, and infectious disease. With regard to their susceptibility to infectious agents, they are exquisite NHP models for viral, protozoan and bacterial agents, as well as prions. The marmoset provides the advantages of a small animal model in high containment coupled with the immunological repertoire of a nonhuman primate and susceptibility to wild type, non-adapted viruses.
Collapse
|
209
|
Abstract
Influenza A virus reservoirs in animals have provided novel genetic elements leading to the emergence of global pandemics in humans. Most influenza A viruses circulate in waterfowl, but those that infect mammalian hosts are thought to pose the greatest risk for zoonotic spread to humans and the generation of pandemic or panzootic viruses. We have identified an influenza A virus from little yellow-shouldered bats captured at two locations in Guatemala. It is significantly divergent from known influenza A viruses. The HA of the bat virus was estimated to have diverged at roughly the same time as the known subtypes of HA and was designated as H17. The neuraminidase (NA) gene is highly divergent from all known influenza NAs, and the internal genes from the bat virus diverged from those of known influenza A viruses before the estimated divergence of the known influenza A internal gene lineages. Attempts to propagate this virus in cell cultures and chicken embryos were unsuccessful, suggesting distinct requirements compared with known influenza viruses. Despite its divergence from known influenza A viruses, the bat virus is compatible for genetic exchange with human influenza viruses in human cells, suggesting the potential capability for reassortment and contributions to new pandemic or panzootic influenza A viruses.
Collapse
|
210
|
Takada A. Filovirus tropism: cellular molecules for viral entry. Front Microbiol 2012; 3:34. [PMID: 22363323 PMCID: PMC3277274 DOI: 10.3389/fmicb.2012.00034] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 01/19/2012] [Indexed: 11/13/2022] Open
Abstract
In human and non-human primates, filoviruses (Ebola and Marburg viruses) cause severe hemorrhagic fever. Recently, other animals such as pigs and some species of fruit bats have also been shown to be susceptible to these viruses. While having a preference for some cell types such as hepatocytes, endothelial cells, dendritic cells, monocytes, and macrophages, filoviruses are known to be pantropic in infection of primates. The envelope glycoprotein (GP) is responsible for both receptor binding and fusion of the virus envelope with the host cell membrane. It has been demonstrated that filovirus GP interacts with multiple molecules for entry into host cells, whereas none of the cellular molecules so far identified as a receptor/co-receptor fully explains filovirus tissue tropism and host range. Available data suggest that the mucin-like region (MLR) on GP plays an important role in attachment to the preferred target cells, whose infection is likely involved in filovirus pathogenesis, whereas the MLR is not essential for the fundamental function of the GP in viral entry into cells in vitro. Further studies elucidating the mechanisms of cellular entry of filoviruses may shed light on the development of strategies for prophylaxis and treatment of Ebola and Marburg hemorrhagic fevers.
Collapse
Affiliation(s)
- Ayato Takada
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University Sapporo, Japan
| |
Collapse
|
211
|
dsRNA binding characterization of full length recombinant wild type and mutants Zaire ebolavirus VP35. Antiviral Res 2012; 93:354-63. [PMID: 22289166 PMCID: PMC7114247 DOI: 10.1016/j.antiviral.2012.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 12/16/2011] [Accepted: 01/15/2012] [Indexed: 12/13/2022]
Abstract
The Ebola viruses (EBOVs) VP35 protein is a multifunctional major virulence factor involved in EBOVs replication and evasion of the host immune system. EBOV VP35 is an essential component of the viral RNA polymerase, it is a key participant of the nucleocapsid assembly and it inhibits the innate immune response by antagonizing RIG-I like receptors through its dsRNA binding function and, hence, by suppressing the host type I interferon (IFN) production. Insights into the VP35 dsRNA recognition have been recently revealed by structural and functional analysis performed on its C-terminus protein. We report the biochemical characterization of the Zaire ebolavirus (ZEBOV) full-length recombinant VP35 (rVP35)–dsRNA binding function. We established a novel in vitro magnetic dsRNA binding pull down assay, determined the rVP35 optimal dsRNA binding parameters, measured the rVP35 equilibrium dissociation constant for heterologous in vitro transcribed dsRNA of different length and short synthetic dsRNA of 8 bp, and validated the assay for compound screening by assessing the inhibitory ability of auryntricarboxylic acid (IC50 value of 50 μg/mL). Furthermore, we compared the dsRNA binding properties of full length wt rVP35 with those of R305A, K309A and R312A rVP35 mutants, which were previously reported to be defective in dsRNA binding-mediated IFN inhibition, showing that the latter have measurably increased Kd values for dsRNA binding and modified migration patterns in mobility shift assays with respect to wt rVP35. Overall, these results provide the first characterization of the full-length wt and mutants VP35–dsRNA binding functions.
Collapse
|
212
|
Lethality and pathogenesis of airborne infection with filoviruses in A129 α/β −/− interferon receptor-deficient mice. J Med Microbiol 2012; 61:8-15. [DOI: 10.1099/jmm.0.036210-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
213
|
|
214
|
Abstract
Filoviruses (Ebola and Marburg viruses) cause severe hemorrhagic fever in humans and nonhuman primates. No effective prophylaxis or treatment for filovirus diseases is yet commercially available. Recent studies have advanced our knowledge of filovirus protein functions and interaction between viral and host factors in the replication cycle. Current findings on the ecology of filoviruses (i.e., natural infection of nonprimate animals and discovery of a new member of filoviruses in Europe) have also provided new insights into the epidemiology of Ebola and Marburg hemorrhagic fever. This article reviews the fundamental aspects of filovirus biology and the latest topics on filovirus research.
Collapse
Affiliation(s)
- Ayato Takada
- Hokkaido University Research Center for Zoonosis Control
| |
Collapse
|
215
|
|
216
|
Wang LF. Bats as a source of emerging zoonotic diseases – the interface with wildlife. MICROBIOLOGY AUSTRALIA 2012. [DOI: 10.1071/ma12150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Emerging infectious diseases (EIDs) are defined as infections that have newly appeared in a population or have undergone a rapid change in incidence or geographic location1. Since 1940s, more than 300 EIDs have been recorded2, most of which are viruses. Approximately 75% of human EIDs originated from animals. Of all the EIDs, zoonoses from wildlife represent the most significant threat to human health. Zoonotic EIDs have been identified in a variety of wildlife animals, including ungulates, carnivores, rodents, primates, bats and other mammal and non-mammal species. AIDS, the most significant EID of modern times, originated from non-human primates. Rodents have long been recognised as an important source of EIDs including hantavirus, plaque and lyme disease3. In the last few decades, bats (Order Chiroptera) have received growing attention as reservoirs for EIDs. Particularly, a number of high profile zoonotic viruses with significant human and animal morbidity and mortality have been linked to bat reservoirs4,5.
Collapse
|
217
|
Marsh GA, Haining J, Robinson R, Foord A, Yamada M, Barr JA, Payne J, White J, Yu M, Bingham J, Rollin PE, Nichol ST, Wang LF, Middleton D. Ebola Reston virus infection of pigs: clinical significance and transmission potential. J Infect Dis 2011; 204 Suppl 3:S804-9. [PMID: 21987755 DOI: 10.1093/infdis/jir300] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In 2008, Reston ebolavirus (REBOV) was isolated from pigs during a disease investigation in the Philippines. Porcine reproductive and respiratory syndrome virus (PRRSV) and porcine circovirus type 2 (PCV-2) infections were also confirmed in affected herds and the contribution of REBOV to the disease outbreak remains uncertain. We have conducted experimental challenge studies in 5-week-old pigs, with exposure of animals to 10(6) TCID(50) of a 2008 swine isolate of REBOV via either the oronasal or subcutaneous route. Replication of virus in internal organs and viral shedding from the nasopharynx were documented in the absence of clinical signs of disease in infected pigs. These observations confirm not only that asymptomatic infection of pigs with REBOV occurs, but that animals so affected pose a transmission risk to farm, veterinary, and abattoir workers.
Collapse
Affiliation(s)
- Glenn A Marsh
- Australian Animal Health Laboratory, Livestock Industries, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Geelong, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
de Wit E, Munster VJ, Metwally SA, Feldmann H. Assessment of rodents as animal models for Reston ebolavirus. J Infect Dis 2011; 204 Suppl 3:S968-72. [PMID: 21987777 DOI: 10.1093/infdis/jir330] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The emergence of Reston ebolavirus (REBOV) in domestic swine in the Philippines has caused a renewed interest in REBOV pathogenicity. Here, the use of different rodent species as animal disease models for REBOV was investigated. BALB/c and STAT1(-)(/-) mice, Hartley guinea pigs, and Syrian hamsters were inoculated intraperitoneally with REBOV strain Pennsylvania or Reston08-A. Although virus replication occurred in guinea pigs, hamsters, and STAT1(-/-) mice, progression to disease was only observed in STAT1(-)(/-) mice. Moreover, REBOV Pennsylvania was more pathogenic than REBOV Reston08-A in this model. Thus, STAT1(-)(/-) mice may be used for research of REBOV pathogenicity and intervention strategies.
Collapse
Affiliation(s)
- Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | | | | | | |
Collapse
|
219
|
Choi JH, Schafer SC, Zhang L, Kobinger GP, Juelich T, Freiberg AN, Croyle MA. A single sublingual dose of an adenovirus-based vaccine protects against lethal Ebola challenge in mice and guinea pigs. Mol Pharm 2011; 9:156-67. [PMID: 22149096 DOI: 10.1021/mp200392g] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Sublingual (SL) delivery, a noninvasive immunization method that bypasses the intestinal tract for direct entry into the circulation, was evaluated with an adenovirus (Ad5)-based vaccine for Ebola. Mice and guinea pigs were immunized via the intramuscular (IM), nasal (IN), oral (PO) and SL routes. SL immunization elicited strong transgene expression in and attracted CD11c(+) antigen presenting cells to the mucosa. A SL dose of 1 × 10⁸ infectious particles induced Ebola Zaire glycoprotein (ZGP)-specific IFN-γ⁺ T cells in spleen, bronchoalveolar lavage, mesenteric lymph nodes and submandibular lymph nodes (SMLN) of naive mice in a manner similar to the same dose given IN. Ex vivo CFSE and in vivo cytotoxic T lymphocyte (CTL) assays confirmed that SL immunization elicits a notable population of effector memory CD8+ T cells and strong CTL responses in spleen and SMLN. SL immunization induced significant ZGP-specific Th1 and Th2 type responses unaffected by pre-existing immunity (PEI) that protected mice and guinea pigs from lethal challenge. SL delivery protected more mice with PEI to Ad5 than IM injection. SL immunization also reduced systemic anti-Ad5 T and B cell responses in naive mice and those with PEI, suggesting that secondary immunizations could be highly effective for both populations.
Collapse
Affiliation(s)
- Jin Huk Choi
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | | | | | | | | | | | | |
Collapse
|
220
|
Taniguchi S, Watanabe S, Masangkay JS, Omatsu T, Ikegami T, Alviola P, Ueda N, Iha K, Fujii H, Ishii Y, Mizutani T, Fukushi S, Saijo M, Kurane I, Kyuwa S, Akashi H, Yoshikawa Y, Morikawa S. Reston Ebolavirus antibodies in bats, the Philippines. Emerg Infect Dis 2011; 17:1559-60. [PMID: 21801651 PMCID: PMC3381561 DOI: 10.3201/eid1708.101693] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
221
|
Ebola virus glycoprotein needs an additional trigger, beyond proteolytic priming for membrane fusion. PLoS Negl Trop Dis 2011; 5:e1395. [PMID: 22102923 PMCID: PMC3216919 DOI: 10.1371/journal.pntd.0001395] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 09/29/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Ebolavirus belongs to the family filoviridae and causes severe hemorrhagic fever in humans with 50-90% lethality. Detailed understanding of how the viruses attach to and enter new host cells is critical to development of medical interventions. The virus displays a trimeric glycoprotein (GP(1,2)) on its surface that is solely responsible for membrane attachment, virus internalization and fusion. GP(1,2) is expressed as a single peptide and is cleaved by furin in the host cells to yield two disulphide-linked fragments termed GP1 and GP2 that remain associated in a GP(1,2) trimeric, viral surface spike. After entry into host endosomes, GP(1,2) is enzymatically cleaved by endosomal cathepsins B and L, a necessary step in infection. However, the functional effects of the cleavage on the glycoprotein are unknown. PRINCIPAL FINDINGS We demonstrate by antibody binding and Hydrogen-Deuterium Exchange Mass Spectrometry (DXMS) of glycoproteins from two different ebolaviruses that although enzymatic priming of GP(1,2) is required for fusion, the priming itself does not initiate the required conformational changes in the ectodomain of GP(1,2). Further, ELISA binding data of primed GP(1,2) to conformational antibody KZ52 suggests that the low pH inside the endosomes also does not trigger dissociation of GP1 from GP2 to effect membrane fusion. SIGNIFICANCE The results reveal that the ebolavirus GP(1,2) ectodomain remains in the prefusion conformation upon enzymatic cleavage in low pH and removal of the glycan cap. The results also suggest that an additional endosomal trigger is necessary to induce the conformational changes in GP(1,2) and effect fusion. Identification of this trigger will provide further mechanistic insights into ebolavirus infection.
Collapse
|
222
|
Kühl A, Hoffmann M, Müller MA, Munster VJ, Gnirss K, Kiene M, Tsegaye TS, Behrens G, Herrler G, Feldmann H, Drosten C, Pöhlmann S. Comparative analysis of Ebola virus glycoprotein interactions with human and bat cells. J Infect Dis 2011; 204 Suppl 3:S840-9. [PMID: 21987760 PMCID: PMC3189982 DOI: 10.1093/infdis/jir306] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Infection with Ebola virus (EBOV) causes hemorrhagic fever in humans with high case-fatality rates. The EBOV-glycoprotein (EBOV-GP) facilitates viral entry and promotes viral release from human cells. African fruit bats are believed not to develop disease upon EBOV infection and have been proposed as a natural reservoir of EBOV. We compared EBOV-GP interactions with human cells and cells from African fruit bats. We found that susceptibility to EBOV-GP-dependent infection was not limited to bat cells from potential reservoir species, and we observed that GP displayed similar biological properties in human and bat cells. The only exception was GP localization, which was to a greater extent intracellular in bat cells as compared to human cells. Collectively, our results suggest that GP interactions with fruit bat and human cells are similar and do not limit EBOV tropism for certain bat species.
Collapse
Affiliation(s)
- Annika Kühl
- Institute of Virology, Hannover Medical School, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Negredo A, Palacios G, Vázquez-Morón S, González F, Dopazo H, Molero F, Juste J, Quetglas J, Savji N, de la Cruz Martínez M, Herrera JE, Pizarro M, Hutchison SK, Echevarría JE, Lipkin WI, Tenorio A. Discovery of an ebolavirus-like filovirus in europe. PLoS Pathog 2011; 7:e1002304. [PMID: 22039362 PMCID: PMC3197594 DOI: 10.1371/journal.ppat.1002304] [Citation(s) in RCA: 302] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 08/23/2011] [Indexed: 12/04/2022] Open
Abstract
Filoviruses, amongst the most lethal of primate pathogens, have only been reported as natural infections in sub-Saharan Africa and the Philippines. Infections of bats with the ebolaviruses and marburgviruses do not appear to be associated with disease. Here we report identification in dead insectivorous bats of a genetically distinct filovirus, provisionally named Lloviu virus, after the site of detection, Cueva del Lloviu, in Spain. A novel filovirus, provisionally named Lloviu virus (LLOV), was detected during the investigation of Miniopterus schreibersii die-offs in Cueva del Lloviu in southern Europe. LLOV is genetically distinct from other marburgviruses and ebolaviruses and is the first filovirus detected in Europe that was not imported from an endemic area in Africa. Filoviruses, amongst the most lethal of primate pathogens, have only been reported as natural infections in sub-Saharan Africa and the Philippines. Infections of bats with the ebolaviruses and marburgviruses do not appear to be associated with disease. Here we report identification of genetically distinct filovirus in dead insectivorous bats in caves in Spain.
Collapse
Affiliation(s)
- Ana Negredo
- National Center of Microbiology, (ISCIII), Madrid, Spain
| | - Gustavo Palacios
- Center for Infection and Immunity, and WHO Collaborating Centre for Diagnostics, Surveillance and Immunotherapeutics for Emerging Infectious and Zoonotic Diseases, Mailman School of Public Health of Columbia University, New York, New York, United States of America
- * E-mail:
| | | | - Félix González
- Grupo Asturiano para el Estudio y Conservacion de los Murciélagos, Posada de Llanera, Principado de Asturias, Spain
| | - Hernán Dopazo
- Evolutionary Genomics Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | - Javier Juste
- Evolutionary Biology Unit, Estación Biológica Doñana (CSIC), Sevilla, Spain
| | - Juan Quetglas
- Evolutionary Biology Unit, Estación Biológica Doñana (CSIC), Sevilla, Spain
| | - Nazir Savji
- Center for Infection and Immunity, and WHO Collaborating Centre for Diagnostics, Surveillance and Immunotherapeutics for Emerging Infectious and Zoonotic Diseases, Mailman School of Public Health of Columbia University, New York, New York, United States of America
| | | | - Jesus Enrique Herrera
- Center for Infection and Immunity, and WHO Collaborating Centre for Diagnostics, Surveillance and Immunotherapeutics for Emerging Infectious and Zoonotic Diseases, Mailman School of Public Health of Columbia University, New York, New York, United States of America
| | - Manuel Pizarro
- Service of Pathology, Veterinary Teaching Hospital, Veterinary School, Complutense University, Madrid, Spain
| | | | | | - W. Ian Lipkin
- Center for Infection and Immunity, and WHO Collaborating Centre for Diagnostics, Surveillance and Immunotherapeutics for Emerging Infectious and Zoonotic Diseases, Mailman School of Public Health of Columbia University, New York, New York, United States of America
| | | |
Collapse
|
224
|
Leroy EM, Gonzalez JP, Baize S. Ebola and Marburg haemorrhagic fever viruses: major scientific advances, but a relatively minor public health threat for Africa. Clin Microbiol Infect 2011; 17:964-76. [PMID: 21722250 DOI: 10.1111/j.1469-0691.2011.03535.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ebola and Marburg viruses are the only members of the Filoviridae family (order Mononegavirales), a group of viruses characterized by a linear, non-segmented, single-strand negative RNA genome. They are among the most virulent pathogens for humans and great apes, causing acute haemorrhagic fever and death within a matter of days. Since their discovery 50 years ago, filoviruses have caused only a few outbreaks, with 2317 clinical cases and 1671 confirmed deaths, which is negligible compared with the devastation caused by malnutrition and other infectious diseases prevalent in Africa (malaria, cholera, AIDS, dengue, tuberculosis …). Yet considerable human and financial resourses have been devoted to research on these viruses during the past two decades, partly because of their potential use as bioweapons. As a result, our understanding of the ecology, host interactions, and control of these viruses has improved considerably.
Collapse
Affiliation(s)
- E M Leroy
- Centre International de Recherches Médicales de Franceville, Franceville, Gabon.
| | | | | |
Collapse
|
225
|
Abstract
From the emergence of Hendra virus and Menangle virus in Australia to the global pandemics of severe acute respiratory syndrome and influenza viruses (both H5N1 and H1N1), there has been a surge of zoonotic virus outbreaks in the last two decades. Although the drivers for virus emergence remain poorly understood, the rate of discovery of new viruses is accelerating. This is due to a combination of true emergence of new pathogens and the advance of new technologies making rapid detection and characterisation possible. While molecular approaches will continue to lead the way in virus discovery, other technological platforms are required to increase the chance of success. The lessons learnt in the last 20 years confirm that the One Health approach, involving inclusive collaborations between physicians, veterinarians and other health and environmental professionals, will be the key to combating future zoonotic disease outbreaks.
Collapse
Affiliation(s)
- Lin-Fa Wang
- CSIRO Livestock Industries, Australian Animal Health Laboratory.
| |
Collapse
|
226
|
Ramanan P, Shabman RS, Brown CS, Amarasinghe GK, Basler CF, Leung DW. Filoviral immune evasion mechanisms. Viruses 2011; 3:1634-49. [PMID: 21994800 PMCID: PMC3187693 DOI: 10.3390/v3091634] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 08/15/2011] [Indexed: 01/09/2023] Open
Abstract
The Filoviridae family of viruses, which includes the genera Ebolavirus (EBOV) and Marburgvirus (MARV), causes severe and often times lethal hemorrhagic fever in humans. Filoviral infections are associated with ineffective innate antiviral responses as a result of virally encoded immune antagonists, which render the host incapable of mounting effective innate or adaptive immune responses. The Type I interferon (IFN) response is critical for establishing an antiviral state in the host cell and subsequent activation of the adaptive immune responses. Several filoviral encoded components target Type I IFN responses, and this innate immune suppression is important for viral replication and pathogenesis. For example, EBOV VP35 inhibits the phosphorylation of IRF-3/7 by the TBK-1/IKKε kinases in addition to sequestering viral RNA from detection by RIG-I like receptors. MARV VP40 inhibits STAT1/2 phosphorylation by inhibiting the JAK family kinases. EBOV VP24 inhibits nuclear translocation of activated STAT1 by karyopherin-α. The examples also represent distinct mechanisms utilized by filoviral proteins in order to counter immune responses, which results in limited IFN-α/β production and downstream signaling.
Collapse
Affiliation(s)
- Parameshwaran Ramanan
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
- Biochemistry Graduate Program, Iowa State University, Ames, IA 50011, USA
| | - Reed S. Shabman
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Craig S. Brown
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
- Biochemistry Undergraduate Program, Iowa State University, Ames, IA 50011, USA
| | - Gaya K. Amarasinghe
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
- Authors to whom correspondence should be addressed; (G.K.A); (C.F.B); (D.W.L.)
| | - Christopher F. Basler
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA
- Authors to whom correspondence should be addressed; (G.K.A); (C.F.B); (D.W.L.)
| | - Daisy W. Leung
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
- Authors to whom correspondence should be addressed; (G.K.A); (C.F.B); (D.W.L.)
| |
Collapse
|
227
|
Barrette RW, Xu L, Rowland JM, McIntosh MT. Current perspectives on the phylogeny of Filoviridae. INFECTION GENETICS AND EVOLUTION 2011; 11:1514-9. [PMID: 21742058 PMCID: PMC7106080 DOI: 10.1016/j.meegid.2011.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 06/24/2011] [Accepted: 06/25/2011] [Indexed: 11/27/2022]
Abstract
Sporadic fatal outbreaks of disease in humans and non-human primates caused by Ebola or Marburg viruses have driven research into the characterization of these viruses with the hopes of identifying host tropisms and potential reservoirs. Such an understanding of the relatedness of newly discovered filoviruses may help to predict risk factors for outbreaks of hemorrhagic disease in humans and/or non-human primates. Recent discoveries such as three distinct genotypes of Reston ebolavirus, unexpectedly discovered in domestic swine in the Philippines; as well as a new species, Bundibugyo ebolavirus; the recent discovery of Lloviu virus as a potential new genus, Cuevavirus, within Filoviridae; and germline integrations of filovirus-like sequences in some animal species bring new insights into the relatedness of filoviruses, their prevalence and potential for transmission to humans. These new findings reveal that filoviruses are more diverse and may have had a greater influence on the evolution of animals than previously thought. Herein we review these findings with regard to the implications for understanding the host range, prevalence and transmission of Filoviridae.
Collapse
Affiliation(s)
- Roger W Barrette
- Foreign Animal Disease Diagnostic Laboratory, National Veterinary Services Laboratories, Animal and Plant Health Inspection Services, USA
| | | | | | | |
Collapse
|
228
|
Bradfute SB, Bavari S. Correlates of immunity to filovirus infection. Viruses 2011; 3:982-1000. [PMID: 21994766 PMCID: PMC3185794 DOI: 10.3390/v3070982] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 06/14/2011] [Accepted: 06/16/2011] [Indexed: 12/13/2022] Open
Abstract
Filoviruses can cause severe, often fatal hemorrhagic fever in humans. Recent advances in vaccine and therapeutic drug development have provided encouraging data concerning treatment of these infections. However, relatively little is known about immune responses in fatal versus non-fatal filovirus infection. This review summarizes the published literature on correlates of immunity to filovirus infection, and highlights deficiencies in our knowledge on this topic. It is likely that there are several types of successful immune responses, depending on the type of filovirus, and the presence and timing of vaccination or drug treatment.
Collapse
Affiliation(s)
- Steven B Bradfute
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Maryland, MD 21702, USA.
| | | |
Collapse
|
229
|
Denner J. Infectious risk in xenotransplantation - what post-transplant screening for the human recipient? Xenotransplantation 2011; 18:151-7. [DOI: 10.1111/j.1399-3089.2011.00636.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
230
|
Ebolavirus delta-peptide immunoadhesins inhibit marburgvirus and ebolavirus cell entry. J Virol 2011; 85:8502-13. [PMID: 21697477 DOI: 10.1128/jvi.02600-10] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
With the exception of Reston and Lloviu viruses, filoviruses (marburgviruses, ebolaviruses, and "cuevaviruses") cause severe viral hemorrhagic fevers in humans. Filoviruses use a class I fusion protein, GP(1,2), to bind to an unknown, but shared, cell surface receptor to initiate virus-cell fusion. In addition to GP(1,2), ebolaviruses and cuevaviruses, but not marburgviruses, express two secreted glycoproteins, soluble GP (sGP) and small soluble GP (ssGP). All three glycoproteins have identical N termini that include the receptor-binding region (RBR) but differ in their C termini. We evaluated the effect of the secreted ebolavirus glycoproteins on marburgvirus and ebolavirus cell entry, using Fc-tagged recombinant proteins. Neither sGP-Fc nor ssGP-Fc bound to filovirus-permissive cells or inhibited GP(1,2)-mediated cell entry of pseudotyped retroviruses. Surprisingly, several Fc-tagged Δ-peptides, which are small C-terminal cleavage products of sGP secreted by ebolavirus-infected cells, inhibited entry of retroviruses pseudotyped with Marburg virus GP(1,2), as well as Marburg virus and Ebola virus infection in a dose-dependent manner and at low molarity despite absence of sequence similarity to filovirus RBRs. Fc-tagged Δ-peptides from three ebolaviruses (Ebola virus, Sudan virus, and Taï Forest virus) inhibited GP(1,2)-mediated entry and infection of viruses comparably to or better than the Fc-tagged RBRs, whereas the Δ-peptide-Fc of an ebolavirus nonpathogenic for humans (Reston virus) and that of an ebolavirus with lower lethality for humans (Bundibugyo virus) had little effect. These data indicate that Δ-peptides are functional components of ebolavirus proteomes. They join cathepsins and integrins as novel modulators of filovirus cell entry, might play important roles in pathogenesis, and could be exploited for the synthesis of powerful new antivirals.
Collapse
|
231
|
Serologic cross-reactivity of human IgM and IgG antibodies to five species of Ebola virus. PLoS Negl Trop Dis 2011; 5:e1175. [PMID: 21666792 PMCID: PMC3110169 DOI: 10.1371/journal.pntd.0001175] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 04/11/2011] [Indexed: 12/04/2022] Open
Abstract
Five species of Ebola virus (EBOV) have been identified, with nucleotide differences of 30–45% between species. Four of these species have been shown to cause Ebola hemorrhagic fever (EHF) in humans and a fifth species (Reston ebolavirus) is capable of causing a similar disease in non-human primates. While examining potential serologic cross-reactivity between EBOV species is important for diagnostic assays as well as putative vaccines, the nature of cross-reactive antibodies following EBOV infection has not been thoroughly characterized. In order to examine cross-reactivity of human serologic responses to EBOV, we developed antigen preparations for all five EBOV species, and compared serologic responses by IgM capture and IgG enzyme-linked immunosorbent assay (ELISA) in groups of convalescent diagnostic sera from outbreaks in Kikwit, Democratic Republic of Congo (n = 24), Gulu, Uganda (n = 20), Bundibugyo, Uganda (n = 33), and the Philippines (n = 18), which represent outbreaks due to four different EBOV species. For groups of samples from Kikwit, Gulu, and Bundibugyo, some limited IgM cross-reactivity was noted between heterologous sera-antigen pairs, however, IgM responses were largely stronger against autologous antigen. In some instances IgG responses were higher to autologous antigen than heterologous antigen, however, in contrast to IgM responses, we observed strong cross-reactive IgG antibody responses to heterologous antigens among all sets of samples. Finally, we examined autologous IgM and IgG antibody levels, relative to time following EHF onset, and observed early peaking and declining IgM antibody levels (by 80 days) and early development and persistence of IgG antibodies among all samples, implying a consistent pattern of antibody kinetics, regardless of EBOV species. Our findings demonstrate limited cross-reactivity of IgM antibodies to EBOV, however, the stronger tendency for cross-reactive IgG antibody responses can largely circumvent limitations in the utility of heterologous antigen for diagnostic assays and may assist in the development of antibody-mediated vaccines to EBOV. Ebola virus (EBOV) is a highly pathogenic virus, capable of causing Ebola hemorrhagic fever in humans and non-human primates. Five species of EBOV have been identified. To examine whether infection with one EBOV species results in antibodies that cross-react with other EBOV species, we selected groups of human diagnostic samples from four outbreaks, which were each due to a different EBOV species, and compared IgM and IgG responses by ELISA to each of the five EBOV species. For samples from an individual outbreak, we found limited IgM reactivity to species of EBOV other than the virus species the individual was infected with. In contrast, for all groups of outbreak samples we observed strong cross-reactive IgG antibodies to all EBOV species. Our study demonstrates that IgG antibody responses tend to be more cross-reactive than IgM antibody responses in people infected with EBOV, a finding that has implications for the development of diagnostic assays and vaccines to EBOV.
Collapse
|
232
|
Bradfute SB, Dye JM, Bavari S. Filovirus vaccines. HUMAN VACCINES 2011; 7:701-11. [PMID: 21519188 PMCID: PMC3219077 DOI: 10.4161/hv.7.6.15398] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 02/18/2011] [Accepted: 02/22/2011] [Indexed: 11/19/2022]
Abstract
Filoviruses can cause severe and often fatal hemorrhagic fever in humans and non-human primates (NHPs). Although there are currently no clinically proven treatments for filovirus disease, much progress has been made in recent years in the discovery of therapeutics and vaccines against these viruses. A variety of vaccine platforms have been shown to be effective against filovirus infection. This review summarizes the literature in this field, focusing on vaccines that have been shown to protect NHPs from infection. Furthermore, the uses of rodent models in vaccine development, as well as correlates of immunity, are discussed.
Collapse
Affiliation(s)
- Steven B Bradfute
- United States Army Medical Research Institute of Infectious Diseases, National Interagency Biodefense Campus; Fort Detrick, Frederick, MD, USA
| | | | | |
Collapse
|
233
|
Bausch DG. Ebola virus as a foodborne pathogen? Cause for consideration, but not panic. J Infect Dis 2011; 204:179-81. [PMID: 21571727 DOI: 10.1093/infdis/jir201] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
234
|
Kobinger GP, Leung A, Neufeld J, Richardson JS, Falzarano D, Smith G, Tierney K, Patel A, Weingartl HM. Replication, pathogenicity, shedding, and transmission of Zaire ebolavirus in pigs. J Infect Dis 2011; 204:200-8. [PMID: 21571728 DOI: 10.1093/infdis/jir077] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
UNLABELLED (See the editorial commentary by Bausch, on pages 179-81.) BACKGROUND Reston ebolavirus was recently detected in pigs in the Philippines. Specific antibodies were found in pig farmers, indicating exposure to the virus. This important observation raises the possibility that pigs may be susceptible to Ebola virus infection, including from other species, such as Zaire ebolavirus (ZEBOV), and can transmit to other susceptible hosts. METHODS This study investigated whether ZEBOV, a species commonly reemerging in central Africa, can replicate and induce disease in pigs and can be transmitted to naive animals. Domesticated Landrace pigs were challenged through mucosal exposure with a total of 1 ×10(6) plaque-forming units of ZEBOV and monitored for virus replication, shedding, and pathogenesis. Using similar conditions, virus transmission from infected to naive animals was evaluated in a second set of pigs. RESULTS Following mucosal exposure, pigs replicated ZEBOV to high titers (reaching 10(7) median tissue culture infective doses/mL), mainly in the respiratory tract, and developed severe lung pathology. Shedding from the oronasal mucosa was detected for up to 14 days after infection, and transmission was confirmed in all naive pigs cohabiting with inoculated animals. CONCLUSIONS These results shed light on the susceptibility of pigs to ZEBOV infection and identify an unexpected site of virus amplification and shedding linked to transmission of infectious virus.
Collapse
Affiliation(s)
- Gary P Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Smith TC, Harper AL, Nair R, Wardyn SE, Hanson BM, Ferguson DD, Dressler AE. Emerging swine zoonoses. Vector Borne Zoonotic Dis 2011; 11:1225-34. [PMID: 21395424 DOI: 10.1089/vbz.2010.0182] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The majority of emerging infectious diseases are zoonotic in origin. Swine represent a potential reservoir for many novel pathogens and may transmit these to humans via direct contact with live animals (such as swine farmers and large animal veterinarians), or to the general human population via contaminated meat. We review recent emerging microbes associated with swine and discuss public health implications.
Collapse
Affiliation(s)
- Tara C Smith
- Department of Epidemiology, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | | | | | | | | | |
Collapse
|
236
|
Abstract
Ebola viruses are the causative agents of a severe form of viral haemorrhagic fever in man, designated Ebola haemorrhagic fever, and are endemic in regions of central Africa. The exception is the species Reston Ebola virus, which has not been associated with human disease and is found in the Philippines. Ebola virus constitutes an important local public health threat in Africa, with a worldwide effect through imported infections and through the fear of misuse for biological terrorism. Ebola virus is thought to also have a detrimental effect on the great ape population in Africa. Case-fatality rates of the African species in man are as high as 90%, with no prophylaxis or treatment available. Ebola virus infections are characterised by immune suppression and a systemic inflammatory response that causes impairment of the vascular, coagulation, and immune systems, leading to multiorgan failure and shock, and thus, in some ways, resembling septic shock.
Collapse
Affiliation(s)
- 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
|
237
|
de Wit E, Feldmann H, Munster VJ. Tackling Ebola: new insights into prophylactic and therapeutic intervention strategies. Genome Med 2011; 3:5. [PMID: 21349211 PMCID: PMC3092090 DOI: 10.1186/gm219] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since its discovery in 1976, Ebolavirus has caused periodic outbreaks of viral hemorrhagic fever associated with severe and often fatal disease. Ebolavirus is endemic in Central Africa and the Philippines. Although there is currently no approved treatment available, the past 10 years has seen remarkable progress in our understanding of the pathogenicity of Ebolavirus and the development of prophylactic and post-exposure therapies against it. In vitro and in vivo experiments have shown that Ebolavirus pathogenicity is multifactorial, including viral and host determinants. Besides their function in the virus replication cycle, the viral glycoprotein, nucleoprotein, minor matrix protein and polymerase cofactor are viral determinants of pathogenicity, with evasion of the host innate and adaptive immune responses as the main mechanism. Although no licensed Ebolavirus vaccines are currently available, vaccine research in non-human primates, the 'gold standard' animal model for Ebolavirus, has produced several promising candidates. A combination of DNA vaccination and a recombinant adenovirus serotype 5 boost resulted in cross-protective immunity in non-human primates. A recombinant vesicular stomatitis vaccine vector protected non-human primates in pre- and post-exposure challenge studies. Several antiviral therapies are currently under investigation, but only a few of these have been tested in non-human primate models. Antisense therapies, in which oligonucleotides inhibit viral replication, have shown promising results in non-human primates following post-exposure treatment. In light of the severity of Ebolavirus disease and the observed increase in Ebolavirus outbreaks over the past decade, the expedited translation of potential candidate therapeutics and vaccines from bench to bedside is currently the most challenging task for the field. Here, we review the current state of Ebolavirus research, with emphasis on prophylactic and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, 903 South 4th Street, MT 59840, USA.
| | | | | |
Collapse
|
238
|
Falzarano D, Geisbert TW, Feldmann H. Progress in filovirus vaccine development: evaluating the potential for clinical use. Expert Rev Vaccines 2011; 10:63-77. [PMID: 21162622 PMCID: PMC3398800 DOI: 10.1586/erv.10.152] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Marburg and Ebola viruses cause severe hemorrhagic fever in humans and nonhuman primates. Currently, there are no effective treatments and no licensed vaccines; although a number of vaccine platforms have proven successful in animal models. The ideal filovirus vaccine candidate should be able to provide rapid protection following a single immunization, have the potential to work postexposure and be cross-reactive or multivalent against all Marburg virus strains and all relevant Ebola virus species and strains. Currently, there are multiple platforms that have provided prophylactic protection in nonhuman primates, including DNA, recombinant adenovirus serotype 5, recombinant human parainfluenza virus 3 and virus-like particles. In addition, a single platform, recombinant vesicular stomatitis virus, has demonstrated both prophylactic and postexposure protection in nonhuman primates. These results demonstrate that achieving a vaccine that is protective against filoviruses is possible; the challenge now is to prove its safety and efficacy in order to obtain a vaccine that is ready for human use.
Collapse
Affiliation(s)
- Darryl Falzarano
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA
| | - Thomas W Geisbert
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
239
|
Abstract
Platforms for pathogen discovery have improved since the days of Koch and Pasteur; nonetheless, the challenges of proving causation are at least as daunting as they were in the late 1800 s. Although we will almost certainly continue to accumulate low-hanging fruit, where simple relationships will be found between the presence of a cultivatable agent and a disease, these successes will be increasingly infrequent. The future of the field rests instead in our ability to follow footprints of infectious agents that cannot be characterized using classical microbiological techniques and to develop the laboratory and computational infrastructure required to dissect complex host-microbe interactions. I have tried to refine the criteria used by Koch and successors to prove linkage to disease. These refinements are working constructs that will continue to evolve in light of new technologies, new models, and new insights. What will endure is the excitement of the chase. Happy hunting!
Collapse
|
240
|
Kuhn JH, Becker S, Ebihara H, Geisbert TW, Johnson KM, Kawaoka Y, Lipkin WI, Negredo AI, Netesov SV, Nichol ST, Palacios G, Peters CJ, Tenorio A, Volchkov VE, Jahrling PB. Proposal for a revised taxonomy of the family Filoviridae: classification, names of taxa and viruses, and virus abbreviations. Arch Virol 2010; 155:2083-103. [PMID: 21046175 PMCID: PMC3074192 DOI: 10.1007/s00705-010-0814-x] [Citation(s) in RCA: 296] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 09/16/2010] [Indexed: 11/30/2022]
Abstract
The taxonomy of the family Filoviridae (marburgviruses and ebolaviruses) has changed several times since the discovery of its members, resulting in a plethora of species and virus names and abbreviations. The current taxonomy has only been partially accepted by most laboratory virologists. Confusion likely arose for several reasons: species names that consist of several words or which (should) contain diacritical marks, the current orthographic identity of species and virus names, and the similar pronunciation of several virus abbreviations in the absence of guidance for the correct use of vernacular names. To rectify this problem, we suggest (1) to retain the current species names Reston ebolavirus, Sudan ebolavirus, and Zaire ebolavirus, but to replace the name Cote d'Ivoire ebolavirus [sic] with Taï Forest ebolavirus and Lake Victoria marburgvirus with Marburg marburgvirus; (2) to revert the virus names of the type marburgviruses and ebolaviruses to those used for decades in the field (Marburg virus instead of Lake Victoria marburgvirus and Ebola virus instead of Zaire ebolavirus); (3) to introduce names for the remaining viruses reminiscent of jargon used by laboratory virologists but nevertheless different from species names (Reston virus, Sudan virus, Taï Forest virus), and (4) to introduce distinct abbreviations for the individual viruses (RESTV for Reston virus, SUDV for Sudan virus, and TAFV for Taï Forest virus), while retaining that for Marburg virus (MARV) and reintroducing that used over decades for Ebola virus (EBOV). Paying tribute to developments in the field, we propose (a) to create a new ebolavirus species (Bundibugyo ebolavirus) for one member virus (Bundibugyo virus, BDBV); (b) to assign a second virus to the species Marburg marburgvirus (Ravn virus, RAVV) for better reflection of now available high-resolution phylogeny; and (c) to create a new tentative genus (Cuevavirus) with one tentative species (Lloviu cuevavirus) for the recently discovered Lloviu virus (LLOV). Furthermore, we explain the etymological derivation of individual names, their pronunciation, and their correct use, and we elaborate on demarcation criteria for each taxon and virus.
Collapse
Affiliation(s)
- Jens H Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, National Interagency Biodefense Campus, B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Cross-platform evaluation of commercial real-time reverse transcription PCR master mix kits using a quantitative 5′nuclease assay for Ebola virus. Mol Cell Probes 2010; 24:370-5. [DOI: 10.1016/j.mcp.2010.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 08/06/2010] [Accepted: 08/09/2010] [Indexed: 11/19/2022]
|
242
|
Katzourakis A, Gifford RJ. Endogenous viral elements in animal genomes. PLoS Genet 2010; 6:e1001191. [PMID: 21124940 PMCID: PMC2987831 DOI: 10.1371/journal.pgen.1001191] [Citation(s) in RCA: 471] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 09/30/2010] [Indexed: 12/31/2022] Open
Abstract
Integration into the nuclear genome of germ line cells can lead to vertical inheritance of retroviral genes as host alleles. For other viruses, germ line integration has only rarely been documented. Nonetheless, we identified endogenous viral elements (EVEs) derived from ten non-retroviral families by systematic in silico screening of animal genomes, including the first endogenous representatives of double-stranded RNA, reverse-transcribing DNA, and segmented RNA viruses, and the first endogenous DNA viruses in mammalian genomes. Phylogenetic and genomic analysis of EVEs across multiple host species revealed novel information about the origin and evolution of diverse virus groups. Furthermore, several of the elements identified here encode intact open reading frames or are expressed as mRNA. For one element in the primate lineage, we provide statistically robust evidence for exaptation. Our findings establish that genetic material derived from all known viral genome types and replication strategies can enter the animal germ line, greatly broadening the scope of paleovirological studies and indicating a more significant evolutionary role for gene flow from virus to animal genomes than has previously been recognized. The presence of retrovirus sequences in animal genomes has been recognized since the 1970s, but is readily explained by the fact that these viruses integrate into chromosomal DNA as part of their normal replication cycle. Unexpectedly, however, we identified a large and diverse population of sequences in animal genomes that are derived from non-retroviral viruses. Analysis of these sequences—which represent all known virus genome types and replication strategies—reveals new information about the evolutionary history of viruses, in many cases providing the first and only direct evidence for their ancient origins. Additionally, we provide evidence that the functionality of one of these sequences has been maintained in the host genome over many millions of years, raising the possibility that captured viral sequences may have played a larger than expected role in host evolution.
Collapse
Affiliation(s)
- Aris Katzourakis
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- * E-mail: (RJG); (AK)
| | - Robert J. Gifford
- Aaron Diamond AIDS Research Center, New York, New York, United States of America
- * E-mail: (RJG); (AK)
| |
Collapse
|
243
|
Abstract
For more than 30 years the filoviruses, Marburg virus and Ebola virus, have been associated with periodic outbreaks of hemorrhagic fever that produce severe and often fatal disease. The filoviruses are endemic primarily in resource-poor regions in Central Africa and are also potential agents of bioterrorism. Although no vaccines or antiviral drugs for Marburg or Ebola are currently available, remarkable progress has been made over the last decade in developing candidate preventive vaccines against filoviruses in nonhuman primate models. Due to the generally remote locations of filovirus outbreaks, a single-injection vaccine is desirable. Among the prospective vaccines that have shown efficacy in nonhuman primate models of filoviral hemorrhagic fever, two candidates, one based on a replication-defective adenovirus serotype 5 and the other on a recombinant VSV (rVSV), were shown to provide complete protection to nonhuman primates when administered as a single injection. The rVSV-based vaccine has also shown utility when administered for postexposure prophylaxis against filovirus infections. A VSV-based Ebola vaccine was recently used to manage a potential laboratory exposure.
Collapse
Affiliation(s)
- Thomas W Geisbert
- Galveston National Laboratory1 and Department of Microbiology and Immunology2, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, USA.
| | | | | |
Collapse
|
244
|
Ledgerwood JE, Costner P, Desai N, Holman L, Enama ME, Yamshchikov G, Mulangu S, Hu Z, Andrews CA, Sheets RA, Koup RA, Roederer M, Bailer R, Mascola JR, Pau MG, Sullivan NJ, Goudsmit J, Nabel GJ, Graham BS. A replication defective recombinant Ad5 vaccine expressing Ebola virus GP is safe and immunogenic in healthy adults. Vaccine 2010; 29:304-13. [PMID: 21034824 DOI: 10.1016/j.vaccine.2010.10.037] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 09/17/2010] [Accepted: 10/13/2010] [Indexed: 10/18/2022]
Abstract
Ebola virus causes irregular outbreaks of severe hemorrhagic fever in equatorial Africa. Case mortality remains high; there is no effective treatment and outbreaks are sporadic and unpredictable. Studies of Ebola virus vaccine platforms in non-human primates have established that the induction of protective immunity is possible and safety and human immunogenicity has been demonstrated in a previous Phase I clinical trial of a 1st generation Ebola DNA vaccine. We now report the safety and immunogenicity of a recombinant adenovirus serotype 5 (rAd5) vaccine encoding the envelope glycoprotein (GP) from the Zaire and Sudan Ebola virus species, in a randomized, placebo-controlled, double-blinded, dose escalation, Phase I human study. Thirty-one healthy adults received vaccine at 2×10(9) (n=12), or 2×10(10) (n=11) viral particles or placebo (n=8) as an intramuscular injection. Antibody responses were assessed by ELISA and neutralizing assays; and T cell responses were assessed by ELISpot and intracellular cytokine staining assays. This recombinant Ebola virus vaccine was safe and subjects developed antigen specific humoral and cellular immune responses.
Collapse
Affiliation(s)
- J E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 40 Convent Drive, Bethesda, MD 20892-3017, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Wauquier N, Becquart P, Padilla C, Baize S, Leroy EM. Human fatal zaire ebola virus infection is associated with an aberrant innate immunity and with massive lymphocyte apoptosis. PLoS Negl Trop Dis 2010; 4:e837. [PMID: 20957152 PMCID: PMC2950153 DOI: 10.1371/journal.pntd.0000837] [Citation(s) in RCA: 284] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 09/03/2010] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Ebolavirus species Zaire (ZEBOV) causes highly lethal hemorrhagic fever, resulting in the death of 90% of patients within days. Most information on immune responses to ZEBOV comes from in vitro studies and animal models. The paucity of data on human immune responses to this virus is mainly due to the fact that most outbreaks occur in remote areas. Published studies in this setting, based on small numbers of samples and limited panels of immunological markers, have given somewhat different results. METHODOLOGY/PRINCIPAL FINDINGS Here, we studied a unique collection of 56 blood samples from 42 nonsurvivors and 14 survivors, obtained during the five outbreaks that occurred between 1996 and 2003 in Gabon and Republic of Congo. Using Luminex technology, we assayed 50 cytokines in all 56 samples and performed phenotypic analyses by flow cytometry. We found that fatal outcome was associated with hypersecretion of numerous proinflammatory cytokines (IL-1β, IL-1RA, IL-6, IL-8, IL-15 and IL-16), chemokines and growth factors (MIP-1α, MIP-1β, MCP-1, M-CSF, MIF, IP-10, GRO-α and eotaxin). Interestingly, no increase of IFNα2 was detected in patients. Furthermore, nonsurvivors were also characterized by very low levels of circulating cytokines produced by T lymphocytes (IL-2, IL-3, IL-4, IL-5, IL-9, IL-13) and by a significant drop of CD3+CD4+ and CD3+CD8+ peripheral cells as well as a high increase in CD95 expression on T lymphocytes. CONCLUSIONS/SIGNIFICANCE This work, the largest study to be conducted to date in humans, showed that fatal outcome is associated with aberrant innate immune responses and with global suppression of adaptive immunity. The innate immune reaction was characterized by a "cytokine storm," with hypersecretion of numerous proinflammatory cytokines, chemokines and growth factors, and by the noteworthy absence of antiviral IFNα2. Immunosuppression was characterized by very low levels of circulating cytokines produced by T lymphocytes and by massive loss of peripheral CD4 and CD8 lymphocytes, probably through Fas/FasL-mediated apoptosis.
Collapse
Affiliation(s)
- Nadia Wauquier
- Unité des Maladies Virales Émergentes, Centre International de Recherches Médicales de Franceville, Franceville, Gabon
| | - Pierre Becquart
- Unité des Maladies Virales Émergentes, Centre International de Recherches Médicales de Franceville, Franceville, Gabon
- UMR190 Emergence des Pathologies Virales, Université Aix-Marseille II and Institut de Recherche pour le Développement, Marseille, France
| | - Cindy Padilla
- Unité des Maladies Virales Émergentes, Centre International de Recherches Médicales de Franceville, Franceville, Gabon
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, IFR128-Biosciences Gerland-Lyon Sud, Lyon, France
| | - Eric M. Leroy
- Unité des Maladies Virales Émergentes, Centre International de Recherches Médicales de Franceville, Franceville, Gabon
- UMR190 Emergence des Pathologies Virales, Université Aix-Marseille II and Institut de Recherche pour le Développement, Marseille, France
| |
Collapse
|
246
|
Enzyme-linked immunosorbent assay for detection of filovirus species-specific antibodies. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1723-8. [PMID: 20861331 DOI: 10.1128/cvi.00170-10] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several enzyme-linked immunosorbent assays (ELISAs) for the detection of filovirus-specific antibodies have been developed. However, diagnostic methods to distinguish antibodies specific to the respective species of filoviruses, which provide the basis for serological classification, are not readily available. We established an ELISA using His-tagged secreted forms of the transmembrane glycoproteins (GPs) of five different Ebola virus (EBOV) species and one Marburg virus (MARV) strain as antigens for the detection of filovirus species-specific antibodies. The GP-based ELISA was evaluated by testing antisera collected from mice immunized with virus-like particles as well as from humans and nonhuman primates infected with EBOV or MARV. In our ELISA, little cross-reactivity of IgG antibodies was observed in most of the mouse antisera. Although sera and plasma from some patients and monkeys showed notable cross-reactivity with the GPs from multiple filovirus species, the highest reactions of IgG were uniformly detected against the GP antigen homologous to the virus species that infected individuals. We further confirmed that MARV-specific IgM antibodies were specifically detected in specimens collected from patients during the acute phase of infection. These results demonstrate the usefulness of our ELISA for diagnostics as well as ecological and serosurvey studies.
Collapse
|
247
|
Beeching NJ, Fletcher TE, Hill DR, Thomson GL. Travellers and viral haemorrhagic fevers: what are the risks? Int J Antimicrob Agents 2010; 36 Suppl 1:S26-35. [PMID: 20705436 DOI: 10.1016/j.ijantimicag.2010.06.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Viral haemorrhagic fevers (VHF) are caused by zoonotic viral infections transmitted to humans directly or by ticks or mosquitoes. The overall risk to travellers is conservatively estimated at <1 in 1 million travel episodes to African countries where infection is present, and febrile patients returning from these countries are at least 1000 times more likely to have malaria than Lassa fever or another VHF. No cases have been reported in fellow travellers exposed to a travelling case and only one asymptomatic seroconversion (to Lassa) has been reported in over 2000 contacts following care of VHF cases in modern Western hospital settings. However, healthcare-associated transmission of infection has been a major problem in some endemic settings. The potential for healthcare-associated infection and the threats posed by unrecognised or new agents necessitate a high index of suspicion and a standardised risk assessment approach to febrile travellers. Travel-related hantavirus infections are increasingly being reported from Europe and the Americas. This article summarises the epidemiology and reports of travel-related VHF cases in the past 40 years, together with strategies for their recognition, management and prevention.
Collapse
Affiliation(s)
- Nick J Beeching
- Tropical and Infectious Disease Unit, Royal Liverpool University Hospital, Liverpool, UK.
| | | | | | | |
Collapse
|
248
|
Hayman DTS, Emmerich P, Yu M, Wang LF, Suu-Ire R, Fooks AR, Cunningham AA, Wood JLN. Long-term survival of an urban fruit bat seropositive for Ebola and Lagos bat viruses. PLoS One 2010; 5:e11978. [PMID: 20694141 PMCID: PMC2915915 DOI: 10.1371/journal.pone.0011978] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 07/12/2010] [Indexed: 11/26/2022] Open
Abstract
Ebolaviruses (EBOV) (family Filoviridae) cause viral hemorrhagic fevers in humans and non-human primates when they spill over from their wildlife reservoir hosts with case fatality rates of up to 90%. Fruit bats may act as reservoirs of the Filoviridae. The migratory fruit bat, Eidolon helvum, is common across sub-Saharan Africa and lives in large colonies, often situated in cities. We screened sera from 262 E. helvum using indirect fluorescent tests for antibodies against EBOV subtype Zaire. We detected a seropositive bat from Accra, Ghana, and confirmed this using western blot analysis. The bat was also seropositive for Lagos bat virus, a Lyssavirus, by virus neutralization test. The bat was fitted with a radio transmitter and was last detected in Accra 13 months after release post-sampling, demonstrating long-term survival. Antibodies to filoviruses have not been previously demonstrated in E. helvum. Radio-telemetry data demonstrates long-term survival of an individual bat following exposure to viruses of families that can be highly pathogenic to other mammal species. Because E. helvum typically lives in large urban colonies and is a source of bushmeat in some regions, further studies should determine if this species forms a reservoir for EBOV from which spillover infections into the human population may occur.
Collapse
Affiliation(s)
- David T S Hayman
- Cambridge Infectious Diseases Consortium, University of Cambridge, Cambridge, UK.
| | | | | | | | | | | | | | | |
Collapse
|
249
|
Belyi VA, Levine AJ, Skalka AM. Unexpected inheritance: multiple integrations of ancient bornavirus and ebolavirus/marburgvirus sequences in vertebrate genomes. PLoS Pathog 2010; 6:e1001030. [PMID: 20686665 PMCID: PMC2912400 DOI: 10.1371/journal.ppat.1001030] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 07/02/2010] [Indexed: 01/01/2023] Open
Abstract
Vertebrate genomes contain numerous copies of retroviral sequences, acquired over the course of evolution. Until recently they were thought to be the only type of RNA viruses to be so represented, because integration of a DNA copy of their genome is required for their replication. In this study, an extensive sequence comparison was conducted in which 5,666 viral genes from all known non-retroviral families with single-stranded RNA genomes were matched against the germline genomes of 48 vertebrate species, to determine if such viruses could also contribute to the vertebrate genetic heritage. In 19 of the tested vertebrate species, we discovered as many as 80 high-confidence examples of genomic DNA sequences that appear to be derived, as long ago as 40 million years, from ancestral members of 4 currently circulating virus families with single strand RNA genomes. Surprisingly, almost all of the sequences are related to only two families in the Order Mononegavirales: the Bornaviruses and the Filoviruses, which cause lethal neurological disease and hemorrhagic fevers, respectively. Based on signature landmarks some, and perhaps all, of the endogenous virus-like DNA sequences appear to be LINE element-facilitated integrations derived from viral mRNAs. The integrations represent genes that encode viral nucleocapsid, RNA-dependent-RNA-polymerase, matrix and, possibly, glycoproteins. Integrations are generally limited to one or very few copies of a related viral gene per species, suggesting that once the initial germline integration was obtained (or selected), later integrations failed or provided little advantage to the host. The conservation of relatively long open reading frames for several of the endogenous sequences, the virus-like protein regions represented, and a potential correlation between their presence and a species' resistance to the diseases caused by these pathogens, are consistent with the notion that their products provide some important biological advantage to the species. In addition, the viruses could also benefit, as some resistant species (e.g. bats) may serve as natural reservoirs for their persistence and transmission. Given the stringent limitations imposed in this informatics search, the examples described here should be considered a low estimate of the number of such integration events that have persisted over evolutionary time scales. Clearly, the sources of genetic information in vertebrate genomes are much more diverse than previously suspected.
Collapse
Affiliation(s)
- Vladimir A. Belyi
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, New Jersey, United States of America
| | - Arnold J. Levine
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, New Jersey, United States of America
| | - Anna Marie Skalka
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
250
|
Abstract
The filoviruses, Ebola and Marburg, utilize a multifaceted mechanism for assembly and budding of infectious virions from mammalian cells. Growing evidence not only demonstrates the importance of multiple viral proteins for efficient assembly and budding, but also the exploitation of various host proteins/pathways by the virus during this late stage of filovirus replication, including endocytic compartments, vacuolar protein sorting pathways, ubiquitination machinery, lipid rafts and cytoskeletal components. Continued elucidation of these complex and orchestrated virus-host interactions will provide a fundamental understanding of the molecular mechanisms of filovirus assembly/budding and ultimately lead to the development of novel viral- and/or host-oriented therapeutics to inhibit filovirus egress and spread. This article will focus on the most recent studies on host interactions and modulation of filovirus budding and summarize the key findings from these investigations.
Collapse
Affiliation(s)
- Yuliang Liu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce St., Philadelphia, PA 19104, USA
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce St., Philadelphia, PA 19104, USA
| |
Collapse
|