1
|
Zhou CM, Jiang ZZ, Liu N, Yu XJ. Current insights into human pathogenic phenuiviruses and the host immune system. Virulence 2024; 15:2384563. [PMID: 39072499 PMCID: PMC11290763 DOI: 10.1080/21505594.2024.2384563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Phenuiviruses are a class of segmented negative-sense single-stranded RNA viruses, typically consisting of three RNA segments that encode four distinct proteins. The emergence of pathogenic phenuivirus strains, such as Rift Valley fever phlebovirus (RVFV) in sub-Saharan Africa, Severe Fever with Thrombocytopenia Syndrome Virus (SFTSV) in East and Southeast Asia, and Heartland Virus (HRTV) in the United States has presented considerable challenges to global public health in recent years. The innate immune system plays a crucial role as the initial defense mechanism of the host against invading pathogens. In addition to continued research aimed at elucidating the epidemiological characteristics of phenuivirus, significant advancements have been made in investigating its viral virulence factors (glycoprotein, non-structural protein, and nucleoprotein) and potential host-pathogen interactions. Specifically, efforts have focused on understanding mechanisms of viral immune evasion, viral assembly and egress, and host immune networks involving immune cells, programmed cell death, inflammation, nucleic acid receptors, etc. Furthermore, a plethora of technological advancements, including metagenomics, metabolomics, single-cell transcriptomics, proteomics, gene editing, monoclonal antibodies, and vaccines, have been utilized to further our understanding of phenuivirus pathogenesis and host immune responses. Hence, this review aims to provide a comprehensive overview of the current understanding of the mechanisms of host recognition, viral immune evasion, and potential therapeutic approaches during human pathogenic phenuivirus infections focusing particularly on RVFV and SFTSV.
Collapse
Affiliation(s)
- Chuan-Min Zhou
- Gastrointestinal Disease Diagnosis and Treatment Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ze-Zheng Jiang
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan, China
| | - Ning Liu
- Department of Quality and Operations Management, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xue-Jie Yu
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan, China
| |
Collapse
|
2
|
Shalamova L, Barth P, Pickin MJ, Kouti K, Ott B, Humpert K, Janssen S, Lorenzo G, Brun A, Goesmann A, Hain T, Hartmann RK, Rossbach O, Weber F. Nucleocapsids of the Rift Valley fever virus ambisense S segment contain an exposed RNA element in the center that overlaps with the intergenic region. Nat Commun 2024; 15:7602. [PMID: 39217162 PMCID: PMC11365940 DOI: 10.1038/s41467-024-52058-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen. Its RNA genome consists of two negative-sense segments (L and M) with one gene each, and one ambisense segment (S) with two opposing genes separated by the noncoding "intergenic region" (IGR). These vRNAs and the complementary cRNAs are encapsidated by nucleoprotein (N). Using iCLIP2 (individual-nucleotide resolution UV crosslinking and immunoprecipitation) to map all N-vRNA and N-cRNA interactions, we detect N coverage along the L and M segments. However, the S segment vRNA and cRNA each contain approximately 100 non-encapsidated nucleotides stretching from the IGR into the 5'-adjacent reading frame. These exposed regions are RNase-sensitive and predicted to form stem-loop structures with the mRNA transcription termination motif positioned near the top. Moreover, optimal S segment transcription and replication requires the entire exposed region rather than only the IGR. Thus, the RVFV S segment contains a central, non-encapsidated RNA region with a functional role.
Collapse
Affiliation(s)
- Lyudmila Shalamova
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Patrick Barth
- Bioinformatics & Systems Biology, Justus-Liebig University, Giessen, Germany
- Cell Biology and Plant Biochemistry, University of Regensburg, Regensburg, Germany
| | - Matthew J Pickin
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Kiriaki Kouti
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Benjamin Ott
- Institute for Medical Microbiology, FB11-Medicine, Justus-Liebig University, Giessen, Germany
| | - Katharina Humpert
- Institute for Medical Microbiology, FB11-Medicine, Justus-Liebig University, Giessen, Germany
- Institute of Molecular Oncology, Genomics Core Facility, Philipps-University, Marburg, Germany
| | - Stefan Janssen
- Algorithmic Bioinformatics, Justus-Liebig University, Giessen, Germany
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, Madrid, Spain
| | - Alejandro Brun
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, Madrid, Spain
| | - Alexander Goesmann
- Bioinformatics & Systems Biology, Justus-Liebig University, Giessen, Germany
| | - Torsten Hain
- Institute for Medical Microbiology, FB11-Medicine, Justus-Liebig University, Giessen, Germany
| | - Roland K Hartmann
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Oliver Rossbach
- Institute for Biochemistry, FB 08-Biology and Chemistry, Justus-Liebig University, Giessen, Germany
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany.
| |
Collapse
|
3
|
Huang J, Adungo F, Konongoi SL, Inoue S, Zhan L, Sang R, Ashur S, Kwallah AO, Mwau M, Morita K, Yu F. Monoclonal Antibodies for Rift Valley Fever Virus Nucleocapsid: Application in IgG/IgM ELISA for Sero-Diagnosis. Pathogens 2024; 13:582. [PMID: 39057809 PMCID: PMC11279541 DOI: 10.3390/pathogens13070582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/07/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION Rift Valley fever virus (RVFV) belonging to the Phenuiviridae family is responsible for a zoonotic disease called Rift Valley fever (RVF). Currently, RVFV has spread from Africa to Asia, and due to its ability to cause high mortality rates, it has significantly impacted human health and economic development in many societies. Highly specific and sensitive systems for sero-diagnosis of RVFV infection are needed for clinical use. METHOD BALB/c mice were immunized with recombinant RVFV nucleocapsid (rRVFV-N) protein and the spleen cells fused with SP2/0 myeloma cells to create hybridoma cell lines. The secreted monoclonal antibodies (MAbs) were purified and characterized. Enzyme-linked immunosorbent assay (ELISA) systems for the detection of IgG and IgM using the new MAbs were established and evaluated. Serum samples from 96 volunteers and 93 patients of suspected RVF from Kenya were tested compared with the ELISA systems based on inactivated viruses and the rabbit polyclonal antibody. RESULT Three monoclonal antibodies against rRVFV-N protein were established. The performance of the MAb-based sandwich IgG ELISA and the IgM capture ELISA perfectly matched the ELISA systems using the inactivated virus or the polyclonal antibody. CONCLUSIONS Recombinant RVFV-N protein-specific MAbs were developed and they offer useful tools for RVFV studies. The MAb-based ELISA systems for detecting IgG and IgM offer safe and useful options for diagnosing RVFV infections in humans.
Collapse
Affiliation(s)
- Jiansheng Huang
- School of Medicine, Guizhou University, Guiyang 550002, China;
| | - Ferdinard Adungo
- Kenya Medical Research Institute, Nairobi 54840-00200, Kenya; (F.A.); (S.L.K.); (R.S.); (S.A.); (A.o.K.); (M.M.)
| | - Samson Limbaso Konongoi
- Kenya Medical Research Institute, Nairobi 54840-00200, Kenya; (F.A.); (S.L.K.); (R.S.); (S.A.); (A.o.K.); (M.M.)
| | - Shingo Inoue
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan; (S.I.); (K.M.)
| | - Lin Zhan
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, Guiyang 550002, China;
- Guizhou Provincial People’s Hospital, Guizhou University, Guiyang 550002, China
| | - Rosemary Sang
- Kenya Medical Research Institute, Nairobi 54840-00200, Kenya; (F.A.); (S.L.K.); (R.S.); (S.A.); (A.o.K.); (M.M.)
| | - Salame Ashur
- Kenya Medical Research Institute, Nairobi 54840-00200, Kenya; (F.A.); (S.L.K.); (R.S.); (S.A.); (A.o.K.); (M.M.)
| | - Allan ole Kwallah
- Kenya Medical Research Institute, Nairobi 54840-00200, Kenya; (F.A.); (S.L.K.); (R.S.); (S.A.); (A.o.K.); (M.M.)
| | - Matilu Mwau
- Kenya Medical Research Institute, Nairobi 54840-00200, Kenya; (F.A.); (S.L.K.); (R.S.); (S.A.); (A.o.K.); (M.M.)
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan; (S.I.); (K.M.)
| | - Fuxun Yu
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, Guiyang 550002, China;
- Guizhou Provincial People’s Hospital, Guizhou University, Guiyang 550002, China
| |
Collapse
|
4
|
Intact Type I Interferon Receptor Signaling Prevents Hepatocellular Necrosis but Not Encephalitis in a Dose-Dependent Manner in Rift Valley Fever Virus Infected Mice. Int J Mol Sci 2022; 23:ijms232012492. [DOI: 10.3390/ijms232012492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Rift Valley fever (RVF) is a zoonotic and emerging disease, caused by the RVF virus (RVFV). In ruminants, it leads to “abortion storms” and enhanced mortality rates in young animals, whereas in humans it can cause symptoms like severe hemorrhagic fever or encephalitis. The role of the innate and adaptive immune response in disease initiation and progression is still poorly defined. The present study used the attenuated RVFV strain clone 13 to investigate viral spread, tissue tropism, and histopathological lesions after intranasal infection in C57BL/6 wild type (WT) and type I interferon (IFN-I) receptor I knockout (IFNAR−/−) mice. In WT mice, 104 PFU RVFV (high dose) resulted in a fatal encephalitis, but no hepatitis 7–11 days post infection (dpi), whereas 103 PFU RVFV (low dose) did not cause clinical disease or significant histopathological lesions in liver and the central nervous system (CNS). In contrast, IFNAR−/− mice infected with 103 PFU RVFV developed hepatocellular necrosis resulting in death at 2–5 dpi and lacked encephalitis. These results show that IFNAR signaling prevents systemic spread of the attenuated RVFV strain clone 13, but not the dissemination to the CNS and subsequent fatal disease. Consequently, neurotropic viruses may be able to evade antiviral IFN-I signaling pathways by using the transneuronal instead of the hematogenous route.
Collapse
|
5
|
Fatima I, Ahmad S, Abbasi SW, Ashfaq UA, Shahid F, Tahir Ul Qamar M, Rehman A, Allemailem KS. Designing of a multi-epitopes-based peptide vaccine against rift valley fever virus and its validation through integrated computational approaches. Comput Biol Med 2022; 141:105151. [PMID: 34942394 DOI: 10.1016/j.compbiomed.2021.105151] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 01/23/2023]
Abstract
Since its discovery, the Rift Valley Fever virus (RVFV) has been the source of numerous outbreaks in the Arab Peninsulas and Africa, wreaking havoc on humans and animals. The lack of therapeutics or licensed human vaccines limits the options for controlling RVFV outbreaks. Therefore, RVFV has been prioritized for rapid research and innovation of prevention strategies to control and prevent its outbreaks. The purpose of this study was to design a multi-epitope-based peptide vaccine (MEBPV) against RVFV. Bioinformatics approaches were used to design a potent MEBPV that can potentially activate both CD8+ and CD4+ T-cell immune responses, and several computational tools were employed to investigate its biological activities. Three antigenic proteins (Nucleocapsid (N), Glycoprotein C (GC), and Glycoprotein N (GN)) from the RVFV were chosen and potential immunogenic T- and B -cell epitopes were predicted from them. Based on in silico analysis, a MEBPV based on highly scored T and B-cell epitopes (6 CTL, 5 HTL, and 4 LBL) combined with linkers and adjuvants was developed. The finest predicted model was used for docking studies with Toll-like receptors (TLR3 and TLR8) and MHC molecules (MHC I and MHC II) after predicting and analyzing the tertiary structure of MEBPV. The designed MEBPV was then tested for stability with TLR3 and TLR8 receptors using molecular dynamics (MD) simulation and MMGBSA analysis. The MEBPV -TLR3, MEBPV -TLR8, MEBPV-MHC I and MEBPV -MHC II docked models were found stable during simulation time in MD and MMGBSA studies. In silico analysis revealed that the constructed vaccine could elicit both cell-mediated and humoral immune responses simultaneously. The proposed MEBPV could be a strong candidate against RVFV, but it will need to be tested in the laboratory to guarantee its safety and immunogenicity.
Collapse
Affiliation(s)
- Israr Fatima
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan.
| | - Sumra Wajid Abbasi
- NUMS Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Farah Shahid
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | | | - Abdur Rehman
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia.
| |
Collapse
|
6
|
Host Cell Restriction Factors of Bunyaviruses and Viral Countermeasures. Viruses 2021; 13:v13050784. [PMID: 33925004 PMCID: PMC8146327 DOI: 10.3390/v13050784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 01/01/2023] Open
Abstract
The Bunyavirales order comprises more than 500 viruses (generally defined as bunyaviruses) classified into 12 families. Some of these are highly pathogenic viruses infecting different hosts, including humans, mammals, reptiles, arthropods, birds, and/or plants. Host cell sensing of infection activates the innate immune system that aims at inhibiting viral replication and propagation. Upon recognition of pathogen-associated molecular patterns (PAMPs) by cellular pattern recognition receptors (PRRs), numerous signaling cascades are activated, leading to the production of interferons (IFNs). IFNs act in an autocrine and paracrine manner to establish an antiviral state by inducing the expression of hundreds of IFN-stimulated genes (ISGs). Some of these ISGs are known to restrict bunyavirus infection. Along with other constitutively expressed host cellular factors with antiviral activity, these proteins (hereafter referred to as “restriction factors”) target different steps of the viral cycle, including viral entry, genome transcription and replication, and virion egress. In reaction to this, bunyaviruses have developed strategies to circumvent this antiviral response, by avoiding cellular recognition of PAMPs, inhibiting IFN production or interfering with the IFN-mediated response. Herein, we review the current knowledge on host cellular factors that were shown to restrict infections by bunyaviruses. Moreover, we focus on the strategies developed by bunyaviruses in order to escape the antiviral state developed by the infected cells.
Collapse
|
7
|
A Look into Bunyavirales Genomes: Functions of Non-Structural (NS) Proteins. Viruses 2021; 13:v13020314. [PMID: 33670641 PMCID: PMC7922539 DOI: 10.3390/v13020314] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
In 2016, the Bunyavirales order was established by the International Committee on Taxonomy of Viruses (ICTV) to incorporate the increasing number of related viruses across 13 viral families. While diverse, four of the families (Peribunyaviridae, Nairoviridae, Hantaviridae, and Phenuiviridae) contain known human pathogens and share a similar tri-segmented, negative-sense RNA genomic organization. In addition to the nucleoprotein and envelope glycoproteins encoded by the small and medium segments, respectively, many of the viruses in these families also encode for non-structural (NS) NSs and NSm proteins. The NSs of Phenuiviridae is the most extensively studied as a host interferon antagonist, functioning through a variety of mechanisms seen throughout the other three families. In addition, functions impacting cellular apoptosis, chromatin organization, and transcriptional activities, to name a few, are possessed by NSs across the families. Peribunyaviridae, Nairoviridae, and Phenuiviridae also encode an NSm, although less extensively studied than NSs, that has roles in antagonizing immune responses, promoting viral assembly and infectivity, and even maintenance of infection in host mosquito vectors. Overall, the similar and divergent roles of NS proteins of these human pathogenic Bunyavirales are of particular interest in understanding disease progression, viral pathogenesis, and developing strategies for interventions and treatments.
Collapse
|
8
|
Kroeker AL, Babiuk S, Pickering BS, Richt JA, Wilson WC. Livestock Challenge Models of Rift Valley Fever for Agricultural Vaccine Testing. Front Vet Sci 2020; 7:238. [PMID: 32528981 PMCID: PMC7266933 DOI: 10.3389/fvets.2020.00238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 04/07/2020] [Indexed: 11/13/2022] Open
Abstract
Since the discovery of Rift Valley Fever virus (RVFV) in Kenya in 1930, the virus has become widespread throughout most of Africa and is characterized by sporadic outbreaks. A mosquito-borne pathogen, RVFV is poised to move beyond the African continent and the Middle East and emerge in Europe and Asia. There is a risk that RVFV could also appear in the Americas, similar to the West Nile virus. In light of this potential threat, multiple studies have been undertaken to establish international surveillance programs and diagnostic tools, develop models of transmission dynamics and risk factors for infection, and to develop a variety of vaccines as countermeasures. Furthermore, considerable efforts to establish reliable challenge models of Rift Valley fever virus have been made and platforms for testing potential vaccines and therapeutics in target species have been established. This review emphasizes the progress and insights from a North American perspective to establish challenge models in target livestock such as cattle, sheep, and goats in comparisons to other researchers' reports. A brief summary of the potential role of wildlife, such as buffalo and white-tailed deer as reservoir species will also be discussed.
Collapse
Affiliation(s)
- Andrea Louise Kroeker
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Bradley S Pickering
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Juergen A Richt
- Center of Excellence for Emerging and Zoonotic Animal Diseases (CEEZAD), Manhattan, KS, United States
| | - William C Wilson
- USDA, Arthropod-Borne Animal Diseases Research Unit (ABADRU), Manhattan, KS, United States
| |
Collapse
|
9
|
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: 49] [Impact Index Per Article: 9.8] [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
|
10
|
Zivcec M, Spiropoulou CF, Spengler JR. The use of mice lacking type I or both type I and type II interferon responses in research on hemorrhagic fever viruses. Part 2: Vaccine efficacy studies. Antiviral Res 2020; 174:104702. [PMID: 31982149 DOI: 10.1016/j.antiviral.2019.104702] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/13/2019] [Accepted: 12/20/2019] [Indexed: 12/24/2022]
Abstract
For more than 20 years, researchers have used laboratory mice lacking type I or both type I and II interferon (IFN) responses to study high-containment viruses that cause hemorrhagic fevers (HF) in humans. With the exception of Rift Valley fever virus, agents that cause viral HF in humans, such as Ebola and Lassa virus, do not cause disease in mature immunocompetent mice. In contrast, IFN-deficient mice typically develop severe or fatal disease when inoculated with these agents. The sensitivity of IFN-deficient mice to disease has led to their widespread use in biocontainment laboratories to assess the efficacy of novel vaccines against HF viruses, often without considering whether adaptive immune responses in IFN-deficient mice accurately mirror those in immunocompetent humans. Failure to recognize these questions may lead to inappropriate expectations of the predictive value of mouse experiments. In two invited articles, we investigate these questions. The present article reviews the use of IFN-deficient mice for assessing novel vaccines against HF viruses, including Ebola, Lassa, Crimean-Congo hemorrhagic fever and Rift Valley fever viruses. A companion paper examines the general question of how the lack of IFN signaling may affect adaptive immune responses and the outcome of vaccine studies in mice.
Collapse
Affiliation(s)
- Marko Zivcec
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
11
|
Ter Horst S, Conceição-Neto N, Neyts J, Rocha-Pereira J. Structural and functional similarities in bunyaviruses: Perspectives for pan-bunya antivirals. Rev Med Virol 2019; 29:e2039. [PMID: 30746831 PMCID: PMC7169261 DOI: 10.1002/rmv.2039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/29/2018] [Accepted: 01/17/2019] [Indexed: 01/03/2023]
Abstract
The order of Bunyavirales includes numerous (re)emerging viruses that collectively have a major impact on human and animal health worldwide. There are no vaccines for human use or antiviral drugs available to prevent or treat infections with any of these viruses. The development of efficacious and safe drugs and vaccines is a pressing matter. Ideally, such antivirals possess pan‐bunyavirus antiviral activity, allowing the containment of every bunya‐related threat. The fact that many bunyaviruses need to be handled in laboratories with biosafety level 3 or 4, the great variety of species and the frequent emergence of novel species complicate such efforts. We here examined the potential druggable targets of bunyaviruses, together with the level of conservation of their biological functions, structure, and genetic similarity by means of heatmap analysis. In the light of this, we revised the available models and tools currently available, pointing out directions for antiviral drug discovery.
Collapse
Affiliation(s)
- Sebastiaan Ter Horst
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Nádia Conceição-Neto
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Johan Neyts
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Joana Rocha-Pereira
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| |
Collapse
|
12
|
The One Health Approach is Necessary for the Control of Rift Valley Fever Infections in Egypt: A Comprehensive Review. Viruses 2019; 11:v11020139. [PMID: 30736362 PMCID: PMC6410127 DOI: 10.3390/v11020139] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/02/2019] [Accepted: 02/03/2019] [Indexed: 11/17/2022] Open
Abstract
Rift Valley fever (RVF) is an emerging transboundary, mosquito-borne, zoonotic viral disease caused high morbidity and mortality in both human and ruminant populations. It is considered an important threat to both agriculture and public health in African and the Middle Eastern countries including Egypt. Five major RVF epidemics have been reported in Egypt (1977, 1993, 1994, 1997, and 2003). The virus is transmitted in Egypt by different mosquito’s genera such as Aedes, Culex, Anopheles, and Mansonia, leading to abortions in susceptible animal hosts especially sheep, goat, cattle, and buffaloes. Recurrent RVF outbreaks in Egypt have been attributed in part to the lack of routine surveillance for the virus. These periodic epizootics have resulted in severe economic losses. We posit that there is a critical need for new approaches to RVF control that will prevent or at least reduce future morbidity and economic stress. One Health is an integrated approach for the understanding and management of animal, human, and environmental determinants of complex problems such as RVF. Employing the One Health approach, one might engage local communities in surveillance and control of RVF efforts, rather than continuing their current status as passive victims of the periodic RVF incursions. This review focuses upon endemic and epidemic status of RVF in Egypt, the virus vectors and their ecology, transmission dynamics, risk factors, and the ecology of the RVF at the animal/human interface, prevention, and control measures, and the use of environmental and climate data in surveillance systems to predict disease outbreaks.
Collapse
|
13
|
Pinkham C, Ahmed A, Bracci N, Narayanan A, Kehn-Hall K. Host-based processes as therapeutic targets for Rift Valley fever virus. Antiviral Res 2018; 160:64-78. [PMID: 30316916 DOI: 10.1016/j.antiviral.2018.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/27/2018] [Accepted: 10/05/2018] [Indexed: 12/28/2022]
Abstract
Rift Valley fever virus (RVFV) is an enveloped, segmented, negative sense RNA virus that replicates within the host's cytoplasm. To facilitate its replication, RVFV must utilize host cell processes and as such, these processes may serve as potential therapeutic targets. This review summarizes key host cell processes impacted by RVFV infection. Specifically the influence of RVFV on host transcriptional regulation, post-transcriptional regulation, protein half-life and availability, host signal transduction, trafficking and secretory pathways, cytoskeletal modulation, and mitochondrial processes and oxidative stress are discussed. Therapeutics targeted towards host processes that are essential for RVFV to thrive as well as their efficacy and importance to viral pathogenesis are highlighted.
Collapse
Affiliation(s)
- Chelsea Pinkham
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Aslaa Ahmed
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Nicole Bracci
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA.
| |
Collapse
|
14
|
Kim HJ, Lyoo HR, Choi JS, Lee YH, Kim BH, Yoo HS. Development of a Quantitative RT-PCR Assay to Differentiate Rift Valley Fever Virus Smithburn Vaccine Strain from Clone 13 Vaccine Strain. Vector Borne Zoonotic Dis 2018; 19:121-127. [PMID: 30300113 DOI: 10.1089/vbz.2018.2342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A new quantitative RT-PCR assay was developed to differentiate Rift Valley fever (RVF) Smithburn vaccine strain from Clone 13 vaccine strain. The new qRT-PCR assay targeting the S segment (NSs and N gene) was tested on synthesized standard RNA and MP-12 strain viruses. The detection limit of the new qRT-PCR assay is 1 copy/μL of NSs and N, and is able to differentiate the Smithburn vaccine strain of RVF from the Clone 13 vaccine strain. No cross-reactivity with other vector-borne viruses was observed, a factor that is especially important in the Republic of Korea (ROK). To examine the performance of the qRT-PCR, intra- and inter-assay variability data were analyzed and showed high reproducibility. These results indicate that the new qRT-PCR can be used as a safe and cost-effective test. Furthermore, this result suggests the possibility of differentiation between infected and vaccinated animals diagnostic test in RVF-free countries including ROK.
Collapse
Affiliation(s)
- Hyun-Joo Kim
- 1 Foreign Animal Disease Division, Animal and Plant Quarantine Agency , Gimcheon-si, Republic of Korea.,2 Department of Infectious Diseases, Colleges of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| | - Hye-Rhyoung Lyoo
- 3 Department of Infectious Diseases and Immunology, Virology Division, Faculty of Veterinary Medicine, Utrecht University , Utrecht, the Netherlands
| | - Jeong-Soo Choi
- 1 Foreign Animal Disease Division, Animal and Plant Quarantine Agency , Gimcheon-si, Republic of Korea
| | - Yoon-Hee Lee
- 1 Foreign Animal Disease Division, Animal and Plant Quarantine Agency , Gimcheon-si, Republic of Korea
| | - Byoung-Han Kim
- 1 Foreign Animal Disease Division, Animal and Plant Quarantine Agency , Gimcheon-si, Republic of Korea
| | - Han Sang Yoo
- 2 Department of Infectious Diseases, Colleges of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| |
Collapse
|
15
|
Smith DR, Johnston SC, Piper A, Botto M, Donnelly G, Shamblin J, Albariño CG, Hensley LE, Schmaljohn C, Nichol ST, Bird BH. Attenuation and efficacy of live-attenuated Rift Valley fever virus vaccine candidates in non-human primates. PLoS Negl Trop Dis 2018; 12:e0006474. [PMID: 29742102 PMCID: PMC5962102 DOI: 10.1371/journal.pntd.0006474] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/21/2018] [Accepted: 04/23/2018] [Indexed: 11/18/2022] Open
Abstract
Rift Valley fever virus (RVFV) is an important mosquito-borne veterinary and human pathogen that has caused large outbreaks of severe disease throughout Africa and the Arabian Peninsula. Currently, no licensed vaccine or therapeutics exists to treat this potentially deadly disease. The explosive nature of RVFV outbreaks and the severe consequences of its accidental or intentional introduction into RVFV-free areas provide the impetus for the development of novel vaccine candidates for use in both livestock and humans. Rationally designed vaccine candidates using reverse genetics have been used to develop deletion mutants of two known RVFV virulence factors, the NSs and NSm genes. These recombinant viruses were demonstrated to be protective and immunogenic in rats, mice, and sheep, without producing clinical illness in these animals. Here, we expand upon those findings and evaluate the single deletion mutant (ΔNSs rRVFV) and double deletion mutant (ΔNSs-ΔNSm rRVFV) vaccine candidates in the common marmoset (Callithrix jacchus), a non-human primate (NHP) model resembling severe human RVF disease. We demonstrate that both the ΔNSs and ΔNSs-ΔNSm rRVFV vaccine candidates were found to be safe and immunogenic in the current study. The vaccinated animals received a single dose of vaccine that led to the development of a robust antibody response. No vaccine-induced adverse reactions, signs of clinical illness or infectious virus were detected in the vaccinated marmosets. All vaccinated animals that were subsequently challenged with RVFV were protected against viremia and liver disease. In summary, our results provide the basis for further development of the ΔNSs and ΔNSs-ΔNSm rRVFV as safe and effective human RVFV vaccines for this significant public health threat. Rift Valley fever (RVF) is an important neglected tropical disease that has caused severe epidemics and epizootics throughout Africa and the Arabian Peninsula. Severe outbreaks have involved tens of thousands of both human and livestock cases for which no effective, commercially available human vaccines are available. Vaccine candidates have been developed based on the complete deletion of two known RVF virus virulence factors, the NSs and NSm genes. These vaccines were previously demonstrated to be protective in rats, mice, and sheep. In this study, we expand upon those results and evaluate the vaccine candidates in a non-human primate model for RVF. The animals received a single dose of vaccine that led to the development of a robust immune response. No vaccine-induced adverse reactions, signs of clinical illness or infectious virus were detected in the vaccinated animals. All vaccinated animals that were subsequently challenged with RVF virus were protected against viremia and liver disease. These results demonstrate that the vaccines are safe and effective in non-human primates, which provides the impetus for further development of these candidates for use in humans.
Collapse
Affiliation(s)
- Darci R. Smith
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
- * E-mail:
| | - Sara C. Johnston
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - Ashley Piper
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - Miriam Botto
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - Ginger Donnelly
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - Joshua Shamblin
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - César G. Albariño
- Centers for Disease Control and Prevention, Viral Special Pathogens Branch, Atlanta, GA, United States of America
| | - Lisa E. Hensley
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - Connie Schmaljohn
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - Stuart T. Nichol
- Centers for Disease Control and Prevention, Viral Special Pathogens Branch, Atlanta, GA, United States of America
| | - Brian H. Bird
- Centers for Disease Control and Prevention, Viral Special Pathogens Branch, Atlanta, GA, United States of America
| |
Collapse
|
16
|
Atkins C, Freiberg AN. Recent advances in the development of antiviral therapeutics for Rift Valley fever virus infection. Future Virol 2017; 12:651-665. [PMID: 29181086 DOI: 10.2217/fvl-2017-0060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/26/2017] [Indexed: 12/25/2022]
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus endemic to sub-Saharan Africa and the Arabian Peninsula and the etiological agent of Rift Valley fever. Rift Valley fever is a disease of major public health and economic concern, affecting livestock and humans. In ruminants, RVFV infection is characterized by high mortality rates in newborns and near 100% abortion rates in pregnant animals. Infection in humans is typically manifested as a self-limiting febrile illness, but can lead to severe and fatal hepatitis, encephalitis, hemorrhagic fever or retinitis with partial or complete blindness. Currently, there are no specific treatment options available for RVFV infection. This review presents a summary of the therapeutic approaches that have been explored on the treatment of RVFV infection.
Collapse
Affiliation(s)
- Colm Atkins
- Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Alexander N Freiberg
- Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Sealy Center for Vaccine Development, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Center for Biodefense & Emerging Infectious Diseases, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Sealy Center for Vaccine Development, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Center for Biodefense & Emerging Infectious Diseases, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| |
Collapse
|
17
|
Barski M, Brennan B, Miller OK, Potter JA, Vijayakrishnan S, Bhella D, Naismith JH, Elliott RM, Schwarz-Linek U. Rift Valley fever phlebovirus NSs protein core domain structure suggests molecular basis for nuclear filaments. eLife 2017; 6. [PMID: 28915104 PMCID: PMC5601994 DOI: 10.7554/elife.29236] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/21/2017] [Indexed: 12/24/2022] Open
Abstract
Rift Valley fever phlebovirus (RVFV) is a clinically and economically important pathogen increasingly likely to cause widespread epidemics. RVFV virulence depends on the interferon antagonist non-structural protein (NSs), which remains poorly characterized. We identified a stable core domain of RVFV NSs (residues 83–248), and solved its crystal structure, a novel all-helical fold organized into highly ordered fibrils. A hallmark of RVFV pathology is NSs filament formation in infected cell nuclei. Recombinant virus encoding the NSs core domain induced intranuclear filaments, suggesting it contains all essential determinants for nuclear translocation and filament formation. Mutations of key crystal fibril interface residues in viruses encoding full-length NSs completely abrogated intranuclear filament formation in infected cells. We propose the fibrillar arrangement of the NSs core domain in crystals reveals the molecular basis of assembly of this key virulence factor in cell nuclei. Our findings have important implications for fundamental understanding of RVFV virulence. Rift Valley fever phlebovirus (RVFV) is a virus of humans and livestock, transmitted by mosquitos and contact with infected animals. Infection can cause severe disease, including hemorrhagic fever, and may lead to death. Historically, the virus was only found in central Africa but it has spread for instance to the Arabian Peninsula. There is a risk that the virus may appear in temperate regions including Europe because global warming is allowing the mosquitos that carry the virus to extend their geographic range. There are no vaccines or treatments available for use in humans so if there is a serious outbreak of the virus it could become an epidemic and cause great economic losses and severe human disease. RVFV relies on a protein called NSs to cause disease. In cells of infected animals and humans NSs forms filaments inside the nucleus, the control center of the cell, and disarms the immune system. However, it is not known precisely how NSs works. To address this question, Barski, Brennan et al. used a technique called X-ray crystallography to study the atomic three-dimensional structure of NSs. This revealed that the center of the protein contains a core domain that causes the filaments to form. Further experiments identified how the NSs core comes together to build the filaments inside the cell nucleus. These findings represent an important step towards understanding how the NSs protein helps RVFV to cause disease in humans and livestock. In the future, this work may aid the development of much needed drugs and vaccines against RVFV.
Collapse
Affiliation(s)
- Michal Barski
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Benjamin Brennan
- MRC-University of Glasgow Centre for Virus Research, London, United Kingdom
| | - Ona K Miller
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Jane A Potter
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | | | - David Bhella
- MRC-University of Glasgow Centre for Virus Research, London, United Kingdom
| | - James H Naismith
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Richard M Elliott
- MRC-University of Glasgow Centre for Virus Research, London, United Kingdom
| | - Ulrich Schwarz-Linek
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
18
|
Mapping of Transcription Termination within the S Segment of SFTS Phlebovirus Facilitated Generation of NSs Deletant Viruses. J Virol 2017; 91:JVI.00743-17. [PMID: 28592543 PMCID: PMC5533932 DOI: 10.1128/jvi.00743-17] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/31/2017] [Indexed: 12/15/2022] Open
Abstract
SFTS phlebovirus (SFTSV) is an emerging tick-borne bunyavirus that was first reported in China in 2009. Here we report the generation of a recombinant SFTSV (rHB29NSsKO) that cannot express the viral nonstructural protein (NSs) upon infection of cells in culture. We show that rHB29NSsKO replication kinetics are greater in interferon (IFN)-incompetent cells and that the virus is unable to suppress IFN induced in response to viral replication. The data confirm for the first time in the context of virus infection that NSs acts as a virally encoded IFN antagonist and that NSs is dispensable for virus replication. Using 3' rapid amplification of cDNA ends (RACE), we mapped the 3' end of the N and NSs mRNAs, showing that the mRNAs terminate within the coding region of the opposite open reading frame. We show that the 3' end of the N mRNA terminates upstream of a 5'-GCCAGCC-3' motif present in the viral genomic RNA. With this knowledge, and using virus-like particles, we could demonstrate that the last 36 nucleotides of the NSs open reading frame (ORF) were needed to ensure the efficient termination of the N mRNA and were required for recombinant virus rescue. We demonstrate that it is possible to recover viruses lacking NSs (expressing just a 12-amino-acid NSs peptide or encoding enhanced green fluorescent protein [eGFP]) or an NSs-eGFP fusion protein in the NSs locus. This opens the possibility for further studies of NSs and potentially the design of attenuated viruses for vaccination studies.IMPORTANCE SFTS phlebovirus (SFTSV) and related tick-borne viruses have emerged globally since 2009. SFTSV has been shown to cause severe disease in humans. For bunyaviruses, it has been well documented that the nonstructural protein (NSs) enables the virus to counteract the human innate antiviral defenses and that NSs is one of the major determinants of virulence in infection. Therefore, the use of reverse genetics systems to engineer viruses lacking NSs is an attractive strategy to rationally attenuate bunyaviruses. Here we report the generation of several recombinant SFTS viruses that cannot express the NSs protein or have the NSs open reading frame replaced with a reporter gene. These viruses cannot antagonize the mammalian interferon (IFN) response mounted to virus infection. The generation of NSs-lacking viruses was achieved by mapping the transcriptional termination of two S-segment-derived subgenomic mRNAs, which revealed that transcription termination occurs upstream of a 5'-GCCAGCC-3' motif present in the virus genomic S RNA.
Collapse
|
19
|
Wichgers Schreur PJ, van Keulen L, Kant J, Kortekaas J. Four-segmented Rift Valley fever virus-based vaccines can be applied safely in ewes during pregnancy. Vaccine 2017; 35:3123-3128. [PMID: 28457675 DOI: 10.1016/j.vaccine.2017.04.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 12/30/2022]
Abstract
Rift Valley fever virus (RVFV) causes severe and recurrent outbreaks on the African continent and the Arabian Peninsula and continues to expand its habitat. This mosquito-borne virus, belonging to the genus Phlebovirus of the family Bunyaviridae contains a tri-segmented negative-strand RNA genome. Previously, we developed four-segmented RVFV (RVFV-4s) variants by splitting the M-genome segment into two M-type segments each encoding one of the structural glycoproteins; Gn or Gc. Vaccination/challenge experiments with mice and lambs subsequently showed that RVFV-4s induces protective immunity against wild-type virus infection after a single administration. To demonstrate the unprecedented safety of RVFV-4s, we here report that the virus does not cause encephalitis after intranasal inoculation of mice. A study with pregnant ewes subsequently revealed that RVFV-4s does not cause viremia and does not cross the ovine placental barrier, as evidenced by the absence of teratogenic effects and virus in the blood and organs of the fetuses. Altogether, these results show that the RVFV-4s vaccine virus can be applied safely in pregnant ewes.
Collapse
Affiliation(s)
| | - Lucien van Keulen
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Jet Kant
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Jeroen Kortekaas
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| |
Collapse
|
20
|
Rissmann M, Ulrich R, Schröder C, Hammerschmidt B, Hanke D, Mroz C, Groschup MH, Eiden M. Vaccination of alpacas against Rift Valley fever virus: Safety, immunogenicity and pathogenicity of MP-12 vaccine. Vaccine 2016; 35:655-662. [PMID: 28012779 DOI: 10.1016/j.vaccine.2016.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/25/2016] [Accepted: 12/01/2016] [Indexed: 02/07/2023]
Abstract
Rift Valley fever (RVF) is an emerging zoonosis of major public health concern in Africa and Arabia. Previous outbreaks attributed camelids a significant role in the epidemiology of Rift Valley fever virus (RVFV), making them an important target species for vaccination. Using three alpacas as model-organisms for dromedary camels, the safety, immunogenicity and pathogenicity of the MP-12 vaccine were evaluated in this study. To compare both acute and subacute effects, animals were euthanized at 3 and 31days post infection (dpi). Clinical monitoring, analysis of liver enzymes and hematological parameters demonstrated the tolerability of the vaccine, as no significant adverse effects were observed. Comprehensive analysis of serological parameters illustrated the immunogenicity of the vaccine, eliciting high neutralizing antibody titers and antibodies targeting different viral antigens. RVFV was detected in serum and liver of the alpaca euthanized 3dpi, whereas no virus was detectable at 31dpi. Viral replication was confirmed by detection of various RVFV-antigens in hepatocytes by immunohistochemistry and the presence of mild multifocal necrotizing hepatitis. In conclusion, results indicate that MP-12 is a promising vaccine candidate but still has a residual pathogenicity, which requires further investigation.
Collapse
Affiliation(s)
- M Rissmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - R Ulrich
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - C Schröder
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - B Hammerschmidt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - D Hanke
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - C Mroz
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - M H Groschup
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - M Eiden
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany.
| |
Collapse
|
21
|
Wuerth JD, Weber F. Phleboviruses and the Type I Interferon Response. Viruses 2016; 8:v8060174. [PMID: 27338447 PMCID: PMC4926194 DOI: 10.3390/v8060174] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022] Open
Abstract
The genus Phlebovirus of the family Bunyaviridae contains a number of emerging virus species which pose a threat to both human and animal health. Most prominent members include Rift Valley fever virus (RVFV), sandfly fever Naples virus (SFNV), sandfly fever Sicilian virus (SFSV), Toscana virus (TOSV), Punta Toro virus (PTV), and the two new members severe fever with thrombocytopenia syndrome virus (SFTSV) and Heartland virus (HRTV). The nonstructural protein NSs is well established as the main phleboviral virulence factor in the mammalian host. NSs acts as antagonist of the antiviral type I interferon (IFN) system. Recent progress in the elucidation of the molecular functions of a growing list of NSs proteins highlights the astonishing variety of strategies employed by phleboviruses to evade the IFN system.
Collapse
Affiliation(s)
- Jennifer Deborah Wuerth
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen 35392, Germany.
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen 35392, Germany.
| |
Collapse
|
22
|
Hobson-Peters J, Warrilow D, McLean BJ, Watterson D, Colmant AMG, van den Hurk AF, Hall-Mendelin S, Hastie ML, Gorman JJ, Harrison JJ, Prow NA, Barnard RT, Allcock R, Johansen CA, Hall RA. Discovery and characterisation of a new insect-specific bunyavirus from Culex mosquitoes captured in northern Australia. Virology 2016; 489:269-81. [PMID: 26773387 DOI: 10.1016/j.virol.2015.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 09/21/2015] [Accepted: 11/05/2015] [Indexed: 11/25/2022]
Abstract
Insect-specific viruses belonging to significant arboviral families have recently been discovered. These viruses appear to be maintained within the insect population without the requirement for replication in a vertebrate host. Mosquitoes collected from Badu Island in the Torres Strait in 2003 were analysed for insect-specific viruses. A novel bunyavirus was isolated in high prevalence from Culex spp. The new virus, provisionally called Badu virus (BADUV), replicated in mosquito cells of both Culex and Aedes origin, but failed to replicate in vertebrate cells. Genomic sequencing revealed that the virus was distinct from sequenced bunyavirus isolates reported to date, but phylogenetically clustered most closely with recently discovered mosquito-borne, insect-specific bunyaviruses in the newly proposed Goukovirus genus. The detection of a functional furin cleavage motif upstream of the two glycoproteins in the M segment-encoded polyprotein suggests that BADUV may employ a unique strategy to process the virion glycoproteins.
Collapse
Affiliation(s)
- Jody Hobson-Peters
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia.
| | - David Warrilow
- Public Health Virology Forensic and Scientific Services, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland 4108, Australia
| | - Breeanna J McLean
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Daniel Watterson
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Agathe M G Colmant
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Andrew F van den Hurk
- Public Health Virology Forensic and Scientific Services, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland 4108, Australia
| | - Sonja Hall-Mendelin
- Public Health Virology Forensic and Scientific Services, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland 4108, Australia
| | - Marcus L Hastie
- Protein Discovery Centre, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Jeffrey J Gorman
- Protein Discovery Centre, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Jessica J Harrison
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Natalie A Prow
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Ross T Barnard
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Richard Allcock
- Lottery West State Biomedical Facility - Genomics, School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia, Australia; Department of Clinical Immunology, Pathwest Laboratory Medicine Western Australia, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Cheryl A Johansen
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Roy A Hall
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia.
| |
Collapse
|
23
|
Mansfield KL, Banyard AC, McElhinney L, Johnson N, Horton DL, Hernández-Triana LM, Fooks AR. Rift Valley fever virus: A review of diagnosis and vaccination, and implications for emergence in Europe. Vaccine 2015; 33:5520-5531. [PMID: 26296499 DOI: 10.1016/j.vaccine.2015.08.020] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/12/2015] [Accepted: 08/05/2015] [Indexed: 12/14/2022]
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne virus, and is the causative agent of Rift Valley fever (RVF), a zoonotic disease characterised by an increased incidence of abortion or foetal malformation in ruminants. Infection in humans can also lead to clinical manifestations that in severe cases cause encephalitis or haemorrhagic fever. The virus is endemic throughout much of the African continent. However, the emergence of RVFV in the Middle East, northern Egypt and the Comoros Archipelago has highlighted that the geographical range of RVFV may be increasing, and has led to the concern that an incursion into Europe may occur. At present, there is a limited range of veterinary vaccines available for use in endemic areas, and there is no licensed human vaccine. In this review, the methods available for diagnosis of RVFV infection, the current status of vaccine development and possible implications for RVFV emergence in Europe, are discussed.
Collapse
Affiliation(s)
- Karen L Mansfield
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency, Woodham Lane, New Haw KT15 3NB, UK.
| | - Ashley C Banyard
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency, Woodham Lane, New Haw KT15 3NB, UK
| | - Lorraine McElhinney
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency, Woodham Lane, New Haw KT15 3NB, UK; NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool L69 7BE, UK
| | - Nicholas Johnson
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency, Woodham Lane, New Haw KT15 3NB, UK
| | - Daniel L Horton
- School of Veterinary Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Luis M Hernández-Triana
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency, Woodham Lane, New Haw KT15 3NB, UK
| | - Anthony R Fooks
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency, Woodham Lane, New Haw KT15 3NB, UK; NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool L69 7BE, UK; Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
| |
Collapse
|
24
|
Lo MM, Mbao V, Sierra P, Thiongane Y, Diop M, Donadeu M, Dungu B. Safety and immunogenicity of Onderstepoort Biological Products' Rift Valley fever Clone 13 vaccine in sheep and goats under field conditions in Senegal. ACTA ACUST UNITED AC 2015; 82:857. [PMID: 26244679 PMCID: PMC6238691 DOI: 10.4102/ojvr.v82i1.857] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 11/29/2022]
Abstract
This blinded field safety study was conducted in Senegal to assess safety and immunogenicity of administration of the registered dose of Rift Valley fever virus (RVFV) Clone 13 vaccine (Onderstepoort Biological Products) to sheep and goats of West African breeds under natural conditions. A total of 267 small ruminants (220 sheep, 47 goats) were included; half received RVFV Clone 13 vaccine at the recommended dose and half received the diluent (as placebo) only. The study was performed on three commercial farms in the northern and eastern region of Senegal in accordance with veterinary good clinical practices. The animals were observed daily for 3 days after vaccination, and then weekly for 1 year. In both sheep and goats vaccinated against RVFV seroconversion rates above 70% were recorded. No seroconversion related to RVFV was observed in placebo-treated animals. No statistically significant differences were determined between placebo and vaccinated groups for mean rectal temperatures for the first 3 days after administration (p > 0.05). No abnormal clinical signs related to treatment were noted, and only one slight injection site reaction was observed in one vaccinated animal for 2 days after vaccination. Out of 176 births assessed over 1 year (93 from the vaccinated group, 83 from the placebo group), 9 were abnormal in the placebo group and 3 in the vaccinated group (p > 0.05). The frequency of adverse events was similar in the placebo and vaccinated groups. RVFV Clone 13 vaccine administered according to the manufacturer's instructions was safe and well tolerated in West African breeds of sheep and goats, including animals of approximately 6 months of age and pregnant females, under field conditions in Senegal. Antibody levels persisted up to 1 year after vaccination.
Collapse
Affiliation(s)
| | - Victor Mbao
- Global Alliance for Livestock Veterinary Medicines.
| | | | | | | | | | | |
Collapse
|
25
|
Keck F, Amaya M, Kehn-Hall K, Roberts B, Bailey C, Narayanan A. Characterizing the effect of Bortezomib on Rift Valley Fever Virus multiplication. Antiviral Res 2015; 120:48-56. [PMID: 26001632 DOI: 10.1016/j.antiviral.2015.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/02/2015] [Accepted: 05/14/2015] [Indexed: 12/13/2022]
Abstract
Rift Valley Fever Virus (RVFV) belongs to the family Bunyaviridae and is a known cause of epizootics and epidemics in Africa and the Middle East. With no FDA approved therapeutics available to treat RVFV infection, understanding the interactions between the virus and the infected host is crucial to developing novel therapeutic strategies. Here, we investigated the requirement of the ubiquitin-proteasome system (UPS) for the establishment of a productive RVFV infection. It was previously shown that the UPS plays a central role in RVFV multiplication involving degradation of PKR and p62 subunit of TFIIH. Using the FDA-approved proteasome inhibitor Bortezomib, we observed robust inhibition of intracellular and extracellular viral loads. Bortezomib treatment did not affect the nuclear/cytoplasmic distribution of the non-structural S-segment protein (NSs); however, the ability of NSs to form nuclear filaments was abolished as a result of Bortezomib treatment. In silico ubiquitination prediction analysis predicted that known NSs interactors (SAP30, YY1, and mSin3A) have multiple putative ubiquitination sites, while NSs itself was not predicted to be ubiquitinated. Immunoprecipitation studies indicated a decrease in interaction between SAP30 - NSs, and mSin3A - NSs in the context of Bortezomib treatment. This decrease in association between SAP30 - NSs also correlated with a decrease in the ubiquitination status of SAP30 with Bortezomib treatment. Bortezomib treatment, however, resulted in increased ubiquitination of mSin3A, suggesting that Bortezomib dynamically affects the ubiquitination status of host proteins that interact with NSs. Finally, we observed that expression of interferon beta (IFN-β) was increased in Bortezomib treated cells which indicated that the cellular antiviral mechanism was revived as a result of treatment and may contribute to control of viral multiplication.
Collapse
Affiliation(s)
- Forrest Keck
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA 20110, United States
| | - Moushimi Amaya
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA 20110, United States
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA 20110, United States
| | - Brian Roberts
- Leidos Health Life Sciences, 5202 Presidents Court, Suite 110, Frederick, MD 21703, United States
| | - Charles Bailey
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA 20110, United States
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA 20110, United States.
| |
Collapse
|
26
|
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: 5.3] [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
|
27
|
A ΩXaV motif in the Rift Valley fever virus NSs protein is essential for degrading p62, forming nuclear filaments and virulence. Proc Natl Acad Sci U S A 2015; 112:6021-6. [PMID: 25918396 DOI: 10.1073/pnas.1503688112] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a single-stranded RNA virus capable of inducing fatal hemorrhagic fever in humans. A key component of RVFV virulence is its ability to form nuclear filaments through interactions between the viral nonstructural protein NSs and the host general transcription factor TFIIH. Here, we identify an interaction between a ΩXaV motif in NSs and the p62 subunit of TFIIH. This motif in NSs is similar to ΩXaV motifs found in nucleotide excision repair (NER) factors and transcription factors known to interact with p62. Structural and biophysical studies demonstrate that NSs binds to p62 in a similar manner as these other factors. Functional studies in RVFV-infected cells show that the ΩXaV motif is required for both nuclear filament formation and degradation of p62. Consistent with the fact that the RVFV can be distinguished from other Bunyaviridae-family viruses due to its ability to form nuclear filaments in infected cells, the motif is absent in the NSs proteins of other Bunyaviridae-family viruses. Taken together, our studies demonstrate that p62 binding to NSs through the ΩXaV motif is essential for degrading p62, forming nuclear filaments and enhancing RVFV virulence. In addition, these results show how the RVFV incorporates a simple motif into the NSs protein that enables it to functionally mimic host cell proteins that bind the p62 subunit of TFIIH.
Collapse
|
28
|
Terasaki K, Makino S. Interplay between the Virus and Host in Rift Valley Fever Pathogenesis. J Innate Immun 2015; 7:450-8. [PMID: 25766761 DOI: 10.1159/000373924] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/08/2015] [Indexed: 12/22/2022] Open
Abstract
Rift Valley fever virus (RVFV) belongs to the genus Phlebovirus, family Bunyaviridae, and carries single-stranded tripartite RNA segments. The virus is transmitted by mosquitoes and has caused large outbreaks among ruminants and humans in sub-Saharan African and Middle East countries. The disease is characterized by a sudden onset of fever, headache, muscle pain, joint pain, photophobia, and weakness. In most cases, patients recover from the disease after a period of weeks, but some also develop retinal or macular changes, which result in vision impairment that lasts for an undefined period of time, and severe disease, characterized by hemorrhagic fever or encephalitis. The virus also causes febrile illness resulting in a high rate of spontaneous abortions in ruminants. The handling of wild-type RVFV requires high-containment facilities, including biosafety level 4 or enhanced biosafety level 3 laboratories. Nonetheless, studies clarifying the mechanisms of the RVFV-induced diseases and preventing them are areas of active research throughout the world. By primarily referring to recent studies using several animal model systems, protein expression systems, and specific mutant viruses, this review describes the current knowledge about the mechanisms of pathogenesis of RVF and biological functions of various viral proteins that affect RVFV pathogenicity.
Collapse
Affiliation(s)
- Kaori Terasaki
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Tex., USA
| | | |
Collapse
|
29
|
Tokuda S, Do Valle TZ, Batista L, Simon-Chazottes D, Guillemot L, Bouloy M, Flamand M, Montagutelli X, Panthier JJ. The genetic basis for susceptibility to Rift Valley fever disease in MBT/Pas mice. Genes Immun 2015; 16:206-12. [PMID: 25569261 DOI: 10.1038/gene.2014.79] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 11/09/2022]
Abstract
The large variation in individual response to infection with Rift Valley fever virus (RVFV) suggests that host genetic determinants play a role in determining virus-induced disease outcomes. These genetic factors are still unknown. The systemic inoculation of mice with RVFV reproduces major pathological features of severe human disease, notably the hepatitis and encephalitis. A genome scan performed on 546 (BALB/c × MBT) F2 progeny identified three quantitative trait loci (QTLs), denoted Rvfs-1 to Rvfs-3, that were associated with disease susceptibility in MBT/Pas mice. Non-parametric interval-mapping revealed one significant and two suggestive linkages with survival time on chromosomes 2 (Rvfs-1), 5 (Rvfs-3) and 11 (Rvfs-2) with respective logarithm of odds (LOD) scores of 4.58, 2.95 and 2.99. The two-part model, combining survival time and survival/death, identified one significant linkage to Rvfs-2 and one suggestive linkage to Rvfs-1 with respective LOD scores of 5.12 and 4.55. Under a multiple model, with additive effects and sex as a covariate, the three QTLs explained 8.3% of the phenotypic variance. Sex had the strongest influence on susceptibility. The contribution of Rvfs-1, Rvfs-2 and Rvfs-3 to survival time of RVFV-infected mice was further confirmed in congenic mice.
Collapse
Affiliation(s)
- S Tokuda
- 1] Institut Pasteur, Developmental & Stem Cell Biology Department, Mouse functional Genetics, Paris, France [2] Centre National de la Recherche Scientifique, URA 2578, Paris, France
| | - T Z Do Valle
- 1] Institut Pasteur, Developmental & Stem Cell Biology Department, Mouse functional Genetics, Paris, France [2] Centre National de la Recherche Scientifique, URA 2578, Paris, France [3] Instituto Oswaldo Cruz, Laboratório de Imunomodulação e Protozoologia, Fiocruz, Rio de Janeiro, Brasil
| | - L Batista
- 1] Institut Pasteur, Developmental & Stem Cell Biology Department, Mouse functional Genetics, Paris, France [2] Centre National de la Recherche Scientifique, URA 2578, Paris, France [3] Sorbonne Universités, UPMC Univ Paris 06, IFD, Paris, France
| | - D Simon-Chazottes
- 1] Institut Pasteur, Developmental & Stem Cell Biology Department, Mouse functional Genetics, Paris, France [2] Centre National de la Recherche Scientifique, URA 2578, Paris, France
| | - L Guillemot
- 1] Institut Pasteur, Developmental & Stem Cell Biology Department, Mouse functional Genetics, Paris, France [2] Centre National de la Recherche Scientifique, URA 2578, Paris, France
| | - M Bouloy
- Institut Pasteur, Bunyaviruses Molecular Genetics, Paris, France
| | - M Flamand
- Institut Pasteur, Structural Virology, Paris, France
| | - X Montagutelli
- 1] Institut Pasteur, Developmental & Stem Cell Biology Department, Mouse functional Genetics, Paris, France [2] Centre National de la Recherche Scientifique, URA 2578, Paris, France
| | - J-J Panthier
- 1] Institut Pasteur, Developmental & Stem Cell Biology Department, Mouse functional Genetics, Paris, France [2] Centre National de la Recherche Scientifique, URA 2578, Paris, France
| |
Collapse
|
30
|
Kortekaas J, Oreshkova N, van Keulen L, Kant J, Bosch B, Bouloy M, Moulin V, Goovaerts D, Moormann R. Comparative efficacy of two next-generation Rift Valley fever vaccines. Vaccine 2014; 32:4901-8. [DOI: 10.1016/j.vaccine.2014.07.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 06/11/2014] [Accepted: 07/09/2014] [Indexed: 12/17/2022]
|
31
|
Chandler JA, Thongsripong P, Green A, Kittayapong P, Wilcox BA, Schroth GP, Kapan DD, Bennett SN. Metagenomic shotgun sequencing of a Bunyavirus in wild-caught Aedes aegypti from Thailand informs the evolutionary and genomic history of the Phleboviruses. Virology 2014; 464-465:312-319. [PMID: 25108381 DOI: 10.1016/j.virol.2014.06.036] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 06/10/2014] [Accepted: 06/22/2014] [Indexed: 01/17/2023]
Abstract
Arthropod-borne viruses significantly impact human health. They span multiple families, all of which include viruses not known to cause disease. Characterizing these representatives could provide insights into the origins of their disease-causing counterparts. Field-caught Aedes aegypti mosquitoes from Nakhon Nayok, Thailand, underwent metagenomic shotgun sequencing to reveal a Bunyavirus closely related to Phasi Charoen (PhaV) virus, isolated in 2009 from Ae. aegypti near Bangkok. Phylogenetic analysis of this virus suggests it is basal to the Phlebovirus genus thus making it ideally positioned phylogenetically for understanding the evolution of these clinically important viruses. Genomic analysis finds that a gene necessary for virulence in vertebrates, but not essential for viral replication in arthropods, is missing. The sequencing of this phylogenetically-notable and genomically-unique Phlebovirus from wild mosquitoes exemplifies the utility and efficacy of metagenomic shotgun sequencing for virus characterization in arthropod vectors of human diseases.
Collapse
Affiliation(s)
- James Angus Chandler
- Department of Microbiology, California Academy of Sciences, 55 Music Concourse Drive, Golden Gate Park, San Francisco, CA 94118, United States of America.
| | - Panpim Thongsripong
- Department of Microbiology, California Academy of Sciences, 55 Music Concourse Drive, Golden Gate Park, San Francisco, CA 94118, United States of America; Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawai׳i at Manoa, 615 Ilalo Street, BioScience Building, Suite 320, Honolulu, HI 96813, United States of America.
| | - Amy Green
- Department of Microbiology, University of Hawai׳i at Manoa, 2538 McCarthy Mall, Snyder 207, Honolulu, HI 96822, United States of America.
| | - Pattamaporn Kittayapong
- Center of Excellence for Vectors and Vector-Borne Diseases, Faculty of Science, Mahidol University at Salaya, 999 Phuttamonthon 4, Road Nakhon Pathom 73170, Thailand.
| | - Bruce A Wilcox
- Center of Excellence for Vectors and Vector-Borne Diseases, Faculty of Science, Mahidol University at Salaya, 999 Phuttamonthon 4, Road Nakhon Pathom 73170, Thailand; Integrative Research and Education Program, Faculty of Public Health, Mahidol University, 420/1 Ratchawithi Road, Ratchathewi District, Bangkok 10400, Thailand.
| | - Gary P Schroth
- Illumina Inc, 25861 Industrial Blvd, Hayward, CA 94545, United States of America.
| | - Durrell D Kapan
- Department of Entomology and Center for Comparative Genomics, California Academy of Sciences, 55 Music Concourse Drive, Golden Gate Park, San Francisco, CA 94118, United States of America; Center for Conservation Research Training, Pacific Biosciences Research Center, University of Hawai׳i at Manoa, 3050 Maile Way, Gilmore Hall 406, Honolulu, HI 96822, United States of America.
| | - Shannon N Bennett
- Department of Microbiology, California Academy of Sciences, 55 Music Concourse Drive, Golden Gate Park, San Francisco, CA 94118, United States of America.
| |
Collapse
|
32
|
Barry G, Varela M, Ratinier M, Blomström AL, Caporale M, Seehusen F, Hahn K, Schnettler E, Baumgärtner W, Kohl A, Palmarini M. NSs protein of Schmallenberg virus counteracts the antiviral response of the cell by inhibiting its transcriptional machinery. J Gen Virol 2014; 95:1640-1646. [PMID: 24828331 PMCID: PMC4103064 DOI: 10.1099/vir.0.065425-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/09/2014] [Indexed: 01/02/2023] Open
Abstract
Bunyaviruses have evolved a variety of strategies to counteract the antiviral defence systems of mammalian cells. Here we show that the NSs protein of Schmallenberg virus (SBV) induces the degradation of the RPB1 subunit of RNA polymerase II and consequently inhibits global cellular protein synthesis and the antiviral response. In addition, we show that the SBV NSs protein enhances apoptosis in vitro and possibly in vivo, suggesting that this protein could be involved in SBV pathogenesis in different ways.
Collapse
Affiliation(s)
- Gerald Barry
- MRC–University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Mariana Varela
- MRC–University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Maxime Ratinier
- MRC–University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Anne-Lie Blomström
- MRC–University of Glasgow Centre for Virus Research, Glasgow, UK
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, P.O. Box 7028, Uppsala SE-750 07, Sweden
| | - Marco Caporale
- MRC–University of Glasgow Centre for Virus Research, Glasgow, UK
- Istituto Zooprofilattico Sperimentale dell’Abruzzo e Molise ‘G. Caporale’, Teramo, Italy
| | - Frauke Seehusen
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Kerstin Hahn
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | | | - Wolfgang Baumgärtner
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Alain Kohl
- MRC–University of Glasgow Centre for Virus Research, Glasgow, UK
| | | |
Collapse
|
33
|
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: 3.7] [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]
|
34
|
Margaria P, Bosco L, Vallino M, Ciuffo M, Mautino GC, Tavella L, Turina M. The NSs protein of tomato spotted wilt virus is required for persistent infection and transmission by Frankliniella occidentalis. J Virol 2014; 88:5788-802. [PMID: 24623427 PMCID: PMC4019118 DOI: 10.1128/jvi.00079-14] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/04/2014] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Tomato spotted wilt virus (TSWV) is the type member of tospoviruses (genus Tospovirus), plant-infecting viruses that cause severe damage to ornamental and vegetable crops. Tospoviruses are transmitted by thrips in the circulative propagative mode. We generated a collection of NSs-defective TSWV isolates and showed that TSWV coding for truncated NSs protein could not be transmitted by Frankliniella occidentalis. Quantitative reverse transcription (RT)-PCR and immunostaining of individual insects detected the mutant virus in second-instar larvae and adult insects, demonstrating that insects could acquire and accumulate the NSs-defective virus. Nevertheless, adults carried a significantly lower viral load, resulting in the absence of transmission. Genome sequencing and analyses of reassortant isolates showed genetic evidence of the association between the loss of competence in transmission and the mutation in the NSs coding sequence. Our findings offer new insight into the TSWV-thrips interaction and Tospovirus pathogenesis and highlight, for the first time in the Bunyaviridae family, a major role for the S segment, and specifically for the NSs protein, in virulence and efficient infection in insect vector individuals. IMPORTANCE Our work is the first to show a role for the NSs protein in virus accumulation in the insect vector in the Bunyaviridae family: demonstration was obtained for the system TSWV-F. occidentalis, arguably one of the most damaging combination for vegetable crops. Genetic evidence of the involvement of the NSs protein in vector transmission was provided with multiple approaches.
Collapse
Affiliation(s)
- P. Margaria
- Istituto di Virologia Vegetale, Sez. di Torino, CNR, Turin, Italy
| | - L. Bosco
- Dipartimento di Scienze Agrarie, Forestali e Alimentari (DISAFA), University of Turin, Grugliasco (TO), Italy
| | - M. Vallino
- Istituto di Virologia Vegetale, Sez. di Torino, CNR, Turin, Italy
| | - M. Ciuffo
- Istituto di Virologia Vegetale, Sez. di Torino, CNR, Turin, Italy
| | - G. C. Mautino
- Dipartimento di Scienze Agrarie, Forestali e Alimentari (DISAFA), University of Turin, Grugliasco (TO), Italy
| | - L. Tavella
- Dipartimento di Scienze Agrarie, Forestali e Alimentari (DISAFA), University of Turin, Grugliasco (TO), Italy
| | - M. Turina
- Istituto di Virologia Vegetale, Sez. di Torino, CNR, Turin, Italy
| |
Collapse
|
35
|
Murakami S, Terasaki K, Ramirez SI, Morrill JC, Makino S. Development of a novel, single-cycle replicable rift valley Fever vaccine. PLoS Negl Trop Dis 2014; 8:e2746. [PMID: 24651859 PMCID: PMC3961198 DOI: 10.1371/journal.pntd.0002746] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/30/2014] [Indexed: 12/25/2022] Open
Abstract
Rift Valley fever virus (RVFV) (genus Phlebovirus, family Bunyaviridae) is an arbovirus that causes severe disease in humans and livestock in sub-Saharan African countries. Although the MP-12 strain of RVFV is a live attenuated vaccine candidate, neuroinvasiveness and neurovirulence of MP-12 in mice may be a concern when vaccinating certain individuals, especially those that are immunocompromised. We have developed a novel, single-cycle replicable MP-12 (scMP-12), which carries an L RNA, M RNA mutant encoding a mutant envelope protein lacking an endoplasmic reticulum retrieval signal and defective for membrane fusion function, and S RNA encoding N protein and green fluorescent protein. The scMP-12 underwent efficient amplification, then formed plaques and retained the introduced mutation after serial passages in a cell line stably expressing viral envelope proteins. However, inoculation of the scMP-12 into naïve cells resulted in a single round of viral replication, and production of low levels of noninfectious virus-like particles. Intracranial inoculation of scMP-12 into suckling mice did not cause clinical signs or death, a finding which demonstrated that the scMP-12 lacked neurovirulence. Mice immunized with a single dose of scMP-12 produced neutralizing antibodies, whose titers were higher than in mice immunized with replicon particles carrying L RNA and S RNA encoding N protein and green fluorescent protein. Moreover, 90% of the scMP-12-immunized mice were protected from wild-type RVFV challenge by efficiently suppressing viremia and replication of the challenge virus in the liver and the spleen. These data demonstrated that scMP-12 is a safe and immunogenic RVFV vaccine candidate.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Disease Models, Animal
- Female
- Mice
- Mutant Proteins/genetics
- Mutant Proteins/metabolism
- Rift Valley Fever/prevention & control
- Rift Valley fever virus/genetics
- Rift Valley fever virus/immunology
- Rift Valley fever virus/physiology
- Survival Analysis
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/adverse effects
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/adverse effects
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Virus Internalization
- Virus Replication
Collapse
Affiliation(s)
- Shin Murakami
- Department of Microbiology and Immunology, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kaori Terasaki
- Department of Microbiology and Immunology, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sydney I. Ramirez
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - John C. Morrill
- Department of Microbiology and Immunology, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Shinji Makino
- Department of Microbiology and Immunology, 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
- UTMB Center for Tropical Diseases, 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
- * E-mail:
| |
Collapse
|
36
|
Palacios G, Tesh RB, Savji N, Travassos da Rosa APA, Guzman H, Bussetti AV, Desai A, Ladner J, Sanchez-Seco M, Lipkin WI. Characterization of the Sandfly fever Naples species complex and description of a new Karimabad species complex (genus Phlebovirus, family Bunyaviridae). J Gen Virol 2014; 95:292-300. [PMID: 24096318 PMCID: PMC3917069 DOI: 10.1099/vir.0.056614-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 09/23/2013] [Indexed: 12/24/2022] Open
Abstract
Genomic and antigenic characterization of members of the Sandfly fever Naples virus (SFNV) complex reveals the presence of five clades that differ in their geographical distribution. Saint Floris and Gordil viruses, both found in Africa, form one clade; Punique, Granada and Massilia viruses, all isolated in the western Mediterranean, constitute a second; Toscana virus, a third; SFNV isolates from Italy, Cyprus, Egypt and India form a fourth; while Tehran virus and a Serbian isolate Yu 8/76, represent a fifth. Interestingly, this last clade appears not to express the second non-structural protein ORF. Karimabad virus, previously classified as a member of the SFNV complex, and Gabek Forest virus are distinct and form a new species complex (named Karimabad) in the Phlebovirus genus. In contrast with the high reassortment frequency observed in some South American phleboviruses, the only virus of the SFNV complex with evidence of reassortment was Granada virus.
Collapse
Affiliation(s)
- Gustavo Palacios
- Center for Genomic Sciences, United States Army Medical Research Institute for Infectious Diseases, Frederick, MD, USA
| | - Robert B. Tesh
- Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nazir Savji
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Amelia P. A. Travassos da Rosa
- Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hilda Guzman
- Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ana Valeria Bussetti
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Aaloki Desai
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jason Ladner
- Center for Genomic Sciences, United States Army Medical Research Institute for Infectious Diseases, Frederick, MD, USA
| | - Maripaz Sanchez-Seco
- Centro Nacional de Microbiologia, Instituto de Salud ‘Carlos III’, Madrid, Spain
| | - W. Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
37
|
Faburay B, Wilson W, McVey DS, Drolet BS, Weingartl H, Madden D, Young A, Ma W, Richt JA. Rift Valley fever virus structural and nonstructural proteins: recombinant protein expression and immunoreactivity against antisera from sheep. Vector Borne Zoonotic Dis 2013; 13:619-29. [PMID: 23962238 DOI: 10.1089/vbz.2012.1285] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The Rift Valley fever virus (RVFV) encodes the structural proteins nucleoprotein (N), aminoterminal glycoprotein (Gn), carboxyterminal glycoprotein (Gc), and L protein, 78-kD, and the nonstructural proteins NSm and NSs. Using the baculovirus system, we expressed the full-length coding sequence of N, NSs, NSm, Gc, and the ectodomain of the coding sequence of the Gn glycoprotein derived from the virulent strain of RVFV ZH548. Western blot analysis using anti-His antibodies and monoclonal antibodies against Gn and N confirmed expression of the recombinant proteins, and in vitro biochemical analysis showed that the two glycoproteins, Gn and Gc, were expressed in glycosylated form. Immunoreactivity profiles of the recombinant proteins in western blot and in indirect enzyme-linked immunosorbent assay against a panel of antisera obtained from vaccinated or wild type (RVFV)-challenged sheep confirmed the results obtained with anti-His antibodies and demonstrated the suitability of the baculo-expressed antigens for diagnostic assays. In addition, these recombinant proteins could be valuable for the development of diagnostic methods that differentiate infected from vaccinated animals (DIVA).
Collapse
Affiliation(s)
- Bonto Faburay
- 1 Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University , Manhattan, Kansas
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
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: 3.2] [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
|
39
|
Palacios G, Savji N, Travassos da Rosa A, Guzman H, Yu X, Desai A, Rosen GE, Hutchison S, Lipkin WI, Tesh R. Characterization of the Uukuniemi virus group (Phlebovirus: Bunyaviridae): evidence for seven distinct species. J Virol 2013; 87:3187-95. [PMID: 23283959 PMCID: PMC3592153 DOI: 10.1128/jvi.02719-12] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/24/2012] [Indexed: 11/20/2022] Open
Abstract
Evolutionary insights into the phleboviruses are limited because of an imprecise classification scheme based on partial nucleotide sequences and scattered antigenic relationships. In this report, the serologic and phylogenetic relationships of the Uukuniemi group viruses and their relationships with other recently characterized tick-borne phleboviruses are described using full-length genome sequences. We propose that the viruses currently included in the Uukuniemi virus group be assigned to five different species as follows: Uukuniemi virus, EgAn 1825-61 virus, Fin V707 virus, Chizé virus, and Zaliv Terpenia virus would be classified into the Uukuniemi species; Murre virus, RML-105-105355 virus, and Sunday Canyon virus would be classified into a Murre virus species; and Grand Arbaud virus, Precarious Point virus, and Manawa virus would each be given individual species status. Although limited sequence similarity was detected between current members of the Uukuniemi group and Severe fever with thrombocytopenia syndrome virus (SFTSV) and Heartland virus, a clear serological reaction was observed between some of them, indicating that SFTSV and Heartland virus should be considered part of the Uukuniemi virus group. Moreover, based on the genomic diversity of the phleboviruses and given the low correlation observed between complement fixation titers and genetic distance, we propose a system for classification of the Bunyaviridae based on genetic as well as serological data. Finally, the recent descriptions of SFTSV and Heartland virus also indicate that the public health importance of the Uukuniemi group viruses must be reevaluated.
Collapse
Affiliation(s)
- Gustavo Palacios
- United States Army Medical Research Institute for Infectious Diseases, Frederick, MD, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Varela M, Schnettler E, Caporale M, Murgia C, Barry G, McFarlane M, McGregor E, Piras IM, Shaw A, Lamm C, Janowicz A, Beer M, Glass M, Herder V, Hahn K, Baumgärtner W, Kohl A, Palmarini M. Schmallenberg virus pathogenesis, tropism and interaction with the innate immune system of the host. PLoS Pathog 2013; 9:e1003133. [PMID: 23326235 PMCID: PMC3542112 DOI: 10.1371/journal.ppat.1003133] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 12/01/2012] [Indexed: 12/27/2022] Open
Abstract
Schmallenberg virus (SBV) is an emerging orthobunyavirus of ruminants associated with outbreaks of congenital malformations in aborted and stillborn animals. Since its discovery in November 2011, SBV has spread very rapidly to many European countries. Here, we developed molecular and serological tools, and an experimental in vivo model as a platform to study SBV pathogenesis, tropism and virus-host cell interactions. Using a synthetic biology approach, we developed a reverse genetics system for the rapid rescue and genetic manipulation of SBV. We showed that SBV has a wide tropism in cell culture and "synthetic" SBV replicates in vitro as efficiently as wild type virus. We developed an experimental mouse model to study SBV infection and showed that this virus replicates abundantly in neurons where it causes cerebral malacia and vacuolation of the cerebral cortex. These virus-induced acute lesions are useful in understanding the progression from vacuolation to porencephaly and extensive tissue destruction, often observed in aborted lambs and calves in naturally occurring Schmallenberg cases. Indeed, we detected high levels of SBV antigens in the neurons of the gray matter of brain and spinal cord of naturally affected lambs and calves, suggesting that muscular hypoplasia observed in SBV-infected lambs is mostly secondary to central nervous system damage. Finally, we investigated the molecular determinants of SBV virulence. Interestingly, we found a biological SBV clone that after passage in cell culture displays increased virulence in mice. We also found that a SBV deletion mutant of the non-structural NSs protein (SBVΔNSs) is less virulent in mice than wild type SBV. Attenuation of SBV virulence depends on the inability of SBVΔNSs to block IFN synthesis in virus infected cells. In conclusion, this work provides a useful experimental framework to study the biology and pathogenesis of SBV.
Collapse
Affiliation(s)
- Mariana Varela
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Esther Schnettler
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marco Caporale
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Istituto G. Caporale, Teramo, Italy
| | - Claudio Murgia
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gerald Barry
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Melanie McFarlane
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Eva McGregor
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ilaria M. Piras
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Sassari, Italy
| | - Andrew Shaw
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Catherine Lamm
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anna Janowicz
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Mandy Glass
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Vanessa Herder
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Kerstin Hahn
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Alain Kohl
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Massimo Palmarini
- MRC Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
41
|
Dicer-2- and Piwi-mediated RNA interference in Rift Valley fever virus-infected mosquito cells. J Virol 2012; 87:1631-48. [PMID: 23175368 DOI: 10.1128/jvi.02795-12] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a Phlebovirus (Bunyaviridae family) transmitted by mosquitoes. It infects humans and ruminants, causing dramatic epidemics and epizootics in Africa, Yemen, and Saudi Arabia. While recent studies demonstrated the importance of the nonstructural protein NSs as a major component of virulence in vertebrates, little is known about infection of mosquito vectors. Here we studied RVFV infection in three different mosquito cell lines, Aag2 cells from Aedes aegypti and U4.4 and C6/36 cells from Aedes albopictus. In contrast with mammalian cells, where NSs forms nuclear filaments, U4.4 and Aag2 cells downregulated NSs expression such that NSs filaments were never formed in nuclei of U4.4 cells and disappeared at an early time postinfection in the case of Aag2 cells. On the contrary, in C6/36 cells, NSs nuclear filaments were visible during the entire time course of infection. Analysis of virus-derived small interfering RNAs (viRNAs) by deep sequencing indicated that production of viRNAs was very low in C6/36 cells, which are known to be Dicer-2 deficient but expressed some viRNAs presenting a Piwi signature. In contrast, Aag2 and U4.4 cells produced large amounts of viRNAs predominantly matching the S segment and displaying Dicer-2 and Piwi signatures. Whereas 21-nucleotide (nt) Dicer-2 viRNAs were prominent during early infection, the population of 24- to 27-nt Piwi RNAs (piRNAs) increased progressively and became predominant later during the acute infection and during persistence. In Aag2 and U4.4 cells, the combined actions of the Dicer-2 and Piwi pathways triggered an efficient antiviral response permitting, among other actions, suppression of NSs filament formation and allowing establishment of persistence. In C6/36 cells, Piwi-mediated RNA interference (RNAi) appeared to be sufficient to mount an antiviral response against a secondary infection with a superinfecting virus. This study provides new insights into the role of Dicer and Piwi in mosquito antiviral defense and the development of the antiviral response in mosquitoes.
Collapse
|
42
|
Lam TTY, Liu W, Bowden TA, Cui N, Zhuang L, Liu K, Zhang YY, Cao WC, Pybus OG. Evolutionary and molecular analysis of the emergent severe fever with thrombocytopenia syndrome virus. Epidemics 2012; 5:1-10. [PMID: 23438426 PMCID: PMC4330987 DOI: 10.1016/j.epidem.2012.09.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 09/13/2012] [Indexed: 01/08/2023] Open
Abstract
In 2009, a novel Bunyavirus, called severe fever with thrombocytopenia syndrome virus (SFTSV) was identified in the vicinity of Huaiyangshan, China. Clinical symptoms of this zoonotic virus included severe fever, thrombocytopenia, and leukocytopenia, with a mortality rate of ∼10%. By the end of 2011 the disease associated with this pathogen had been reported from eleven Chinese provinces and human-to-human transmission suspected. However, current understanding of the evolution and molecular epidemiology of SFTSV before and after its identification is limited. To address this we undertake phylogenetic, evolutionary and structural analyses of all available SFTSV genetic sequences, including a new SFTSV complete genome isolated from a patient from Henan in 2011. Our discovery of a mosaic L segment sequence, which is descended from two major circulating lineages of SFTSV in China, represents the first evidence that homologous recombination plays a role in SFTSV evolution. Selection analyses indicate that negative selection is predominant in SFTSV genes, yet differences in selective forces among genes are consistent between Phlebovirus species. Further analysis reveals structural conservation between SFTSV and Rift Valley fever virus in the residues of their nucleocapsids that are responsible for oligomerisation and RNA-binding, suggesting the viruses share similar modes of higher-order assembly. We reconstruct the epidemic history of SFTSV using molecular clock and coalescent-based methods, revealing that the extant SFTSV lineages originated 50–150 years ago, and that the viral population experienced a recent growth phase that concurs with and extends the earliest serological reports of SFTSV infection. Taken together, our combined structural and phylogenetic analyses shed light into the evolutionary behaviour of SFTSV in the context of other, better-known, pathogenic Phleboviruses.
Collapse
|
43
|
Ayari-Fakhfakh E, do Valle TZ, Guillemot L, Panthier JJ, Bouloy M, Ghram A, Albina E, Cêtre-Sossah C. MBT/Pas mouse: a relevant model for the evaluation of Rift Valley fever vaccines. J Gen Virol 2012; 93:1456-1464. [DOI: 10.1099/vir.0.042754-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Currently, there are no worldwide licensed vaccines for Rift Valley fever (RVF) that are both safe and effective. Development and evaluation of vaccines, diagnostics and treatments depend on the availability of appropriate animal models. Animal models are also necessary to understand the basic pathobiology of infection. Here, we report the use of an inbred MBT/Pas mouse model that consistently reproduces RVF disease and serves our purpose for testing the efficacy of vaccine candidates; an attenuated Rift Valley fever virus (RVFV) and a recombinant RVFV–capripoxvirus. We show that this model is relevant for vaccine testing.
Collapse
Affiliation(s)
- Emna Ayari-Fakhfakh
- Institut Pasteur de Tunis, Université de Tunis El Manar, 1002 Tunis Belvédère, Tunisia
| | - Tânia Zaverucha do Valle
- Centre National de la Recherche Scientifique, Unité de Recherche Associée 2578, Paris, France
- Mouse Functional Genetics, Institut Pasteur, Paris, France
| | - Laurent Guillemot
- Centre National de la Recherche Scientifique, Unité de Recherche Associée 2578, Paris, France
- Mouse Functional Genetics, Institut Pasteur, Paris, France
| | - Jean-Jacques Panthier
- Centre National de la Recherche Scientifique, Unité de Recherche Associée 2578, Paris, France
- Mouse Functional Genetics, Institut Pasteur, Paris, France
| | - Michèle Bouloy
- Molecular Genetics of Bunyaviruses, Institut Pasteur, Paris, France
| | - Abdeljelil Ghram
- Institut Pasteur de Tunis, Université de Tunis El Manar, 1002 Tunis Belvédère, Tunisia
| | - Emmanuel Albina
- CIRAD, UMR Contrôle des Maladies, Montpellier, F-34398, France
| | | |
Collapse
|
44
|
Ikegami T. Molecular biology and genetic diversity of Rift Valley fever virus. Antiviral Res 2012; 95:293-310. [PMID: 22710362 DOI: 10.1016/j.antiviral.2012.06.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 01/05/2012] [Accepted: 06/05/2012] [Indexed: 01/25/2023]
Abstract
Rift Valley fever virus (RVFV), a member of the family Bunyaviridae, genus Phlebovirus, is the causative agent of Rift Valley fever (RVF), a mosquito-borne disease of ruminant animals and humans. The generation of a large sequence database has facilitated studies of the evolution and spread of the virus. Bayesian analyses indicate that currently circulating strains of RVFV are descended from an ancestral species that emerged from a natural reservoir in Africa when large-scale cattle and sheep farming were introduced during the 19th century. Viruses descended from multiple lineages persist in that region, through infection of reservoir animals and vertical transmission in mosquitoes, emerging in years of heavy rainfall to cause epizootics and epidemics. On a number of occasions, viruses from these lineages have been transported outside the enzootic region through the movement of infected animals or mosquitoes, triggering outbreaks in countries such as Egypt, Saudi Arabia, Mauritania and Madagascar, where RVF had not previously been seen. Such viruses could potentially become established in their new environments through infection of wild and domestic ruminants and other animals and vertical transmission in local mosquito species. Despite their extensive geographic dispersion, all strains of RVFV remain closely related at the nucleotide and amino acid level. The high degree of conservation of genes encoding the virion surface glycoproteins suggests that a single vaccine should protect against all currently circulating RVFV strains. Similarly, preservation of the sequence of the RNA-dependent RNA polymerase across viral lineages implies that antiviral drugs targeting the enzyme should be effective against all strains. Researchers should be encouraged to collect additional RVFV isolates and perform whole-genome sequencing and phylogenetic analysis, so as to enhance our understanding of the continuing evolution of this important virus. This review forms part of a series of invited papers in Antiviral Research on the genetic diversity of emerging viruses.
Collapse
Affiliation(s)
- Tetsuro Ikegami
- Department of Pathology, The University of Texas Medical Branch, MMNP3.206D, 301 University Blvd. Galveston, TX 77555-0436, USA.
| |
Collapse
|
45
|
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: 1.5] [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
|
46
|
Austin D, Baer A, Lundberg L, Shafagati N, Schoonmaker A, Narayanan A, Popova T, Panthier JJ, Kashanchi F, Bailey C, Kehn-Hall K. p53 Activation following Rift Valley fever virus infection contributes to cell death and viral production. PLoS One 2012; 7:e36327. [PMID: 22574148 PMCID: PMC3344861 DOI: 10.1371/journal.pone.0036327] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 03/30/2012] [Indexed: 12/31/2022] Open
Abstract
Rift Valley fever virus (RVFV) is an emerging viral zoonosis that is responsible for devastating outbreaks among livestock and is capable of causing potentially fatal disease in humans. Studies have shown that upon infection, certain viruses have the capability of utilizing particular cellular signaling pathways to propagate viral infection. Activation of p53 is important for the DNA damage signaling cascade, initiation of apoptosis, cell cycle arrest and transcriptional regulation of multiple genes. The current study focuses on the role of p53 signaling in RVFV infection and viral replication. These results show an up-regulation of p53 phosphorylation at several serine sites after RVFV MP-12 infection that is highly dependent on the viral protein NSs. qRT-PCR data showed a transcriptional up-regulation of several p53 targeted genes involved in cell cycle and apoptosis regulation following RVFV infection. Cell viability assays demonstrate that loss of p53 results in less RVFV induced cell death. Furthermore, decreased viral titers in p53 null cells indicate that RVFV utilizes p53 to enhance viral production. Collectively, these experiments indicate that the p53 signaling pathway is utilized during RVFV infection to induce cell death and increase viral production.
Collapse
Affiliation(s)
- Dana Austin
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Alan Baer
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Lindsay Lundberg
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Nazly Shafagati
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Annalise Schoonmaker
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Taissia Popova
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | | | - Fatah Kashanchi
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Charles Bailey
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- * E-mail:
| |
Collapse
|
47
|
Grobbelaar AA, Weyer J, Leman PA, Kemp A, Paweska JT, Swanepoel R. Molecular epidemiology of Rift Valley fever virus. Emerg Infect Dis 2012; 17:2270-6. [PMID: 22172568 PMCID: PMC3311189 DOI: 10.3201/eid1712.111035] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Large-scale vaccination of animals might have influenced virus evolution. Phylogenetic relationships were examined for 198 Rift Valley fever virus isolates and 5 derived strains obtained from various sources in Saudi Arabia and 16 countries in Africa during a 67-year period (1944–2010). A maximum-likelihood tree prepared with sequence data for a 490-nt section of the Gn glycoprotein gene showed that 95 unique sequences sorted into 15 lineages. A 2010 isolate from a patient in South Africa potentially exposed to co-infection with live animal vaccine and wild virus was a reassortant. The potential influence of large-scale use of live animal vaccine on evolution of Rift Valley fever virus is discussed.
Collapse
Affiliation(s)
- Antoinette A Grobbelaar
- National Institute for Communicable Diseases of the National Health Service, Sandringham, South Africa
| | | | | | | | | | | |
Collapse
|
48
|
Single-dose immunization with virus replicon particles confers rapid robust protection against Rift Valley fever virus challenge. J Virol 2012; 86:4204-12. [PMID: 22345465 DOI: 10.1128/jvi.07104-11] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Rift Valley fever virus (RVFV) causes outbreaks of severe disease in people and livestock throughout Africa and the Arabian Peninsula. The potential for RVFV introduction outside the area of endemicity highlights the need for fast-acting, safe, and efficacious vaccines. Here, we demonstrate a robust system for the reverse genetics generation of a RVF virus replicon particle (VRP(RVF)) vaccine candidate. Using a mouse model, we show that VRP(RVF) immunization provides the optimal balance of safety and single-dose robust efficacy. VRP(RVF) can actively synthesize viral RNA and proteins but lacks structural glycoprotein genes, preventing spread within immunized individuals and reducing the risk of vaccine-induced pathogenicity. VRP(RVF) proved to be completely safe following intracranial inoculation of suckling mice, a stringent test of vaccine safety. Single-dose subcutaneous immunization with VRP(RVF), although it is highly attenuated, completely protected mice against a virulent RVFV challenge dose which was 100,000-fold greater than the 50% lethal dose (LD(50)). Robust protection from lethal challenge was observed by 24 h postvaccination, with 100% protection induced in as little as 96 h. We show that a single subcutaneous VRP(RVF) immunization initiated a systemic antiviral state followed by an enhanced adaptive response. These data contrast sharply with the much-reduced survivability and immune responses observed among animals immunized with nonreplicating viral particles, indicating that replication, even if confined to the initially infected cells, contributes substantially to protective efficacy at early and late time points postimmunization. These data demonstrate that replicon vaccines successfully bridge the gap between safety and efficacy and provide insights into the kinetics of antiviral protection from RVFV infection.
Collapse
|
49
|
Baer A, Austin D, Narayanan A, Popova T, Kainulainen M, Bailey C, Kashanchi F, Weber F, Kehn-Hall K. Induction of DNA damage signaling upon Rift Valley fever virus infection results in cell cycle arrest and increased viral replication. J Biol Chem 2012; 287:7399-410. [PMID: 22223653 DOI: 10.1074/jbc.m111.296608] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Rift Valley fever virus (RVFV) is a highly pathogenic arthropod-borne virus infecting a wide range of vertebrate hosts. Of particular interest is the nonstructural NSs protein, which forms large filamentous fibril bundles in the nucleus. Past studies have shown NSs to be a multifaceted protein important for virulence through modulation of the interferon response as well acting as a general inhibitor of transcription. Here we investigated the regulation of the DNA damage signaling cascades by RVFV infection and found virally inducted phosphorylation of the classical DNA damage signaling proteins, ataxia-telangiectasia mutated (ATM) (Ser-1981), Chk.2 (Thr-68), H2A.X (Ser-139), and p53 (Ser-15). In contrast, ataxia-telangiectasia mutated and Rad3-related kinase (ATR) (Ser-428) phosphorylation was decreased following RVFV infection. Importantly, both the attenuated vaccine strain MP12 and the fully virulent strain ZH548 showed strong parallels in their up-regulation of the ATM arm of the DNA damage response and in the down-regulation of the ATR pathway. The increase in DNA damage signaling proteins did not result from gross DNA damage as no increase in DNA damage was observed following infection. Rather the DNA damage signaling was found to be dependent on the viral protein NSs, as an NSs mutant virus was not found to induce the equivalent signaling pathways. RVFV MP12-infected cells also displayed an S phase arrest that was found to be dependent on NSs expression. Use of ATM and Chk.2 inhibitors resulted in a marked decrease in S phase arrest as well as viral production. These results indicate that RVFV NSs induces DNA damage signaling pathways that are beneficial for viral replication.
Collapse
Affiliation(s)
- Alan Baer
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia 20110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Scott T, Paweska JT, Arbuthnot P, Weinberg MS. Pathogenic effects of Rift Valley fever virus NSs gene are alleviated in cultured cells by expressed antiviral short hairpin RNAs. Antivir Ther 2012; 17:643-56. [DOI: 10.3851/imp2073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2011] [Indexed: 10/28/2022]
|