1
|
Ebisine K, Quist D, Findlay-Wilson S, Kennedy E, Dowall S. A Review of Nonhuman Primate Models of Rift Valley Fever Virus Infection: Progress, Challenge Strains, and Future Directions. Pathogens 2024; 13:856. [PMID: 39452727 PMCID: PMC11510021 DOI: 10.3390/pathogens13100856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Rift Valley fever (RVF) is a mosquito-borne viral disease that primarily affects animals, especially ruminants, but has the capacity to infect humans and result in outbreaks. Infection with the causative agent, RVF virus (RVFV), causes severe disease in domestic animals, especially sheep, resulting in fever, anorexia, immobility, abortion, and high morbidity and mortality rates in neonate animals. Humans become infected through exposure to infected animals and, less frequently, directly via a mosquito bite. A greater awareness of RVFV and its epidemic potential has resulted in increased investment in the development of interventions, especially vaccines. There is currently no substitute for the use of animal models in order to evaluate these vaccines. As outbreaks of RVF disease are difficult to predict or model, conducting Phase III clinical trials will likely not be feasible. Therefore, representative animal model systems are essential for establishing efficacy data to support licensure. Nonhuman primate (NHP) species are often chosen due to their closeness to humans, reflecting similar susceptibility and disease kinetics. This review covers the use of NHP models in RVFV research, with much of the work having been conducted in rhesus macaques and common marmosets. The future direction of RVF work conducted in NHP is discussed in anticipation of the importance of it being a key element in the development and approval of a human vaccine.
Collapse
Affiliation(s)
| | | | | | | | - Stuart Dowall
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK; (K.E.); (D.Q.); (S.F.-W.); (E.K.)
| |
Collapse
|
2
|
Alkan C, Jurado-Cobena E, Ikegami T. Advancements in Rift Valley fever vaccines: a historical overview and prospects for next generation candidates. NPJ Vaccines 2023; 8:171. [PMID: 37925544 PMCID: PMC10625542 DOI: 10.1038/s41541-023-00769-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/18/2023] [Indexed: 11/06/2023] Open
Abstract
Rift Valley fever (RVF) is a zoonotic viral disease transmitted by mosquitoes and causes abortion storms, fetal malformations, and newborn animal deaths in livestock ruminants. In humans, RVF can manifest as hemorrhagic fever, encephalitis, or retinitis. Outbreaks of RVF have been occurring in Africa since the early 20th century and continue to pose a threat to both humans and animals in various regions such as Africa, Madagascar, the Comoros, Saudi Arabia, and Yemen. The development of RVF vaccines is crucial in preventing mortality and morbidity and reducing the spread of the virus. While several veterinary vaccines have been licensed in endemic countries, there are currently no licensed RVF vaccines for human use. This review provides an overview of the existing RVF vaccines, as well as potential candidates for future studies on RVF vaccine development, including next-generation vaccines that show promise in combating the disease in both humans and animals.
Collapse
Affiliation(s)
- Cigdem Alkan
- Department of Pathology, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Eduardo Jurado-Cobena
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Tetsuro Ikegami
- Department of Pathology, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA.
- The Sealy Institute for Vaccine Sciences, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA.
- The Center for Biodefense and Emerging Infectious Diseases, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA.
| |
Collapse
|
3
|
Xu Y, Wang X, Jiang L, Zhou Y, Liu Y, Wang F, Zhang L. Natural hosts and animal models for Rift Valley fever phlebovirus. Front Vet Sci 2023; 10:1258172. [PMID: 37929288 PMCID: PMC10621046 DOI: 10.3389/fvets.2023.1258172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Rift Valley fever phlebovirus (RVFV) is a zoonotic mosquito-transmitted arbovirus, presenting a serious threat to humans and animals. Susceptible hosts are of great significance for the prevention of RVFV. Appropriate animal models are helpful to better understand the onset and development of diseases, as well as the control measures and vaccine research. This review focuses on the role of animal hosts in the maintenance of the virus, and summarizes the host range of RVFV. We list some common animal models in the process of RVFV research, which would provide some important insights into the prevention and treatment of RVFV, as well as the study of Rift Valley fever (RVF) pathogenesis and vaccines.
Collapse
Affiliation(s)
- Yuqing Xu
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lu Jiang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yixuan Zhou
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yihan Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fei Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- School of Laboratory Animal and Shandong Laboratory Animal Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Leiliang Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
4
|
Nair N, Osterhaus ADME, Rimmelzwaan GF, Prajeeth CK. Rift Valley Fever Virus-Infection, Pathogenesis and Host Immune Responses. Pathogens 2023; 12:1174. [PMID: 37764982 PMCID: PMC10535968 DOI: 10.3390/pathogens12091174] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Rift Valley Fever Virus is a mosquito-borne phlebovirus causing febrile or haemorrhagic illness in ruminants and humans. The virus can prevent the induction of the antiviral interferon response through its NSs proteins. Mutations in the NSs gene may allow the induction of innate proinflammatory immune responses and lead to attenuation of the virus. Upon infection, virus-specific antibodies and T cells are induced that may afford protection against subsequent infections. Thus, all arms of the adaptive immune system contribute to prevention of disease progression. These findings will aid the design of vaccines using the currently available platforms. Vaccine candidates have shown promise in safety and efficacy trials in susceptible animal species and these may contribute to the control of RVFV infections and prevention of disease progression in humans and ruminants.
Collapse
|
5
|
Chapman NS, Hulswit RJG, Westover JLB, Stass R, Paesen GC, Binshtein E, Reidy JX, Engdahl TB, Handal LS, Flores A, Gowen BB, Bowden TA, Crowe JE. Multifunctional human monoclonal antibody combination mediates protection against Rift Valley fever virus at low doses. Nat Commun 2023; 14:5650. [PMID: 37704627 PMCID: PMC10499838 DOI: 10.1038/s41467-023-41171-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 08/22/2023] [Indexed: 09/15/2023] Open
Abstract
The zoonotic Rift Valley fever virus (RVFV) can cause severe disease in humans and has pandemic potential, yet no approved vaccine or therapy exists. Here we describe a dual-mechanism human monoclonal antibody (mAb) combination against RVFV that is effective at minimal doses in a lethal mouse model of infection. We structurally analyze and characterize the binding mode of a prototypical potent Gn domain-A-binding antibody that blocks attachment and of an antibody that inhibits infection by abrogating the fusion process as previously determined. Surprisingly, the Gn domain-A antibody does not directly block RVFV Gn interaction with the host receptor low density lipoprotein receptor-related protein 1 (LRP1) as determined by a competitive assay. This study identifies a rationally designed combination of human mAbs deserving of future investigation for use in humans against RVFV infection. Using a two-pronged mechanistic approach, we demonstrate the potent efficacy of a rationally designed combination mAb therapeutic.
Collapse
Affiliation(s)
- Nathaniel S Chapman
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ruben J G Hulswit
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Jonna L B Westover
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322, USA
| | - Robert Stass
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Guido C Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Elad Binshtein
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joseph X Reidy
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Taylor B Engdahl
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Laura S Handal
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Alejandra Flores
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Brian B Gowen
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322, USA
| | - Thomas A Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
6
|
Chapman NS, Zhao H, Kose N, Westover JB, Kalveram B, Bombardi R, Rodriguez J, Sutton R, Genualdi J, LaBeaud AD, Mutuku FM, Pittman PR, Freiberg AN, Gowen BB, Fremont DH, Crowe JE. Potent neutralization of Rift Valley fever virus by human monoclonal antibodies through fusion inhibition. Proc Natl Acad Sci U S A 2021; 118:e2025642118. [PMID: 33782133 PMCID: PMC8040655 DOI: 10.1073/pnas.2025642118] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rift Valley fever virus (RVFV), an emerging arboviral and zoonotic bunyavirus, causes severe disease in livestock and humans. Here, we report the isolation of a panel of monoclonal antibodies (mAbs) from the B cells of immune individuals following natural infection in Kenya or immunization with MP-12 vaccine. The B cell responses of individuals who were vaccinated or naturally infected recognized similar epitopes on both Gc and Gn proteins. The Gn-specific mAbs and two mAbs that do not recognize either monomeric Gc or Gn alone but recognized the hetero-oligomer glycoprotein complex (Gc+Gn) when Gc and Gn were coexpressed exhibited potent neutralizing activities in vitro, while Gc-specific mAbs exhibited relatively lower neutralizing capacity. The two Gc+Gn-specific mAbs and the Gn domain A-specific mAbs inhibited RVFV fusion to cells, suggesting that mAbs can inhibit the exposure of the fusion loop in Gc, a class II fusion protein, and thus prevent fusion by an indirect mechanism without direct fusion loop contact. Competition-binding analysis with coexpressed Gc/Gn and mutagenesis library screening indicated that these mAbs recognize four major antigenic sites, with two sites of vulnerability for neutralization on Gn. In experimental models of infection in mice, representative mAbs recognizing three of the antigenic sites reduced morbidity and mortality when used at a low dose in both prophylactic and therapeutic settings. This study identifies multiple candidate mAbs that may be suitable for use in humans against RVFV infection and highlights fusion inhibition against bunyaviruses as a potential contributor to potent antibody-mediated neutralization.
Collapse
Affiliation(s)
- Nathaniel S Chapman
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Haiyan Zhao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jonna B Westover
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322
| | - Birte Kalveram
- Department of Pathology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555
| | - Robin Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jessica Rodriguez
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Rachel Sutton
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Joseph Genualdi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - A Desiree LaBeaud
- Department of Pediatrics, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, CA 94305
| | - Francis M Mutuku
- Department of Environment and Health Sciences, Technical University of Mombasa, Mombasa, Kenya
| | - Phillip R Pittman
- Medical Research and Material Command, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702
| | - Alexander N Freiberg
- Department of Pathology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch at Galveston, Galveston, TX 77555
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch at Galveston, Galveston, TX 77555
| | - Brian B Gowen
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232;
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
7
|
Nielsen SS, Alvarez J, Bicout DJ, Calistri P, Depner K, Drewe JA, Garin-Bastuji B, Rojas JLG, Schmidt CG, Michel V, Chueca MÁM, Roberts HC, Sihvonen LH, Stahl K, Calvo AV, Viltrop A, Winckler C, Bett B, Cetre-Sossah C, Chevalier V, Devos C, Gubbins S, Monaco F, Sotiria-Eleni A, Broglia A, Abrahantes JC, Dhollander S, Stede YVD, Zancanaro G. Rift Valley Fever - epidemiological update and risk of introduction into Europe. EFSA J 2020; 18:e06041. [PMID: 33020705 PMCID: PMC7527653 DOI: 10.2903/j.efsa.2020.6041] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Rift Valley fever (RVF) is a vector-borne disease transmitted by a broad spectrum of mosquito species, especially Aedes and Culex genus, to animals (domestic and wild ruminants and camels) and humans. Rift Valley fever is endemic in sub-Saharan Africa and in the Arabian Peninsula, with periodic epidemics characterised by 5-15 years of inter-epizootic periods. In the last two decades, RVF was notified in new African regions (e.g. Sahel), RVF epidemics occurred more frequently and low-level enzootic virus circulation has been demonstrated in livestock in various areas. Recent outbreaks in a French overseas department and some seropositive cases detected in Turkey, Tunisia and Libya raised the attention of the EU for a possible incursion into neighbouring countries. The movement of live animals is the most important pathway for RVF spread from the African endemic areas to North Africa and the Middle East. The movement of infected animals and infected vectors when shipped by flights, containers or road transport is considered as other plausible pathways of introduction into Europe. The overall risk of introduction of RVF into EU through the movement of infected animals is very low in all the EU regions and in all MSs (less than one epidemic every 500 years), given the strict EU animal import policy. The same level of risk of introduction in all the EU regions was estimated also considering the movement of infected vectors, with the highest level for Belgium, Greece, Malta, the Netherlands (one epidemic every 228-700 years), mainly linked to the number of connections by air and sea transports with African RVF infected countries. Although the EU territory does not seem to be directly exposed to an imminent risk of RVFV introduction, the risk of further spread into countries neighbouring the EU and the risks of possible introduction of infected vectors, suggest that EU authorities need to strengthen their surveillance and response capacities, as well as the collaboration with North African and Middle Eastern countries.
Collapse
|
8
|
Characterization of Two Neutralizing Antibodies against Rift Valley Fever Virus Gn Protein. Viruses 2020; 12:v12030259. [PMID: 32120864 PMCID: PMC7150882 DOI: 10.3390/v12030259] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
The Rift Valley fever virus (RVFV) is an arthropod-borne virus that can not only cause severe disease in domestic animals but also in humans. However, the licensed vaccines or available therapeutics for humans do not exist. Here, we report two Gn-specific neutralizing antibodies (NAbs), isolated from a rhesus monkey immunized with recombinant human adenoviruses type 4 expressing Rift Valley fever virus Gn and Gc protein (rHAdV4-GnGcopt). The two NAbs were both able to protect host cells from RVFV infection. The interactions between NAbs and Gn were then characterized to demonstrate that these two NAbs might preclude RVFV glycoprotein rearrangement, hindering the exposure of fusion loops in Gc to endosomal membranes after the virus invades the host cell. The target region for the two NAbs is located in the Gn domain III, implying that Gn is a desired target for developing vaccines and neutralizing antibodies against RVFV.
Collapse
|
9
|
Boumart Z, Daouam S, Bamouh Z, Jazouli M, Tadlaoui KO, Dungu B, Bettinger G, Watts DM, Elharrak M. Safety and immunogenicity of a live attenuated Rift Valley Fever recombinant arMP-12ΔNSm21/384 vaccine candidate for sheep, goats and calves. Vaccine 2019; 37:1642-1650. [PMID: 30773401 DOI: 10.1016/j.vaccine.2019.01.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 01/03/2023]
Abstract
Rift Valley fever (RVF) causes serious health and economic losses to the livestock industry as well as a significant cause of human disease. The prevention of RVF in Africa is a global priority, however, available vaccines have only been partially effective. Therefore, the objective of this study was to evaluate the safety and immunogenicity of a live, attenuated recombinant RVFV arMP-12ΔNSm21/384 nucleotide deletion vaccine candidate in domestic ruminants. Evaluation involved testing to determine the infectivity titer of the vaccine virus in Vero cells for industrial scale up vaccine production. Safety experiments were conducted to determine the potential of the vaccine virus to revert to virulence by serial passages in sheep, the possibility of virus spread from vaccinated sheep and calves to unvaccinated animals, and the potential health effects of administering overdoses of the vaccine to sheep, goats and calves. The immunogenicity of 3 doses of 104, 105 and 106 Tissue Culture Infectious Doses50% (TCID50) of the vaccine was assessed in 3 groups of 10 sheep and 3 groups of 10 goats, and doses of 105, 106 and 107 TCID50 was evaluated in 3 groups of 10 calves subcutaenous vaccintation. The results showed that the infectivity titer of the vaccine virus was 108.4 TCID50/ml, that the vaccine did not spread from vaccinated to un-vaccinated animals, there was no evidence of reversion to virulence in sheep and the vaccine overdoses did not cause any adverse effects. The immunogenicity among sheep, goats and calves indicated that doses of 104-106 TCID50 elicited detectable antibody by day 7 post-vaccination (PV) with antibody titers ranging from 0.6 log to 2.1 log on day 14 PV with sustained titers through day 28 PV. Overall, these findings indicated that the RVFV arMP-12ΔNSm21/384 vaccine is a promising candidate for the prevention of RVF among domestic ruminants.
Collapse
Affiliation(s)
- Z Boumart
- Research and Development Dept., Multi-Chemical Industry, Lot. 157, Z I, Sud-Ouest (ERAC) B.P.: 278, Mohammedia 28810, Morocco.
| | - S Daouam
- Research and Development Dept., Multi-Chemical Industry, Lot. 157, Z I, Sud-Ouest (ERAC) B.P.: 278, Mohammedia 28810, Morocco.
| | - Z Bamouh
- Research and Development Dept., Multi-Chemical Industry, Lot. 157, Z I, Sud-Ouest (ERAC) B.P.: 278, Mohammedia 28810, Morocco.
| | - M Jazouli
- Research and Development Dept., Multi-Chemical Industry, Lot. 157, Z I, Sud-Ouest (ERAC) B.P.: 278, Mohammedia 28810, Morocco.
| | - K O Tadlaoui
- Research and Development Dept., Multi-Chemical Industry, Lot. 157, Z I, Sud-Ouest (ERAC) B.P.: 278, Mohammedia 28810, Morocco.
| | - B Dungu
- Research and Development Dept., Multi-Chemical Industry, Lot. 157, Z I, Sud-Ouest (ERAC) B.P.: 278, Mohammedia 28810, Morocco.
| | - G Bettinger
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - D M Watts
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - M Elharrak
- Research and Development Dept., Multi-Chemical Industry, Lot. 157, Z I, Sud-Ouest (ERAC) B.P.: 278, Mohammedia 28810, Morocco.
| |
Collapse
|
10
|
Nyundo S, Adamson E, Rowland J, Palermo PM, Matiko M, Bettinger GE, Wambura P, Morrill JC, Watts DM. Safety and immunogenicity of Rift Valley fever MP-12 and arMP-12ΔNSm21/384 vaccine candidates in goats (Capra aegagrus hircus) from Tanzania. ACTA ACUST UNITED AC 2019; 86:e1-e8. [PMID: 30843406 PMCID: PMC6407455 DOI: 10.4102/ojvr.v86i1.1683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022]
Abstract
Vaccination of domestic ruminants is considered to be an effective strategy for protecting these animals against Rift Valley fever (RVF), but available vaccines have limitations. Therefore, the aim of this study was to determine the safety and immunogenicity of RVF virus (RVFV) mutagenesis passage 12 (MP-12) and arMP-12ΔNSm21/384 vaccine candidates in goats (Capra aegagrus hircus) in Tanzania. Goats were vaccinated intramuscularly with RVFV MP-12 or arMP-12ΔNSm21/384, and then on Day 87 post-vaccination (PV) all animals were revaccinated using the RVFV MP-12 vaccine candidate. Serum samples were collected from the animals before and after vaccination at various intervals to test for RVFV using a Vero cell culture assay and reverse transcription polymerase chain reaction and for RVFV-neutralising antibody using a plaque reduction neutralisation assay. Serum samples collected before vaccination on Days -14 and 0, and on Days 3, 4 and 5 PV were negative for RVFV and neutralising antibody. All animals remained healthy, and viremia was not detected in any of the animals. Rift Valley fever virus antibody was first detected on Day 5 PV at a 1:10 dilution in five of five animals vaccinated with the MP-12 vaccine and in five of eight animals vaccinated with arMP-12ΔNSm21/384. Titres then increased and were sustained at 1:40 to 1:640 through to Day 87 PV. All animals that were revaccinated on Day 87 PV with MP-12 developed antibody titres ranging from 1:160 to as high as 1:10 240 on Days 14 and 21 PV. Although the antibody titres for goats vaccinated with RVF MP-12 were slightly higher than titres elicited by the arMP-12ΔNSm21/384 vaccine, these findings demonstrated that both vaccines are promising candidates for the prevention of RVF among Tansanian goats.
Collapse
Affiliation(s)
- Salama Nyundo
- Department of Microbiology, Parasitology and Biotechnology, Sokoine University of Agriculture.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Wonderlich ER, Caroline AL, McMillen CM, Walters AW, Reed DS, Barratt-Boyes SM, Hartman AL. Peripheral Blood Biomarkers of Disease Outcome in a Monkey Model of Rift Valley Fever Encephalitis. J Virol 2018; 92:e01662-17. [PMID: 29118127 PMCID: PMC5774883 DOI: 10.1128/jvi.01662-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022] Open
Abstract
Rift Valley Fever (RVF) is an emerging arboviral disease of livestock and humans. Although the disease is caused by a mosquito-borne virus, humans are infected through contact with, or inhalation of, virus-laden particles from contaminated animal carcasses. Some individuals infected with RVF virus (RVFV) develop meningoencephalitis, resulting in morbidity and mortality. Little is known about the pathogenic mechanisms that lead to neurologic sequelae, and thus, animal models that represent human disease are needed. African green monkeys (AGM) exposed to aerosols containing RVFV develop a reproducibly lethal neurological disease that resembles human illness. To understand the disease process and identify biomarkers of lethality, two groups of 5 AGM were infected by inhalation with either a lethal or a sublethal dose of RVFV. Divergence between lethal and sublethal infections occurred as early as 2 days postinfection (dpi), at which point CD8+ T cells from lethally infected AGM expressed activated caspase-3 and simultaneously failed to increase levels of major histocompatibility complex (MHC) class II molecules, in contrast to surviving animals. At 4 dpi, lethally infected animals failed to demonstrate proliferation of total CD4+ and CD8+ T cells, in contrast to survivors. These marked changes in peripheral blood cells occur much earlier than more-established indicators of severe RVF disease, such as granulocytosis and fever. In addition, an early proinflammatory (gamma interferon [IFN-γ], interleukin 6 [IL-6], IL-8, monocyte chemoattractant protein 1 [MCP-1]) and antiviral (IFN-α) response was seen in survivors, while very late cytokine expression was found in animals with lethal infections. By characterizing immunological markers of lethal disease, this study furthers our understanding of RVF pathogenesis and will allow the testing of therapeutics and vaccines in the AGM model.IMPORTANCE Rift Valley Fever (RVF) is an important emerging viral disease for which we lack both an effective human vaccine and treatment. Encephalitis and neurological disease resulting from RVF lead to death or significant long-term disability for infected people. African green monkeys (AGM) develop lethal neurological disease when infected with RVF virus by inhalation. Here we report the similarities in disease course between infected AGM and humans. For the first time, we examine the peripheral immune response during the course of infection in AGM and show that there are very early differences in the immune response between animals that survive infection and those that succumb. We conclude that AGM are a novel and suitable monkey model for studying the neuropathogenesis of RVF and for testing vaccines and therapeutics against this emerging viral pathogen.
Collapse
Affiliation(s)
- Elizabeth R Wonderlich
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy L Caroline
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cynthia M McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aaron W Walters
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Douglas S Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Simon M Barratt-Boyes
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy L Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Current Status of Rift Valley Fever Vaccine Development. Vaccines (Basel) 2017; 5:vaccines5030029. [PMID: 28925970 PMCID: PMC5620560 DOI: 10.3390/vaccines5030029] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/16/2017] [Accepted: 09/18/2017] [Indexed: 01/08/2023] Open
Abstract
Rift Valley Fever (RVF) is a mosquito-borne zoonotic disease that presents a substantial threat to human and public health. It is caused by Rift Valley fever phlebovirus (RVFV), which belongs to the genus Phlebovirus and the family Phenuiviridae within the order Bunyavirales. The wide distribution of competent vectors in non-endemic areas coupled with global climate change poses a significant threat of the transboundary spread of RVFV. In the last decade, an improved understanding of the molecular biology of RVFV has facilitated significant progress in the development of novel vaccines, including DIVA (differentiating infected from vaccinated animals) vaccines. Despite these advances, there is no fully licensed vaccine for veterinary or human use available in non-endemic countries, whereas in endemic countries, there is no clear policy or practice of routine/strategic livestock vaccinations as a preventive or mitigating strategy against potential RVF disease outbreaks. The purpose of this review was to provide an update on the status of RVF vaccine development and provide perspectives on the best strategies for disease control. Herein, we argue that the routine or strategic vaccination of livestock could be the best control approach for preventing the outbreak and spread of future disease.
Collapse
|
13
|
Ikegami T. Rift Valley fever vaccines: an overview of the safety and efficacy of the live-attenuated MP-12 vaccine candidate. Expert Rev Vaccines 2017; 16:601-611. [PMID: 28425834 DOI: 10.1080/14760584.2017.1321482] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Rift Valley fever (RVF) is a mosquito-borne zoonotic viral disease endemic to Africa and the Arabian Peninsula. High rates of abortion among infected ruminants and hemorrhagic fever in infected humans are major public health concerns. Commercially available veterinary RVF vaccines are important for preventing the spread of the Rift Valley fever virus (RVFV) in endemic countries; however, RVFV outbreaks continue to occur frequently in endemic countries in the 21st century. In the U.S., the live-attenuated MP-12 vaccine has been developed for both animal and human vaccination. This vaccine strain is well attenuated, and a single dose induces neutralizing antibodies in both ruminants and humans. Areas covered: This review describes scientific evidences of MP-12 vaccine efficacy and safety, as well as MP-12 variants recently developed by reverse genetics, in comparison with other RVF vaccines. Expert commentary: The containment of active RVF outbreaks and long-term protection from RVF exposure to infected mosquitoes are important goals for RVF vaccination. MP-12 vaccine will allow immediate vaccination of susceptible animals in case of an unexpected RVF outbreak in the U.S., whereas MP-12 vaccine may be also useful for the RVF control in endemic regions.
Collapse
Affiliation(s)
- Tetsuro Ikegami
- a Department of Pathology, Sealy Center for Vaccine Development, Center for Biodefense and Emerging Infectious Diseases , The University of Texas Medical Branch , Galveston , TX , USA
| |
Collapse
|
14
|
The L, M, and S Segments of Rift Valley Fever Virus MP-12 Vaccine Independently Contribute to a Temperature-Sensitive Phenotype. J Virol 2016; 90:3735-44. [PMID: 26819307 DOI: 10.1128/jvi.02241-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/14/2016] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Rift Valley fever (RVF) is endemic to Africa, and the mosquito-borne disease is characterized by "abortion storms" in ruminants and by hemorrhagic fever, encephalitis, and blindness in humans. Rift Valley fever virus (RVFV; family Bunyaviridae, genus Phlebovirus) has a tripartite negative-stranded RNA genome (L, M, and S segments). A live-attenuated vaccine for RVF, the MP-12 vaccine, is conditionally licensed for veterinary use in the United States. MP-12 is fully attenuated by the combination of the partially attenuated L, M, and S segments. Temperature sensitivity (ts) limits viral replication at a restrictive temperature and may be involved with viral attenuation. In this study, we aimed to characterize the ts mutations for MP-12. The MP-12 vaccine showed restricted replication at 38°C and replication shutoff (100-fold or greater reduction in virus titer compared to that at 37°C) at 39°C in Vero and MRC-5 cells. Using rZH501 reassortants with either the MP-12 L, M, or S segment, we found that all three segments encode a temperature-sensitive phenotype. However, the ts phenotype of the S segment was weaker than that of the M or L segment. We identified Gn-Y259H, Gc-R1182G, L-V172A, and L-M1244I as major ts mutations for MP-12. The ts mutations in the L segment decreased viral RNA synthesis, while those in the M segment delayed progeny production from infected cells. We also found that a lack of NSs and/or 78kD/NSm protein expression minimally affected the ts phenotype. Our study revealed that MP-12 is a unique vaccine carrying ts mutations in the L, M, and S segments. IMPORTANCE Rift Valley fever (RVF) is a mosquito-borne viral disease endemic to Africa, characterized by high rates of abortion in ruminants and severe diseases in humans. Vaccination is important to prevent the spread of disease, and a live-attenuated MP-12 vaccine is currently the only vaccine with a conditional license in the United States. This study determined the temperature sensitivity (ts) of MP-12 vaccine to understand virologic characteristics. Our study revealed that MP-12 vaccine contains ts mutations independently in the L, M, and S segments and that MP-12 displays a restrictive replication at 38°C.
Collapse
|
15
|
Laughlin RC, Drake KL, Morrill JC, Adams LG. Correlative Gene Expression to Protective Seroconversion in Rift Valley Fever Vaccinates. PLoS One 2016; 11:e0147027. [PMID: 26783758 PMCID: PMC4718665 DOI: 10.1371/journal.pone.0147027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022] Open
Abstract
Rift Valley fever Virus (RVFV), a negative-stranded RNA virus, is the etiological agent of the vector-borne zoonotic disease, Rift Valley fever (RVF). In both humans and livestock, protective immunity can be achieved through vaccination. Earlier and more recent vaccine trials in cattle and sheep demonstrated a strong neutralizing antibody and total IgG response induced by the RVF vaccine, authentic recombinant MP-12 (arMP-12). From previous work, protective immunity in sheep and cattle vaccinates normally occurs from 7 to 21 days after inoculation with arMP-12. While the serology and protective response induced by arMP-12 has been studied, little attention has been paid to the underlying molecular and genetic events occurring prior to the serologic immune response. To address this, we isolated RNA from whole blood of vaccinated calves over a time course of 21 days before and after vaccination with arMP-12. The time course RNAs were sequenced by RNASeq and bioinformatically analyzed. Our results revealed time-dependent activation or repression of numerous gene ontologies and pathways related to the vaccine induced immune response and its regulation. Additional bioinformatic analyses identified a correlative relationship between specific host immune response genes and protective immunity prior to the detection of protective serum neutralizing antibody responses. These results contribute an important proof of concept for identifying molecular and genetic components underlying the immune response to RVF vaccination and protection prior to serologic detection.
Collapse
Affiliation(s)
- Richard C. Laughlin
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, United States of America
| | - Kenneth L. Drake
- Seralogix LLC, 335 Bee Cave Rd, Suite 607, Austin, TX 78746, United States of America
| | - John C. Morrill
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, United States of America
| | - L. Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, United States of America
- * E-mail:
| |
Collapse
|
16
|
Ly HJ, Lokugamage N, Ikegami T. Application of Droplet Digital PCR to Validate Rift Valley Fever Vaccines. Methods Mol Biol 2016; 1403:207-20. [PMID: 27076132 DOI: 10.1007/978-1-4939-3387-7_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Droplet Digital™ polymerase chain reaction (ddPCR™) is a promising technique that quantitates the absolute concentration of nucleic acids in a given sample. This technique utilizes water-in-oil emulsion technology, a system developed by Bio-Rad Laboratories that partitions a single sample into thousands of nanoliter-sized droplets and counts nucleic acid molecules encapsulated in each individual particle as one PCR reaction. This chapter discusses the applications and methodologies of ddPCR for development of Rift Valley fever (RVF) vaccine, using an example that measures RNA copy numbers of a live-attenuated MP-12 vaccine from virus stocks, infected cells, or animal blood. We also discuss how ddPCR detects a reversion mutant of MP-12 from virus stocks accurately. The use of ddPCR improves the quality control of live-attenuated vaccines in the seed lot systems.
Collapse
Affiliation(s)
- Hoai J Ly
- Department of Pathology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA
| | - Nandadeva Lokugamage
- Department of Pathology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA
| | - Tetsuro Ikegami
- Department of Pathology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
17
|
Rift Valley fever MP-12 vaccine Phase 2 clinical trial: Safety, immunogenicity, and genetic characterization of virus isolates. Vaccine 2015; 34:523-530. [PMID: 26706271 DOI: 10.1016/j.vaccine.2015.11.078] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 11/23/2022]
Abstract
An outbreak or deliberate release of Rift Valley fever (RVF) virus could have serious public health and socioeconomic consequences. A safe RVF vaccine capable of eliciting long-lasting immunity after a single injection is urgently needed. The live attenuated RVF MP-12 vaccine candidate has shown promise in Phase 1 clinical trials; no evidence of reversion to virulence has been identified in numerous animal studies. The objective of this Phase 2 clinical trial was to (a) further examine the safety and immunogenicity of RVF MP-12 in RVF virus-naïve humans and (b) characterize isolates of RVF MP-12 virus recovered from the blood of vaccinated subjects to evaluate the genetic stability of MP-12 attenuation. We found that RVF MP-12 was well tolerated, causing mostly mild reactions that resolved without sequelae. Of 19 subjects, 18 (95%) and 19 (100%) achieved, respectively, 80% and 50% plaque reduction neutralization titers (PRNT80 and PRNT50)≥1:20 by postvaccination day 28. All 18 PRNT80 responders maintained PRNT80 and PRNT50≥1:40 until at least postvaccination month 12. Viremia was undetectable in the plasma of any subject by direct plaque assay techniques. However, 5 of 19 vaccinees were positive for MP-12 isolates in plasma by blind passage of plasma on Vero cells. Vaccine virus was also recovered from buffy coat material from one of those vaccinees and from one additional vaccinee. Through RNA sequencing of MP-12 isolates, we found no reversions of amino acids to those of the parent virulent virus (strain ZH548). Five years after a single dose of RVF MP-12 vaccine, 8 of 9 vaccinees (89%) maintained a PRNT80≥1:20. These findings support the continued development of RVF MP-12 as a countermeasure against RVF virus in humans.
Collapse
|
18
|
Nishiyama S, Ikegami T. Temperature-sensitive mutations for live-attenuated Rift Valley fever vaccines: implications from other RNA viruses. Front Microbiol 2015; 6:787. [PMID: 26322023 PMCID: PMC4531298 DOI: 10.3389/fmicb.2015.00787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
Rift Valley fever (RVF) is a mosquito-borne zoonotic disease endemic to the African continent. RVF is characterized by high rate of abortions in ruminants and hemorrhagic fever, encephalitis, or blindness in humans. RVF is caused by the Rift Valley fever virus (RVFV: genus Phlebovirus, family Bunyaviridae). Vaccination is the only known effective strategy to prevent the disease, but there are no licensed RVF vaccines available for humans. A live-attenuated vaccine candidate derived from the wild-type pathogenic Egyptian ZH548 strain, MP-12, has been conditionally licensed for veterinary use in the U.S. MP-12 displays a temperature-sensitive (ts) phenotype and does not replicate at 41°C. The ts mutation limits viral replication at a specific body temperature and may lead to an attenuation of the virus. Here we will review well-characterized ts mutations for RNA viruses, and further discuss the potential in designing novel live-attenuated vaccines for RVF.
Collapse
Affiliation(s)
- Shoko Nishiyama
- Department of Pathology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| | - Tetsuro Ikegami
- Department of Pathology, The University of Texas Medical Branch at Galveston, Galveston, TX USA ; Sealy Center for Vaccine Development, The University of Texas Medical Branch at Galveston, Galveston, TX USA ; Center for Biodefense and Emerging Infectious Diseases, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| |
Collapse
|
19
|
Evaluation of the Efficacy, Potential for Vector Transmission, and Duration of Immunity of MP-12, an Attenuated Rift Valley Fever Virus Vaccine Candidate, in Sheep. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:930-7. [PMID: 26041042 DOI: 10.1128/cvi.00114-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/30/2015] [Indexed: 11/20/2022]
Abstract
Rift Valley fever virus (RVFV) causes serious disease in ruminants and humans in Africa. In North America, there are susceptible ruminant hosts and competent mosquito vectors, yet there are no fully licensed animal vaccines for this arthropod-borne virus, should it be introduced. Studies in sheep and cattle have found the attenuated strain of RVFV, MP-12, to be both safe and efficacious based on early testing, and a 2-year conditional license for use in U.S. livestock has been issued. The purpose of this study was to further determine the vaccine's potential to infect mosquitoes, the duration of humoral immunity to 24 months postvaccination, and the ability to prevent disease and viremia from a virulent challenge. Vaccination experiments conducted in sheep found no evidence of a potential for vector transmission to 4 North American mosquito species. Neutralizing antibodies were elicited, with titers of >1:40 still present at 24 months postvaccination. Vaccinates were protected from clinical signs and detectable viremia after challenge with virulent virus, while control sheep had fever and high-titered viremia extending for 5 days. Antibodies to three viral proteins (nucleocapsid N, the N-terminal half of glycoprotein GN, and the nonstructural protein from the short segment NSs) were also detected to 24 months using competitive enzyme-linked immunosorbent assays. This study demonstrates that the MP-12 vaccine given as a single dose in sheep generates protective immunity to a virulent challenge with antibody duration of at least 2 years, with no evidence of a risk for vector transmission.
Collapse
|
20
|
Rift Valley Fever Virus MP-12 Vaccine Is Fully Attenuated by a Combination of Partial Attenuations in the S, M, and L Segments. J Virol 2015; 89:7262-76. [PMID: 25948740 DOI: 10.1128/jvi.00135-15] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/28/2015] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Rift Valley fever (RVF) is a mosquito-borne zoonotic disease endemic to Africa and characterized by a high rate of abortion in ruminants and hemorrhagic fever, encephalitis, or blindness in humans. RVF is caused by Rift Valley fever virus (RVFV; family Bunyaviridae, genus Phlebovirus), which has a tripartite negative-stranded RNA genome (consisting of the S, M, and L segments). Further spread of RVF into countries where the disease is not endemic may affect the economy and public health, and vaccination is an effective approach to prevent the spread of RVFV. A live-attenuated MP-12 vaccine is one of the best-characterized RVF vaccines for safety and efficacy and is currently conditionally licensed for use for veterinary purposes in the United States. Meanwhile, as of 2015, no other RVF vaccine has been conditionally or fully licensed for use in the United States. The MP-12 strain is derived from wild-type pathogenic strain ZH548, and its genome encodes 23 mutations in the three genome segments. However, the mechanism of MP-12 attenuation remains unknown. We characterized the attenuation of wild-type pathogenic strain ZH501 carrying a mutation(s) of the MP-12 S, M, or L segment in a mouse model. Our results indicated that MP-12 is attenuated by the mutations in the S, M, and L segments, while the mutations in the M and L segments confer stronger attenuation than those in the S segment. We identified a combination of 3 amino acid changes, Y259H (Gn), R1182G (Gc), and R1029K (L), that was sufficient to attenuate ZH501. However, strain MP-12 with reversion mutations at those 3 sites was still highly attenuated. Our results indicate that MP-12 attenuation is supported by a combination of multiple partial attenuation mutations and a single reversion mutation is less likely to cause a reversion to virulence of the MP-12 vaccine. IMPORTANCE Rift Valley fever (RVF) is a mosquito-transmitted viral disease that is endemic to Africa and that has the potential to spread into other countries. Vaccination is considered an effective way to prevent the disease, and the only available veterinary RVF vaccine in the United States is a live-attenuated MP-12 vaccine, which is conditionally licensed. Strain MP-12 is different from its parental pathogenic RVFV strain, strain ZH548, because of the presence of 23 mutations. This study determined the role of individual mutations in the attenuation of the MP-12 strain. We found that full attenuation of MP-12 occurs by a combination of multiple mutations. Our findings indicate that a single reversion mutation will less likely cause a major reversion to virulence of the MP-12 vaccine.
Collapse
|
21
|
Njenga MK, Njagi L, Thumbi SM, Kahariri S, Githinji J, Omondi E, Baden A, Murithi M, Paweska J, Ithondeka PM, Ngeiywa KJ, Dungu B, Donadeu M, Munyua PM. Randomized controlled field trial to assess the immunogenicity and safety of rift valley fever clone 13 vaccine in livestock. PLoS Negl Trop Dis 2015; 9:e0003550. [PMID: 25756501 PMCID: PMC4355591 DOI: 10.1371/journal.pntd.0003550] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 01/20/2015] [Indexed: 11/23/2022] Open
Abstract
Background Although livestock vaccination is effective in preventing Rift Valley fever (RVF) epidemics, there are concerns about safety and effectiveness of the only commercially available RVF Smithburn vaccine. We conducted a randomized controlled field trial to evaluate the immunogenicity and safety of the new RVF Clone 13 vaccine, recently registered in South Africa. Methods In a blinded randomized controlled field trial, 404 animals (85 cattle, 168 sheep, and 151 goats) in three farms in Kenya were divided into three groups. Group A included males and non-pregnant females that were randomized and assigned to two groups; one vaccinated with RVF Clone 13 and the other given placebo. Groups B included animals in 1st half of pregnancy, and group C animals in 2nd half of pregnancy, which were also randomized and either vaccinated and given placebo. Animals were monitored for one year and virus antibodies titers assessed on days 14, 28, 56, 183 and 365. Results In vaccinated goats (N = 72), 72% developed anti-RVF virus IgM antibodies and 97% neutralizing IgG antibodies. In vaccinated sheep (N = 77), 84% developed IgM and 91% neutralizing IgG antibodies. Vaccinated cattle (N = 42) did not develop IgM antibodies but 67% developed neutralizing IgG antibodies. At day 14 post-vaccination, the odds of being seropositive for IgG in the vaccine group was 3.6 (95% CI, 1.5 – 9.2) in cattle, 90.0 (95% CI, 25.1 – 579.2) in goats, and 40.0 (95% CI, 16.5 – 110.5) in sheep. Abortion was observed in one vaccinated goat but histopathologic analysis did not indicate RVF virus infection. There was no evidence of teratogenicity in vaccinated or placebo animals. Conclusions The results suggest RVF Clone 13 vaccine is safe to use and has high (>90%) immunogenicity in sheep and goats but moderate (> 65%) immunogenicity in cattle. Although livestock vaccination is effective in preventing Rift Valley fever (RVF) outbreaks, there are concerns about safety and effectiveness of the only commercially available vaccine for the disease. Here, we conducted a field trial in Kenya to evaluate the safety and ability to induce protection for a new RVF vaccine, referred to as Clone 13, that was recently registered in South Africa. A total of 404 animals, consisting of cattle, sheep, and goats, were divided two groups and one group was vaccinated with Clone 13 vaccine while the other group was not vaccinated. The animals were followed for one year and analyzed for RVF antibody levels at days 14, 28, 56, 183, and 365 after vaccination. Between 91% and 97% of vaccinated sheep and goats develop antibodies to the vaccine, whereas only 67% of the vaccinated cattle developed antibodies. These finding indicate that the Clone 13 vaccine induces high levels of protective antibodies in sheep and goats and moderate levels in cattle. The vaccine was safe since none of vaccinated animals developed evidence of RVF disease including deformities in newborns, and only 1 out of 120 pregnant animals had an abortion that was not associated with the RVF disease.
Collapse
Affiliation(s)
- M. Kariuki Njenga
- Division of Global Health Protection, United States Centers for Disease Control and Prevention-Kenya, Nairobi, Kenya
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, United States of America
- * E-mail: ,
| | - Leonard Njagi
- Kenya Ministry of Agriculture Livestock and Fisheries, Nairobi, Kenya
| | - S. Mwangi Thumbi
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, United States of America
| | - Samuel Kahariri
- Kenya Ministry of Agriculture Livestock and Fisheries, Nairobi, Kenya
| | - Jane Githinji
- Kenya Ministry of Agriculture Livestock and Fisheries, Nairobi, Kenya
| | - Eunice Omondi
- Kenya Ministry of Agriculture Livestock and Fisheries, Nairobi, Kenya
| | - Amy Baden
- Division of Global Health Protection, United States Centers for Disease Control and Prevention-Kenya, Nairobi, Kenya
| | - Mbabu Murithi
- Kenya Ministry of Agriculture Livestock and Fisheries, Nairobi, Kenya
| | - Janusz Paweska
- National Institute for Communicable Diseases, Pretoria, South Africa
| | | | - Kisa J. Ngeiywa
- Kenya Ministry of Agriculture Livestock and Fisheries, Nairobi, Kenya
| | - Baptiste Dungu
- Global Alliance for Livestock Veterinary Medicines, Edinburg, Scotland, United Kingdom
| | - Meritxell Donadeu
- Global Alliance for Livestock Veterinary Medicines, Edinburg, Scotland, United Kingdom
| | - Peninah M. Munyua
- Division of Global Health Protection, United States Centers for Disease Control and Prevention-Kenya, Nairobi, Kenya
| |
Collapse
|
22
|
Sumaye RD, Abatih EN, Thiry E, Amuri M, Berkvens D, Geubbels E. Inter-epidemic acquisition of Rift Valley fever virus in humans in Tanzania. PLoS Negl Trop Dis 2015; 9:e0003536. [PMID: 25723502 PMCID: PMC4344197 DOI: 10.1371/journal.pntd.0003536] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/13/2015] [Indexed: 11/26/2022] Open
Abstract
Background In East Africa, epidemics of Rift Valley fever (RVF) occur in cycles of 5–15 years following unusually high rainfall. RVF transmission during inter-epidemic periods (IEP) generally passes undetected in absence of surveillance in mammalian hosts and vectors. We studied IEP transmission of RVF and evaluated the demographic, behavioural, occupational and spatial determinants of past RVF infection. Methodology Between March and August 2012 we collected blood samples, and administered a risk factor questionnaire among 606 inhabitants of 6 villages in the seasonally inundated Kilombero Valley, Tanzania. ELISA tests were used to detect RVFV IgM and IgG antibodies in serum samples. Risk factors were examined by mixed effects logistic regression. Findings RVF virus IgM antibodies, indicating recent RVFV acquisition, were detected in 16 participants, representing 2.6% overall and in 22.5% of inhibition ELISA positives (n = 71). Four of 16 (25.0%) IgM positives and 11/71 (15.5%) of individuals with inhibition ELISA sero-positivity reported they had had no previous contact with host animals. Sero-positivity on inhibition ELISA was 11.7% (95% CI 9.2–14.5) and risk was elevated with age (odds ratio (OR) 1.03 per year; 95% CI 1.01–1.04), among milkers (OR 2.19; 95% CI 1.23–3.91), and individuals eating raw meat (OR 4.17; 95% CI 1.18–14.66). Households keeping livestock had a higher probability of having members with evidence of past infection (OR = 3.04, 95% CI = 1.42–6.48) than those that do not keep livestock. Conclusion There is inter-epidemic acquisition of RVFV in Kilombero Valley inhabitants. In the wake of declining malaria incidence, these findings underscore the need for clinicians to consider RVF in the differential diagnosis for febrile illnesses. Several types of direct contact with livestock are important risk factors for past infection with RVFV in this study’s population. However, at least part of RVFV transmission appears to have occurred through bites of infected mosquitoes. Rift Valley fever (RVF) is a disease of animals and people that is caused by the RVF virus. During epidemics, humans get RVF through direct contact with animals or through mosquito bites. In East Africa, epidemics occur every 5–15 years following unusually high rainfall. In between epidemics, the transmission of RVF might occur at low level. In an epidemic-free period, we measured whether people in the Kilombero Valley in Tanzania had evidence of past and recent RVF infection in their blood sample, and studied risk factors. Three per cent of people had been infected recently, and 12% had evidence of past infection, with increased risk with age, among milkers and among people eating raw meat. Some people with past or recent infection reported they had not had contact with animals. Households keeping livestock had more members with evidence of past infection. The findings show that people get infected with RVF in between epidemics, and that various types of contact with livestock are important risk factors. There is also evidence that some people get infected with RVFV by mosquitoes in the epidemic free period. Clinicians in the Kilombero Valley should consider RVF in the differential diagnosis of patients with fever.
Collapse
Affiliation(s)
- Robert David Sumaye
- Ifakara Health Institute, Ifakara, Tanzania
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
- * E-mail:
| | - Emmanuel Nji Abatih
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Etienne Thiry
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | | | - Dirk Berkvens
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | | |
Collapse
|
23
|
Lorenzo G, López-Gil E, Warimwe GM, Brun A. Understanding Rift Valley fever: contributions of animal models to disease characterization and control. Mol Immunol 2015; 66:78-88. [PMID: 25725948 DOI: 10.1016/j.molimm.2015.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 12/26/2014] [Accepted: 02/03/2015] [Indexed: 11/30/2022]
Abstract
Rift Valley fever (RVF) is a mosquito-borne viral zoonosis with devastating health impacts in domestic ruminants and humans. Effective vaccines and accurate disease diagnostic tools are key components in the control of RVF. Animal models reproducing infection with RVF virus are of upmost importance in the development of these disease control tools. Rodent infection models are currently used in the initial steps of vaccine development and for the study of virus induced pathology. Translation of data obtained in these animal models to target species (ruminants and humans) is highly desirable but does not always occur. Small ruminants and non-human primates have been used for pathogenesis and transmission studies, and for testing the efficacy of vaccines and therapeutic antiviral compounds. However, the molecular mechanisms of the immune response elicited by RVF virus infection or vaccination are still poorly understood. The paucity of data in this area offers opportunities for new research activities and programs. This review summarizes our current understanding with respect to immunity and pathogenesis of RVF in animal models with a particular emphasis on small ruminants and non-human primates, including recent experimental infection data in sheep.
Collapse
Affiliation(s)
- Gema Lorenzo
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación Agraria y Alimentaria (INIA-CISA), Valdeolmos, Madrid, Spain
| | - Elena López-Gil
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación Agraria y Alimentaria (INIA-CISA), Valdeolmos, Madrid, Spain
| | - George M Warimwe
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alejandro Brun
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación Agraria y Alimentaria (INIA-CISA), Valdeolmos, Madrid, Spain.
| |
Collapse
|
24
|
Oreshkova N, Cornelissen LAHM, de Haan CAM, Moormann RJM, Kortekaas J. Evaluation of nonspreading Rift Valley fever virus as a vaccine vector using influenza virus hemagglutinin as a model antigen. Vaccine 2014; 32:5323-9. [PMID: 25066737 DOI: 10.1016/j.vaccine.2014.07.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/02/2014] [Accepted: 07/15/2014] [Indexed: 11/30/2022]
Abstract
Virus replicon particles are capable of infection, genome replication and gene expression, but are unable to produce progeny virions, rendering their use inherently safe. By virtue of this unique combination of features, replicon particles hold great promise for vaccine applications. We previously developed replicon particles of Rift Valley fever virus (RVFV) and demonstrated their high efficacy as a RVFV vaccine in the natural target species. We have now investigated the feasibility of using this nonspreading RVFV (NSR) as a vaccine vector using influenza virus hemagglutinin as a model antigen. NSR particles were designed to express either the full-length hemagglutinin of influenza A virus H1N1 (NSR-HA) or the respective soluble ectodomain (NSR-sHA). The efficacies of the two NSR vector vaccines, applied via either the intramuscular or the intranasal route, were evaluated. A single vaccination with NSR-HA protected all mice from a lethal challenge dose, while vaccination with NSR-sHA was not protective. Interestingly, whereas intramuscular vaccination elicited superior systemic immune responses, intranasal vaccination provided optimal clinical protection.
Collapse
Affiliation(s)
- N Oreshkova
- Department of Virology, Central Veterinary Institute of Wageningen University and Research Centre, Lelystad, The Netherlands; Department of Infectious Diseases & Immunology, Virology Division, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - L A H M Cornelissen
- Department of Virology, Central Veterinary Institute of Wageningen University and Research Centre, Lelystad, The Netherlands.
| | - C A M de Haan
- Department of Infectious Diseases & Immunology, Virology Division, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - R J M Moormann
- Department of Virology, Central Veterinary Institute of Wageningen University and Research Centre, Lelystad, The Netherlands; Department of Infectious Diseases & Immunology, Virology Division, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - J Kortekaas
- Department of Virology, Central Veterinary Institute of Wageningen University and Research Centre, Lelystad, The Netherlands.
| |
Collapse
|
25
|
Kortekaas J. One Health approach to Rift Valley fever vaccine development. Antiviral Res 2014; 106:24-32. [DOI: 10.1016/j.antiviral.2014.03.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/10/2014] [Accepted: 03/15/2014] [Indexed: 10/25/2022]
|
26
|
Weingartl HM, Nfon CK, Zhang S, Marszal P, Wilson WC, Morrill J, Bettinger GE, Peters CJ. Efficacy of a recombinant Rift Valley fever virus MP-12 with NSm deletion as a vaccine candidate in sheep. Vaccine 2014; 32:2345-9. [DOI: 10.1016/j.vaccine.2013.12.064] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/09/2013] [Accepted: 12/18/2013] [Indexed: 11/26/2022]
|
27
|
Powell DS, Walker RC, Heflin DT, Fisher D, Kosky JB, Homer LC, Reed DS, Stefano-Cole K, Trichel AM, Hartman AL. Development of novel mechanisms for housing, handling, and remote monitoring of common marmosets at animal biosafety level 3. Pathog Dis 2014; 71:219-26. [PMID: 24453160 DOI: 10.1111/2049-632x.12140] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/13/2013] [Accepted: 01/08/2014] [Indexed: 11/29/2022] Open
Abstract
The use of common marmosets as an alternative non-human primate model for infectious disease research using BSL-3 viruses such as Rift Valley fever virus (RVFV) presents unique challenges with respect to housing, handling, and safety. Subject matter experts from veterinary care, animal husbandry, biosafety, engineering, and research were consulted to design a pilot experiment using marmosets infected with RVFV. This paper reviews the caging, handling, and safety-related adaptations and modifications that were required to humanely utilize marmosets as a model for high-hazard BSL-3 viral diseases.
Collapse
Affiliation(s)
- Diana S Powell
- Regional Biocontainment Laboratory, Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hartman AL, Powell DS, Bethel LM, Caroline AL, Schmid RJ, Oury T, Reed DS. Aerosolized rift valley fever virus causes fatal encephalitis in african green monkeys and common marmosets. J Virol 2014; 88:2235-45. [PMID: 24335307 PMCID: PMC3911574 DOI: 10.1128/jvi.02341-13] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/04/2013] [Indexed: 01/25/2023] Open
Abstract
Rift Valley fever (RVF) is a veterinary and human disease in Africa and the Middle East. The causative agent, RVF virus (RVFV), can be naturally transmitted by mosquito, direct contact, or aerosol. We sought to develop a nonhuman primate (NHP) model of severe RVF in humans to better understand the pathogenesis of RVF and to use for evaluation of medical countermeasures. NHP from four different species were exposed to aerosols containing RVFV. Both cynomolgus and rhesus macaques developed mild fevers after inhalation of RVFV, but no other clinical signs were noted and no macaque succumbed to RVFV infection. In contrast, both marmosets and African green monkeys (AGM) proved susceptible to aerosolized RVF virus. Fever onset was earlier with the marmosets and had a biphasic pattern similar to what has been reported in humans. Beginning around day 8 to day 10 postexposure, clinical signs consistent with encephalitis were noted in both AGM and marmosets; animals of both species succumbed between days 9 and 11 postexposure. Marmosets were susceptible to lower doses of RVFV than AGM. Histological examination confirmed viral meningoencephalitis in both species. Hematological analyses indicated a drop in platelet counts in both AGM and marmosets suggestive of thrombosis, as well as leukocytosis that consisted mostly of granulocytes. Both AGM and marmosets would serve as useful models of aerosol infection with RVFV.
Collapse
Affiliation(s)
- Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Diana S. Powell
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Laura M. Bethel
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy L. Caroline
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Richard J. Schmid
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tim Oury
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
29
|
Sumaye RD, Geubbels E, Mbeyela E, Berkvens D. Inter-epidemic transmission of Rift Valley fever in livestock in the Kilombero River Valley, Tanzania: a cross-sectional survey. PLoS Negl Trop Dis 2013; 7:e2356. [PMID: 23951376 PMCID: PMC3738442 DOI: 10.1371/journal.pntd.0002356] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 06/20/2013] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND In recent years, evidence of Rift Valley fever (RVF) transmission during inter-epidemic periods in parts of Africa has increasingly been reported. The inter-epidemic transmissions generally pass undetected where there is no surveillance in the livestock or human populations. We studied the presence of and the determinants for inter-epidemic RVF transmission in an area experiencing annual flooding in southern Tanzania. METHODOLOGY A cross-sectional sero-survey was conducted in randomly selected cattle, sheep and goats in the Kilombero river valley from May to August 2011, approximately four years after the 2006/07 RVF outbreak in Tanzania. The exposure status to RVF virus (RVFV) was determined using two commercial ELISA kits, detecting IgM and IgG antibodies in serum. Information about determinants was obtained through structured interviews with herd owners. FINDINGS An overall seroprevalence of 11.3% (n = 1680) was recorded; 5.5% in animals born after the 2006/07 RVF outbreak and 22.7% in animals present during the outbreak. There was a linear increase in prevalence in the post-epidemic annual cohorts. Nine inhibition-ELISA positive samples were also positive for RVFV IgM antibodies indicating a recent infection. The spatial distribution of seroprevalence exhibited a few hotspots. The sex difference in seroprevalence in animals born after the previous epidemic was not significant (6.1% vs. 4.6% for females and males respectively, p = 0.158) whereas it was significant in animals present during the outbreak (26.0% vs. 7.8% for females and males respectively, p<0.001). Animals living >15 km from the flood plain were more likely to have antibodies than those living <5 km (OR 1.92; 95% CI 1.04-3.56). Species, breed, herd composition, grazing practices and altitude were not associated with seropositivity. CONCLUSION These findings indicate post-epidemic transmission of RVFV in the study area. The linear increase in seroprevalence in the post-epidemic annual cohorts implies a constant exposure and presence of active foci transmission preceding the survey.
Collapse
|
30
|
Rift Valley fever virus clearance and protection from neurologic disease are dependent on CD4+ T cell and virus-specific antibody responses. J Virol 2013; 87:6161-71. [PMID: 23536675 DOI: 10.1128/jvi.00337-13] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rift Valley fever virus (RVFV) causes outbreaks of severe disease in people and livestock throughout Africa and the Arabian Peninsula. Human RVFV infections generally manifest as a self-limiting febrile illness, but in some individuals, the disease can progress to a fatal encephalitis or hemorrhagic syndrome. Little is known about the host characteristics that predispose development of more severe disease. Early in infection, interferon-mediated antiviral responses are critical for controlling RVFV replication, but the roles of downstream adaptive immune responses in determining clinical outcome have not been examined. Here, using a C57BL/6 mouse disease model, we evaluated the roles of B cells and T cells in RVFV pathogenesis. Given the profound inhibition of the innate response by the viral NSs protein and rapid course of wild-type infection, we utilized an attenuated RVFV lacking NSs to examine host responses following primary infection. Experiments utilizing B-cell-deficient mice or targeted T cell depletions of wild-type mice demonstrated that B cells and CD4(+) T cells, but not CD8(+) T cells, were critical for mediating viral clearance, even in the presence of a functional innate response. One-third of CD4-depleted mice developed severe neurologic disease following infection, in contrast to virus-infected mock-depleted mice that showed no clinical signs. CD4(+) T cells were required for robust IgG and neutralizing antibody responses that correlated with RVFV clearance from peripheral tissues. Furthermore, CD4-depleted mice demonstrated significantly stronger proinflammatory responses relative to controls, suggesting CD4(+) T cells regulate immune responses to RVFV infection. Together, these results indicate CD4(+) T cells are critical determinants of RVFV pathogenesis and play an important role in preventing onset of neurologic disease.
Collapse
|
31
|
Xu W, Watts DM, Costanzo MC, Tang X, Venegas LA, Jiao F, Sette A, Sidney J, Sewell AK, Wooldridge L, Makino S, Morrill JC, Peters CJ, Kan-Mitchell J. The nucleocapsid protein of Rift Valley fever virus is a potent human CD8+ T cell antigen and elicits memory responses. PLoS One 2013; 8:e59210. [PMID: 23527138 PMCID: PMC3601065 DOI: 10.1371/journal.pone.0059210] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/12/2013] [Indexed: 01/10/2023] Open
Abstract
There is no licensed human vaccine currently available for Rift Valley Fever Virus (RVFV), a Category A high priority pathogen and a serious zoonotic threat. While neutralizing antibodies targeting the viral glycoproteins are protective, they appear late in the course of infection, and may not be induced in time to prevent a natural or bioterrorism-induced outbreak. Here we examined the immunogenicity of RVFV nucleocapsid (N) protein as a CD8(+) T cell antigen with the potential for inducing rapid protection after vaccination. HLA-A*0201 (A2)-restricted epitopic determinants were identified with N-specific CD8(+) T cells from eight healthy donors that were primed with dendritic cells transduced to express N, and subsequently expanded in vitro by weekly re-stimulations with monocytes pulsed with 59 15mer overlapping peptides (OLPs) across N. Two immunodominant epitopes, VT9 (VLSEWLPVT, N(121-129)) and IL9 (ILDAHSLYL, N165-173), were defined. VT9- and IL9-specific CD8(+) T cells identified by tetramer staining were cytotoxic and polyfunctional, characteristics deemed important for viral control in vivo. These peptides induced specific CD8(+) T cell responses in A2-transgenic mice, and more importantly, potent N-specific CD8(+) T cell reactivities, including VT9- and IL9-specific ones, were mounted by mice after a booster vaccination with the live attenuated RVF MP-12. Our data suggest that the RVFV N protein is a potent human T cell immunogen capable of eliciting broad, immunodominant CD8(+) T cell responses that are potentially protective. Understanding the immune responses to the nucleocapsid is central to the design of an effective RVFV vaccine irrespective of whether this viral protein is effective as a stand-alone immunogen or only in combination with other RVFV antigens.
Collapse
Affiliation(s)
- Weidong Xu
- Department of Biological Science and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Head JA, Kalveram B, Ikegami T. Functional analysis of Rift Valley fever virus NSs encoding a partial truncation. PLoS One 2012; 7:e45730. [PMID: 23029207 PMCID: PMC3446906 DOI: 10.1371/journal.pone.0045730] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 08/20/2012] [Indexed: 12/13/2022] Open
Abstract
Rift Valley fever virus (RVFV), belongs to genus Phlebovirus of the family Bunyaviridae, causes high rates of abortion and fetal malformation in infected ruminants as well as causing neurological disorders, blindness, or lethal hemorrhagic fever in humans. RVFV is classified as a category A priority pathogen and a select agent in the U.S., and currently there are no therapeutics available for RVF patients. NSs protein, a major virulence factor of RVFV, inhibits host transcription including interferon (IFN)-β mRNA synthesis and promotes degradation of dsRNA-dependent protein kinase (PKR). NSs self-associates at the C-terminus 17 aa., while NSs at aa.210–230 binds to Sin3A-associated protein (SAP30) to inhibit the activation of IFN-β promoter. Thus, we hypothesize that NSs function(s) can be abolished by truncation of specific domains, and co-expression of nonfunctional NSs with intact NSs will result in the attenuation of NSs function by dominant-negative effect. Unexpectedly, we found that RVFV NSs truncated at aa. 6–30, 31–55, 56–80, 81–105, 106–130, 131–155, 156–180, 181–205, 206–230, 231–248 or 249–265 lack functions of IFN–β mRNA synthesis inhibition and degradation of PKR. Truncated NSs were less stable in infected cells, while nuclear localization was inhibited in NSs lacking either of aa.81–105, 106–130, 131–155, 156–180, 181–205, 206–230 or 231–248. Furthermore, none of truncated NSs had exhibited significant dominant-negative functions for NSs-mediated IFN-β suppression or PKR degradation upon co-expression in cells infected with RVFV. We also found that any of truncated NSs except for intact NSs does not interact with RVFV NSs even in the presence of intact C-terminus self-association domain. Our results suggest that conformational integrity of NSs is important for the stability, cellular localization and biological functions of RVFV NSs, and the co-expression of truncated NSs does not exhibit dominant-negative phenotype.
Collapse
Affiliation(s)
- Jennifer A. Head
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Birte Kalveram
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Tetsuro Ikegami
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Vaccine Development, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, The University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
33
|
Koukuntla R, Mandell RB, Flick R. Virus-Like Particle-Based Countermeasures Against Rift Valley Fever Virus. Zoonoses Public Health 2012; 59 Suppl 2:142-50. [DOI: 10.1111/j.1863-2378.2012.01478.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
34
|
The dominant-negative inhibition of double-stranded RNA-dependent protein kinase PKR increases the efficacy of Rift Valley fever virus MP-12 vaccine. J Virol 2012; 86:7650-61. [PMID: 22573861 DOI: 10.1128/jvi.00778-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rift Valley fever virus (RVFV), belonging to the genus Phlebovirus, family Bunyaviridae, is endemic to sub-Saharan Africa and causes a high rate of abortion in ruminants and hemorrhagic fever, encephalitis, or blindness in humans. MP-12 is the only RVFV strain excluded from the select-agent rule and handled at a biosafety level 2 (BSL2) laboratory. MP-12 encodes a functional major virulence factor, the NSs protein, which contributes to its residual virulence in pregnant ewes. We found that 100% of mice subcutaneously vaccinated with recombinant MP-12 (rMP12)-murine PKRN167 (mPKRN167), which encodes a dominant-negative form of mouse double-stranded RNA (dsRNA)-dependent protein kinase (PKR) in place of NSs, were protected from wild-type (wt) RVFV challenge, while 72% of mice vaccinated with MP-12 were protected after challenge. rMP12-mPKRN167 induced alpha interferon (IFN-α) in sera, accumulated RVFV antigens in dendritic cells at the local draining lymph nodes, and developed high levels of neutralizing antibodies, while parental MP-12 induced neither IFN-α nor viral-antigen accumulation at the draining lymph node yet induced a high level of neutralizing antibodies. The present study suggests that the expression of a dominant-negative PKR increases the immunogenicity and efficacy of live-attenuated RVFV vaccine, which will lead to rational design of safe and highly immunogenic RVFV vaccines for livestock and humans.
Collapse
|
35
|
Kortekaas J, Antonis AFG, Kant J, Vloet RPM, Vogel A, Oreshkova N, de Boer SM, Bosch BJ, Moormann RJM. Efficacy of three candidate Rift Valley fever vaccines in sheep. Vaccine 2012; 30:3423-9. [PMID: 22449427 DOI: 10.1016/j.vaccine.2012.03.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 03/04/2012] [Accepted: 03/12/2012] [Indexed: 11/18/2022]
Abstract
Rift Valley fever virus (RVFV) is a mosquito-transmitted Bunyavirus that causes high morbidity and mortality among ruminants and humans. The virus is endemic to the African continent and the Arabian Peninsula and continues to spread into new areas. The explosive nature of RVF outbreaks requires that vaccines provide swift protection after a single vaccination. We recently developed several candidate vaccines and here report their efficacy in lambs within three weeks after a single vaccination. The first vaccine comprises the purified ectodomain of the Gn structural glycoprotein formulated in a water-in-oil adjuvant. The second vaccine is based on a Newcastle disease virus-based vector that produces both RVFV structural glycoproteins Gn and Gc. The third vaccine comprises a recently developed nonspreading RVFV. The latter two vaccines were administered without adjuvant. The inactivated whole virus-based vaccine produced by Onderstepoort Biological Products was used as a positive control. Five out of six mock-vaccinated lambs developed high viremia and fever and one lamb succumbed to the challenge infection. A single vaccination with each vaccine resulted in a neutralizing antibody response within three weeks after vaccination and protected lambs from viremia, pyrexia and mortality.
Collapse
Affiliation(s)
- J Kortekaas
- Department of Virology, Central Veterinary Institute of Wageningen University and Research Centre, 8200 AB Lelystad, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|