1
|
Haas G, Lee B. Reverse Genetics Systems for the De Novo Rescue of Diverse Members of Paramyxoviridae. Methods Mol Biol 2024; 2733:15-35. [PMID: 38064024 DOI: 10.1007/978-1-0716-3533-9_2] [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] [Indexed: 12/18/2023]
Abstract
Paramyxoviruses place significant burdens on both human and wildlife health; while some paramyxoviruses are established within human populations, others circulate within diverse animal reservoirs. Concerningly, bat-borne paramyxoviruses have spilled over into humans with increasing frequency in recent years, resulting in severe disease. The risk of future zoonotic outbreaks, as well as the persistence of paramyxoviruses that currently circulate within humans, highlights the need for efficient tools through which to interrogate paramyxovirus biology. Reverse genetics systems provide scientists with the ability to rescue paramyxoviruses de novo, offering versatile tools for implementation in both research and public health settings. Reverse genetics systems have greatly improved over the past 30 years, with several key innovations optimizing the success of paramyxovirus rescue. Here, we describe the significance of such advances and provide a generally applicable guide for the development and use of reverse genetics systems for the rescue of diverse members of Paramyxoviridae.
Collapse
Affiliation(s)
- Griffin Haas
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benhur Lee
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Dong S, Meng W, Yang Z, Chen J, Liu J, Shen Z, Wang J. Development of a sensitive immunochromatographic method using lanthanide fluorescent microsphere for rapid test for PPRV antibody. J Virol Methods 2023; 321:114809. [PMID: 37683936 DOI: 10.1016/j.jviromet.2023.114809] [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: 07/29/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Peste des petits ruminants virus (PPRV) causes a very devastating disease in sheep and goats. Rapid diagnosis and immunisation have been identified as key strategies for successful prevention of the disease. Therefore, a sensitive fluorescent microsphere immunochromatography test strips (FM-ICTS) was developed for rapid detection of special antibodies of PPRV in goats and sheep serum. The FM-ICTS were successfully prepared by fluorescent microspheres (FM) as tracer, which were covalently coupled to PPRV nucleocapsid protein (NP). The NP and monoclonal antibody of NP were separately dispensed onto a nitrocellulose membrane as test and quality control lines, respectively. The critical threshold for determining negative or positive through the ratio of the fluorescent signal of the test line and the control line (T/C) is 0.050. The repeatability of the FM-ICTS was excellent, with an overall average CV of 3.17 %. The detection limit of this assay was 1:5120. Additionally, the FM-ICTS no cross reaction with the sera of other related diseases was observed, only reacting with anti-PPRV serum. 70 serum samples were tested by FM-ICTS and commercial ELISA kit, and the results showed good agreement. Overall, a promising pen-side diagnostic tool was developed for the rapid qualitatively/semi-quantitatively detection of PPRV antibodies within 15 min.
Collapse
Affiliation(s)
- Shuai Dong
- Shandong Binzhou Animal Science & Veterinary Medicine Academy, Bingzhou 256600, China; College of Life Science and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - Weiqin Meng
- Shandong Binzhou Animal Science & Veterinary Medicine Academy, Bingzhou 256600, China
| | - Zhe Yang
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - Jinlong Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Jianchai Liu
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - Zhiqiang Shen
- Shandong Binzhou Animal Science & Veterinary Medicine Academy, Bingzhou 256600, China
| | - Jinliang Wang
- Shandong Binzhou Animal Science & Veterinary Medicine Academy, Bingzhou 256600, China.
| |
Collapse
|
3
|
Tully M, Batten C, Ashby M, Mahapatra M, Parekh K, Parida S, Njeumi F, Willett B, Bataille A, Libeau G, Kwiatek O, Caron A, Berguido FJ, Lamien CE, Cattoli G, Misinzo G, Keyyu J, Mdetele D, Gakuya F, Bodjo SC, Taha FA, Elbashier HM, Khalafalla AI, Osman AY, Kock R. The evaluation of five serological assays in determining seroconversion to peste des petits ruminants virus in typical and atypical hosts. Sci Rep 2023; 13:14787. [PMID: 37684280 PMCID: PMC10491793 DOI: 10.1038/s41598-023-41630-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Peste des petits ruminants (PPR) is an infectious viral disease, primarily of small ruminants such as sheep and goats, but is also known to infect a wide range of wild and domestic Artiodactyls including African buffalo, gazelle, saiga and camels. The livestock-wildlife interface, where free-ranging animals can interact with captive flocks, is the subject of scrutiny as its role in the maintenance and spread of PPR virus (PPRV) is poorly understood. As seroconversion to PPRV indicates previous infection and/or vaccination, the availability of validated serological tools for use in both typical (sheep and goat) and atypical species is essential to support future disease surveillance and control strategies. The virus neutralisation test (VNT) and enzyme-linked immunosorbent assay (ELISA) have been validated using sera from typical host species. Still, the performance of these assays in detecting antibodies from atypical species remains unclear. We examined a large panel of sera (n = 793) from a range of species from multiple countries (sourced 2015-2022) using three tests: VNT, ID VET N-ELISA and AU-PANVAC H-ELISA. A sub-panel (n = 30) was also distributed to two laboratories and tested using the luciferase immunoprecipitation system (LIPS) and a pseudotyped virus neutralisation assay (PVNA). We demonstrate a 75.0-88.0% agreement of positive results for detecting PPRV antibodies in sera from typical species between the VNT and commercial ELISAs, however this decreased to 44.4-62.3% in sera from atypical species, with an inter-species variation. The LIPS and PVNA strongly correlate with the VNT and ELISAs for typical species but vary when testing sera from atypical species.
Collapse
Affiliation(s)
| | | | - Martin Ashby
- The Pirbright Institute, Pirbright, United Kingdom
| | | | | | - Satya Parida
- The Pirbright Institute, Pirbright, United Kingdom
- Food and Agriculture Organization (FAO), United Nations, Rome, Italy
| | - Felix Njeumi
- Food and Agriculture Organization (FAO), United Nations, Rome, Italy
| | - Brian Willett
- MRC-University of Glasgow Centre for Virus Research (UoG), Glasgow, United Kingdom
| | - Arnaud Bataille
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Genevieve Libeau
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Olivier Kwiatek
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Alexandre Caron
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Francisco J Berguido
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, 2444, Seibersdorf, Austria
| | - Charles E Lamien
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, 2444, Seibersdorf, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, 2444, Seibersdorf, Austria
| | - Gerald Misinzo
- SACIDS Foundation for One Health, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Julius Keyyu
- Tanzania Wildlife Research Institute (TAWIRI), Arusha, Tanzania
| | | | - Francis Gakuya
- Wildlife Research & Training Institute (WRTI), Karagita, Kenya
| | - Sanne Charles Bodjo
- Pan African Veterinary Vaccine Centre for African Union (AU-PANVAC), Debre Zeit, Ethiopia
| | | | | | - Abdelmalik Ibrahim Khalafalla
- Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi, United Arab Emirates
- Faculty of Veterinary Medicine, University of Khartoum, Khartoum, Sudan
| | - Abdinasir Y Osman
- National Institute of Health (NIH), Ministry of Health, Mogadishu, Somalia
- Royal Veterinary College (RVC), London, United Kingdom
| | - Richard Kock
- Royal Veterinary College (RVC), London, United Kingdom
| |
Collapse
|
4
|
Schmitz KS, Eblé PL, van Gennip RGP, Maris-Veldhuis MA, de Vries RD, van Keulen LJM, de Swart RL, van Rijn PA. Pathogenesis of wild-type- and vaccine-based recombinant peste des petits ruminants virus (PPRV) expressing EGFP in experimentally infected domestic goats. J Gen Virol 2023; 104. [PMID: 36757863 DOI: 10.1099/jgv.0.001828] [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: 02/10/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is a highly contagious morbillivirus related to measles and canine distemper virus, mostly affecting small ruminants. The corresponding PPR disease has a high clinical impact in goats and is characterized by fever, oral and nasal erosions, diarrhoea and pneumonia. In addition, massive infection of lymphoid tissues causes lymphopaenia and immune suppression. This results in increased susceptibility to secondary bacterial infections, explaining the observed high mortality in some outbreaks. We studied the pathogenesis of PPR by experimental inoculation of Dutch domestic goats with a recombinant virulent PPRV strain modified to express EGFP and compared it to an EGFP-expressing vaccine strain of PPRV. After intratracheal inoculation with virulent PPRV, animals developed fever, viraemia and leucopaenia, and shed virus from the respiratory and gastro-intestinal tracts. Macroscopic evaluation of fluorescence at the peak of infection 7 days post-inoculation (dpi) showed prominent PPRV infection of the respiratory tract, lymphoid tissues, gastro-intestinal tract, mucosae and skin. Flow cytometry of PBMCs collected over time demonstrated a cell-associated viraemia mediated by infected lymphocytes. At 14 dpi, pathognomonic zebra stripes were detected in the mucosa of the large intestine. In contrast, vaccine strain-inoculated goats remained largely macroscopically fluorescence negative and did not present clinical signs. A low-level viraemia was detected by flow cytometry, but at necropsy no histological lesions were observed. Animals from both groups seroconverted as early as 7 dpi and sera efficiently neutralized virulent PPRV in vitro. Combined, this work presents a study of the pathogenesis of wild type- and vaccine-based PPRV in its natural host. This study shows the strength of recombinant EGFP-expressing viruses in fluorescence-guided pathogenesis studies.
Collapse
Affiliation(s)
| | - Phaedra L Eblé
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | - René G P van Gennip
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | | | - Rory D de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
| | - Lucien J M van Keulen
- Department of Infection Biology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | - Rik L de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, Netherlands.,Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | - Piet A van Rijn
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands.,Department of Biochemistry, Centre of Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
5
|
Research Progress on Emerging Viral Pathogens of Small Ruminants in China during the Last Decade. Viruses 2022; 14:v14061288. [PMID: 35746759 PMCID: PMC9228844 DOI: 10.3390/v14061288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/26/2022] Open
Abstract
China is the country with the largest number of domestic small ruminants in the world. Recently, the intensive and large-scale sheep/goat raising industry has developed rapidly, especially in nonpastoral regions. Frequent trading, allocation, and transportation result in the introduction and prevalence of new pathogens. Several new viral pathogens (peste des petits ruminants virus, caprine parainfluenza virus type 3, border disease virus, enzootic nasal tumor virus, caprine herpesvirus 1, enterovirus) have been circulating and identified in China, which has attracted extensive attention from both farmers and researchers. During the last decade, studies examining the etiology, epidemiology, pathogenesis, diagnostic methods, and vaccines for these emerging viruses have been conducted. In this review, we focus on the latest findings and research progress related to these newly identified viral pathogens in China, discuss the current situation and problems, and propose research directions and prevention strategies for different diseases in the future. Our aim is to provide comprehensive and valuable information for the prevention and control of these emerging viruses and highlight the importance of surveillance of emerging or re-emerging viruses.
Collapse
|
6
|
PPRV-Induced Autophagy Facilitates Infectious Virus Transmission by the Exosomal Pathway. J Virol 2022; 96:e0024422. [PMID: 35319226 DOI: 10.1128/jvi.00244-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. We showed previously that PPRV induced sustained autophagy for their replication in host cells. Many studies have shown that exosomes released from virus-infected cells contain a variety of viral and host cellular factors that are able to modulate the recipient's cellular response and result in productive infection of the recipient host. Here, we show that PPRV infection results in packaging of the viral genomic RNA and partial viral proteins into exosomes of Vero cells and upregulates exosome secretion. We provide evidence showing that the exosomal viral cargo can be transferred to and establish productive infection in a new target cell. Importantly, our study reveals that PPRV-induced autophagy enhances exosome secretion and exosome-mediated virus transmission. Additionally, our data show that TSG101 may be involved in the sorting of the infectious PPRV RNA into exosomes to facilitate the release of PPRV through the exosomal pathway. Taken together, our results suggest a novel mechanism involving autophagy and exosome-mediated PPRV intercellular transmission. IMPORTANCE Autophagy plays an important role in PPRV pathogenesis. The role of exosomes in viral infections is beginning to be appreciated. The present study examined the role of autophagy in secretion of infectious PPRV from Vero cells. Our data provided the first direct evidence that ATG7-mediated autophagy enhances exosome secretion and exosome-mediated PPRV transmission. TSG101 may be involved in the sorting of the infectious PPRV RNA genomes into exosomes to facilitate the release of PPRV through the exosomal pathway. Inhibition of PPRV-induced autophagy or TSG101 expression could be used as a strategy to block exosome-mediated virus transmission.
Collapse
|
7
|
Kinimi E, Mahapatra M, Kgotlele T, Makange MR, Tennakoon C, Njeumi F, Odongo S, Muyldermans S, Kock R, Parida S, Rweyemamu M, Misinzo G. Complete Genome Sequencing of Field Isolates of Peste des Petits Ruminants Virus from Tanzania Revealed a High Nucleotide Identity with Lineage III PPR Viruses. Animals (Basel) 2021; 11:2976. [PMID: 34679994 PMCID: PMC8532778 DOI: 10.3390/ani11102976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 10/12/2021] [Indexed: 01/30/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) causes a highly devastating disease of sheep and goats that threatens food security, small ruminant production and susceptible endangered wild ruminants. With policy directed towards achieving global PPR eradication, the establishment of cost-effective genomic surveillance tools is critical where PPR is endemic. Genomic data can provide sufficient in-depth information to identify the pockets of endemicity responsible for PPRV persistence and viral evolution, and direct an appropriate vaccination response. Yet, access to the required sequencing technology is low in resource-limited settings and is compounded by the difficulty of transporting clinical samples from wildlife across international borders due to the Convention on International Trade in Endangered Species (CITES) of Wild Fauna and Flora, and Nagoya Protocol regulations. Oxford nanopore MinION sequencing technology has recently demonstrated an extraordinary performance in the sequencing of PPRV due to its rapidity, utility in endemic countries and comparatively low cost per sample when compared to other whole-genome (WGS) sequencing platforms. In the present study, Oxford nanopore MinION sequencing was utilised to generate complete genomes of PPRV isolates collected from infected goats in Ngorongoro and Momba districts in the northern and southern highlands of Tanzania during 2016 and 2018, respectively. The tiling multiplex polymerase chain reaction (PCR) was carried out with twenty-five pairs of long-read primers. The resulting PCR amplicons were used for nanopore library preparation and sequencing. The analysis of output data was complete genomes of PPRV, produced within four hours of sequencing (accession numbers: MW960272 and MZ322753). Phylogenetic analysis of the complete genomes revealed a high nucleotide identity, between 96.19 and 99.24% with lineage III PPRV currently circulating in East Africa, indicating a common origin. The Oxford nanopore MinION sequencer can be deployed to overcome diagnostic and surveillance challenges in the PPR Global Control and Eradication program. However, the coverage depth was uneven across the genome and amplicon dropout was observed mainly in the GC-rich region between the matrix (M) and fusion (F) genes of PPRV. Thus, larger field studies are needed to allow the collection of sufficient data to assess the robustness of nanopore sequencing technology.
Collapse
Affiliation(s)
- Edson Kinimi
- SACIDS Africa Centre of Excellence for Infectious Diseases, SACIDS Foundation for One Health, Sokoine University of Agriculture, P.O. Box 3297, Morogoro 67125, Tanzania; (S.P.); (M.R.)
- Department of Veterinary Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3017, Morogoro 67125, Tanzania
- Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3019, Morogoro 67125, Tanzania; (T.K.); (M.R.M.)
| | - Mana Mahapatra
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (M.M.); (C.T.)
| | - Tebogo Kgotlele
- Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3019, Morogoro 67125, Tanzania; (T.K.); (M.R.M.)
| | - Mariam R. Makange
- Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3019, Morogoro 67125, Tanzania; (T.K.); (M.R.M.)
| | - Chandana Tennakoon
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (M.M.); (C.T.)
| | - Felix Njeumi
- Food and Agriculture Organization of the United Nations (FAO), Viale delle Terme di Caracalla, 00153 Rome, Italy;
| | - Steven Odongo
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine, Animal Resources and Biosecurity (COVAB), Makerere University, Kampala P.O. Box 7062, Uganda;
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium;
| | - Richard Kock
- The Royal Veterinary College, University of London, Hawkshead Lane, North Mymms, Hertfordshire, Hatfield AL9 7TA, UK;
| | - Satya Parida
- SACIDS Africa Centre of Excellence for Infectious Diseases, SACIDS Foundation for One Health, Sokoine University of Agriculture, P.O. Box 3297, Morogoro 67125, Tanzania; (S.P.); (M.R.)
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (M.M.); (C.T.)
- Food and Agriculture Organization of the United Nations (FAO), Viale delle Terme di Caracalla, 00153 Rome, Italy;
| | - Mark Rweyemamu
- SACIDS Africa Centre of Excellence for Infectious Diseases, SACIDS Foundation for One Health, Sokoine University of Agriculture, P.O. Box 3297, Morogoro 67125, Tanzania; (S.P.); (M.R.)
| | - Gerald Misinzo
- SACIDS Africa Centre of Excellence for Infectious Diseases, SACIDS Foundation for One Health, Sokoine University of Agriculture, P.O. Box 3297, Morogoro 67125, Tanzania; (S.P.); (M.R.)
- Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3019, Morogoro 67125, Tanzania; (T.K.); (M.R.M.)
| |
Collapse
|
8
|
Rojas JM, Sevilla N, Martín V. A New Look at Vaccine Strategies Against PPRV Focused on Adenoviral Candidates. Front Vet Sci 2021; 8:729879. [PMID: 34568477 PMCID: PMC8455998 DOI: 10.3389/fvets.2021.729879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is a virus that mainly infects goats and sheep causing significant economic loss in Africa and Asia, but also posing a serious threat to Europe, as recent outbreaks in Georgia (2016) and Bulgaria (2018) have been reported. In order to carry out the eradication of PPRV, an objective set for 2030 by the Office International des Epizooties (OIE) and the Food and Agriculture Organization of the United Nations (FAO), close collaboration between governments, pharmaceutical companies, farmers and researchers, among others, is needed. Today, more than ever, as seen in the response to the SARS-CoV2 pandemic that we are currently experiencing, these goals are feasible. We summarize in this review the current vaccination approaches against PPRV in the field, discussing their advantages and shortfalls, as well as the development and generation of new vaccination strategies, focusing on the potential use of adenovirus as vaccine platform against PPRV and more broadly against other ruminant pathogens.
Collapse
Affiliation(s)
| | | | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA-CSIC), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
9
|
Alfred N, Qian B, Qin X, Yin X, Prajapati M, Dou Y, Li Y, Zhang Z. Inhibition of eIF2α Phosphorylation by Peste des Petits Ruminant Virus Phosphoprotein Facilitates Viral Replication. Front Vet Sci 2021; 8:645571. [PMID: 34295932 PMCID: PMC8290123 DOI: 10.3389/fvets.2021.645571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Peste des petits ruminant virus (PPRV) causes a highly contagious disease in small ruminants. The molecular mechanism of PPRV replication and its interactions with hosts are poorly studied. In other paramyxoviruses, the viral phosphoprotein (P) has been associated with multiple functions for key biological processes such as the regulation of transcription, translation, and the control of cell cycle. Phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α) is an important process for gene regulation in host cells under stress, including viral infection. In the present study, molecular mechanisms associated with PPRV replication and viral interaction with host cells were investigated. We describe the ability of PPRV to dephosphorylate eIF2α and the potential of PPRV P protein to induce the host cellular growth arrest DNA damage protein (GADD34), which is known to be associated with eIF2α dephosphorylation. Furthermore, we observed that PPRV P protein alone could block PERK/eIF2α phosphorylation. We speculate that PPRV exploits eIF2α dephosphorylation to facilitate viral replication and that PPRV P protein is involved in this molecular mechanism. This work provides new insights into further understanding PPRV pathobiology and its viral/host interactions.
Collapse
Affiliation(s)
- Niyokwishimira Alfred
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Bang Qian
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiaodong Qin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangping Yin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Meera Prajapati
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Yongxi Dou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yanmin Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Zhidong Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| |
Collapse
|
10
|
Zhao H, Njeumi F, Parida S, Benfield CTO. Progress towards Eradication of Peste des Petits Ruminants through Vaccination. Viruses 2021; 13:v13010059. [PMID: 33466238 PMCID: PMC7824732 DOI: 10.3390/v13010059] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 01/05/2023] Open
Abstract
Peste des petits ruminants (PPR) is a transboundary viral disease that threatens more than 1.74 billion goats and sheep in approximately 70 countries globally. In 2015, the international community set the goal of eradicating PPR by 2030, and, since then, Food and Agriculture Organization of the United Nations (FAO) and World Organization for Animal Health (OIE) have jointly developed and implemented the Global Control and Eradication Strategy for PPR. Here, data from the United Nations Food and Agriculture Organization Statistical Database (FAOSTAT), the OIE World Animal Health Information System (WAHIS), Regional Roadmap Meetings, and countries' responses to PPR Monitoring and Assessment Tool (PMAT) questionnaires were analyzed to inform on current progress towards PPR eradication. OIE recorded the use of over 333 million doses of vaccine in 12 countries from 2015 to 2018, 41.8% of which were used in Asia and 58.2% in Africa. Between 2015 and 2019, a total of 12,757 PPR outbreaks were reported to OIE: 75.1% in Asia, 24.8% in Africa, and 0.1% in Europe. The number of global outbreaks in 2019 fell to 1218, compared with 3688 in 2015. Analysis of vaccine use and PPR outbreaks in countries indicates that disease control strategies, particularly vaccination campaigns and vaccine distribution strategies, still require scientific evaluation. It is imperative that vaccination is undertaken based on the epidemiology of the disease in a region and is coordinated between neighboring countries to restrict transboundary movements. Strengthening surveillance and post-vaccination sero-monitoring at the national level is also essential. The PPR vaccine stock/bank established by FAO, OIE, and other partners have improved the quality assurance and supply of vaccines. However, to achieve PPR eradication, filling the funding gap for vaccination campaigns and other program activities will be critical.
Collapse
Affiliation(s)
- Hang Zhao
- Jiangsu Key Laboratory for Food Quality and Safety–State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Felix Njeumi
- Food and Agriculture Organization of the United Nations (FAO), Viale delle Terme di Caracalla, 00153 Rome, Italy;
| | - Satya Parida
- The Pirbright Institute, Woking GU24 0NF, UK
- Correspondence: (S.P.); (C.T.O.B.)
| | - Camilla T. O. Benfield
- Food and Agriculture Organization of the United Nations (FAO), Viale delle Terme di Caracalla, 00153 Rome, Italy;
- Royal Veterinary College, University of London, London NW1 0TU, UK
- Correspondence: (S.P.); (C.T.O.B.)
| |
Collapse
|
11
|
Yang DK, Kim HH, Park YR, Yoo JY, Park Y, Park J, Hyun BH. Generation of a recombinant rabies virus expressing green fluorescent protein for a virus neutralization antibody assay. J Vet Sci 2021; 22:e56. [PMID: 34313041 PMCID: PMC8318786 DOI: 10.4142/jvs.2021.22.e56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/27/2021] [Accepted: 06/29/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Fluorescent antibody virus neutralization (FAVN) test is a standard assay for quantifying rabies virus-neutralizing antibody (VNA) in serum. However, a safer rabies virus (RABV) should be used in the FAVN assay. There is a need for a new method that is economical and time-saving by eliminating the immunostaining step. OBJECTIVES We aimed to improve the traditional FAVN method by rescuing and characterizing a new recombinant RABV expressing green fluorescent protein (GFP). METHODS A new recombinant RABV expressing GFP designated as ERAGS-GFP was rescued using a reverse genetic system. Immuno-fluorescence assay, peroxidase-linked assay, electron microscopy and reverse transcription polymerase chain reaction were performed to confirm the recombinant ERAGS-GFP virus as a RABV expressing the GFP gene. The safety of ERAGS-GFP was evaluated in 4-week-old mice. The rabies VNA titers were measured and compared with conventional FAVN and FAVN-GFP tests using VERO cells. RESULTS The virus propagated in VERO cells was confirmed as RABV expressing GFP. The ERAGS-GFP showed the highest titer (108.0 TCID50/mL) in VERO cells at 5 days post-inoculation, and GFP expression persisted until passage 30. The body weight of 4-week-old mice inoculated intracranially with ERAGS-GFP continued to increase and the survival rate was 100%. In 62 dog sera, the FAVN-GFP result was significantly correlated with that of conventional FAVN (r = 0.95). CONCLUSIONS We constructed ERAGS-GFP, which could replace the challenge virus standard-11 strain used in FAVN test.
Collapse
Affiliation(s)
- Dong Kun Yang
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea.
| | - Ha Hyun Kim
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Yu Ri Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Jae Young Yoo
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Yeseul Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Jungwon Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Bang Hun Hyun
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| |
Collapse
|
12
|
Prajapati M, Dou Y, Zhu X, Zhao S, Alfred N, Li Y, Zhang Z. Development of an Enzyme-Linked Immunosorbent Assay Based on CD150/SLAM for the Detection of Peste des Petits Ruminant Virus. Front Vet Sci 2020; 7:196. [PMID: 32411735 PMCID: PMC7198758 DOI: 10.3389/fvets.2020.00196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/25/2020] [Indexed: 11/13/2022] Open
Abstract
Peste des petits ruminant (PPR) is an economically important severe viral disease of small ruminants that affects primarily the respiratory and digestive tract. Specific detection of the PPR virus (PPRV) antigen plays an important role in the disease control and eradication program. In this study, an indirect enzyme-linked immunosorbent assay (ELISA) based on the recombinant goat signaling lymphocyte activation molecule (SLAM) as the capture ligand was successfully developed for the detection of the PPRV antigen (PPRV SLAM-iELISA). The assay was highly specific for PPRV with no cross-reactions among foot and mouth disease virus, Orf virus, sheep pox virus, and goat pox virus and had a sensitivity with a detection limit of 1.56 × 101 TCID50/reaction (50 μl). Assessment of 136 samples showed that the developed PPRV SLAM-iELISA was well correlated with real-time RT-qPCR assays and commercially available sandwich ELISA for detection of PPRV and showed relative sensitivity and specificity of 93.75 and 100.83%, respectively. These results suggest that the developed PPRV SLAM-iELISA is suitable for specific detection of the PPRV antigen. This study demonstrated for the first time that the goat SLAM, the cellular receptor for PPRV, can be used for the development of a diagnostic method for the detection of PPRV.
Collapse
Affiliation(s)
- Meera Prajapati
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,CAAS-ILRI Joint Laboratory for Ruminant Disease Control, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China.,Animal Health Research Division, Nepal Agricultural Research Council, Lalitpur, Nepal
| | - Yongxi Dou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xueliang Zhu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shuaiyang Zhao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Niyokwishimira Alfred
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yanmin Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhidong Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,CAAS-ILRI Joint Laboratory for Ruminant Disease Control, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China
| |
Collapse
|
13
|
Jia XX, Wang H, Liu Y, Meng DM, Fan ZC. Development of vaccines for prevention of peste-des-petits-ruminants virus infection. Microb Pathog 2020; 142:104045. [PMID: 32035105 DOI: 10.1016/j.micpath.2020.104045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 01/09/2020] [Accepted: 02/05/2020] [Indexed: 01/22/2023]
Abstract
Peste des petits ruminants (PPR) is a highly contagious and fatal disease of small ruminants, particularly sheep and goats. This disease leads to high morbidity and mortality of small ruminants, thus resulting in devastating economic loss to the livestock industry globally. The severe disease impact has prompted the Food and Agriculture Organization of the United Nations (FAO) and the World Organization for Animal Health (OIE) to develop a global strategy for the control and eradication of PPR by 2030. Over the past decades, the control of PPR is mainly achieved through vaccinating the animals with live-attenuated vaccines, e.g., rinderpest vaccines. As a closely related disease to PPR of large ruminants, rinderpest was eradicated in 2011 and its vaccines subsequently got banned in order to keep rinderpest-free zones. Consequently, it is desirable to develop homologous PPR vaccines to control the disease. The present review summarizes the objectives of PPR control and eradication by focusing on the homologous PPR vaccines.
Collapse
Affiliation(s)
- Xue-Xia Jia
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Hui Wang
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Ying Liu
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - De-Mei Meng
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Zhen-Chuan Fan
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China.
| |
Collapse
|
14
|
Kamel M, El-Sayed A. Toward peste des petits virus (PPRV) eradication: Diagnostic approaches, novel vaccines, and control strategies. Virus Res 2019; 274:197774. [PMID: 31606355 DOI: 10.1016/j.virusres.2019.197774] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
Abstract
Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease affecting domestic and wild small ruminants' species besides camels reared in Africa, Asia and the Middle East. The virus is a serious paramount challenge to the sustainable agriculture advancement in the developing world. The disease outbreak was also detected for the first time in the European Union namely in Bulgaria at 2018. Therefore, the disease has lately been aimed for eradication with the purpose of worldwide clearance by 2030. Radically, the vaccines needed for effectively accomplishing this aim are presently convenient; however, the availableness of innovative modern vaccines to fulfill the desideratum for Differentiating between Infected and Vaccinated Animals (DIVA) may mitigate time spent and financial disbursement of serological monitoring and surveillance in the advanced levels for any disease obliteration campaign. We here highlight what is at the present time well-known about the virus and the different available diagnostic tools. Further, we interject on current updates and insights on several novel vaccines and on the possible current and prospective strategies to be applied for disease control.
Collapse
Affiliation(s)
- Mohamed Kamel
- Faculty of Veterinary Medicine, Department of Medicine and Infectious Diseases, Cairo University, Giza, Egypt.
| | - Amr El-Sayed
- Faculty of Veterinary Medicine, Department of Medicine and Infectious Diseases, Cairo University, Giza, Egypt
| |
Collapse
|
15
|
Host Cellular Receptors for the Peste des Petits Ruminant Virus. Viruses 2019; 11:v11080729. [PMID: 31398809 PMCID: PMC6723671 DOI: 10.3390/v11080729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Peste des Petits Ruminant (PPR) is an important transboundary, OIE-listed contagious viral disease of primarily sheep and goats caused by the PPR virus (PPRV), which belongs to the genus Morbillivirus of the family Paramyxoviridae. The mortality rate is 90–100%, and the morbidity rate may reach up to 100%. PPR is considered economically important as it decreases the production and productivity of livestock. In many endemic poor countries, it has remained an obstacle to the development of sustainable agriculture. Hence, proper control measures have become a necessity to prevent its rapid spread across the world. For this, detailed information on the pathogenesis of the virus and the virus host interaction through cellular receptors needs to be understood clearly. Presently, two cellular receptors; signaling lymphocyte activation molecule (SLAM) and Nectin-4 are known for PPRV. However, extensive information on virus interactions with these receptors and their impact on host immune response is still required. Hence, a thorough understanding of PPRV receptors and the mechanism involved in the induction of immunosuppression is crucial for controlling PPR. In this review, we discuss PPRV cellular receptors, viral host interaction with cellular receptors, and immunosuppression induced by the virus with reference to other Morbilliviruses.
Collapse
|
16
|
Yadav AK, Chaudhary D, Bhadouriya S, Chandrasekar S, Dhanesh VV, Rajak KK, Singh RP, Ramakrishnan MA, Singh RK, Muthuchelvan D. Expression and characterization of the non-structural protein V of small ruminant morbillivirus. Virusdisease 2019; 30:465-468. [PMID: 31803815 DOI: 10.1007/s13337-019-00539-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 06/25/2019] [Indexed: 11/24/2022] Open
Abstract
Peste-des-petits ruminants is a transboundary viral disease of small ruminants caused by small ruminant morbillivirus (SRMV). In the present study, the full-length V gene of SRMV was constructed through site-directed mutagenesis from the P gene transcripts of the vaccine virus (Sungri/96 India) and expressed in a prokaryotic expression system. In animals, the seroconversion against this protein occurs from 14-days and is getting produced from 48 h in cell culture. An indirect ELISA developed using this protein has a relative sensitivity and relative specificity of 77.73% and 73.775%, respectively as compared to c-ELISA. In this ELISA, it was observed that most of the convalescent animals elicited higher level of antibodies than vaccinated animals.
Collapse
Affiliation(s)
- Ajay Kumar Yadav
- 1ICAR, Indian Veterinary Research Institute, Mukteswar, Uttarakhand 263 138 India
| | - Dheeraj Chaudhary
- 1ICAR, Indian Veterinary Research Institute, Mukteswar, Uttarakhand 263 138 India
| | - Sakshi Bhadouriya
- 1ICAR, Indian Veterinary Research Institute, Mukteswar, Uttarakhand 263 138 India
| | - S Chandrasekar
- 1ICAR, Indian Veterinary Research Institute, Mukteswar, Uttarakhand 263 138 India
| | - V V Dhanesh
- 1ICAR, Indian Veterinary Research Institute, Mukteswar, Uttarakhand 263 138 India
| | - Kaushal K Rajak
- 2ICAR, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122 India
| | - R P Singh
- 2ICAR, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122 India
| | - M A Ramakrishnan
- 1ICAR, Indian Veterinary Research Institute, Mukteswar, Uttarakhand 263 138 India
| | - R K Singh
- 2ICAR, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122 India
| | | |
Collapse
|
17
|
Jarikre TA, Taiwo JO, Emikpe BO, Akpavie SO. Protective effect of intranasal peste des petits ruminants virus and bacterin vaccinations: Clinical, hematological, serological, and serum oxidative stress changes in challenged goats. Vet World 2019; 12:945-950. [PMID: 31528016 PMCID: PMC6702579 DOI: 10.14202/vetworld.2019.945-950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 05/15/2019] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: The current vaccination for peste des petits ruminants virus (PPRV) is stalled by myriad challenges and continuous endemicity of pneumonia due to fulminant bacterial complication in goats. The present study evaluated the protective effect of intranasal PPRV linage 1 and bacterine vaccinations. Materials and Methods: Twelve West African Dwarf (WAD) goats aged 6 months were randomly grouped and vaccinated within 2 weeks using a combination of PPRV lineage 1 vaccine (Nig/75), and bacterin from Mannheimia haemolytica (Mh) or Pasteurella multocida intranasally. The goats were observed for 3 weeks post-vaccination before comingled with a known infected WAD goat with apparent clinical signs of peste des petits ruminants and further observed clinically for 5 weeks post-infection (PI). Blood samples were taken for hematology while sera were assayed for antioxidants (glutathione peroxidase, glutathione transferase, and superoxide dismutase) activities and pro-oxidants (malondialdehyde content, reduced glutathione, hydrogen peroxide generation, and myeloperoxidase) using spectrophotometric methods. Data were subjected to parametric statistics at α=0.05 using GraphPad Prism version 21. Results: Clinically, there were pyrexia, oculonasal discharge, diarrhea, anemia, leukopenia, and increased pro-oxidants in the unvaccinated goats, while moderate neutrophilia and leukocytosis were observed in PPRV and bacterin vaccinated goats. Two unvaccinated goats were weak and euthanized at 13 and 28 days PI. The goats vaccinated with PPRV and Mh showed better response clinically and biochemically. Conclusion: The mucosal vaccination of goats with PPRV vaccine and bacterine will protect against exposure and culminate in the development of protective mucosal, humoral, and cell-mediated immune responses. This vaccination strategy will provide framework needed in the prevention and control of endemic caprine pneumonia in Nigeria.
Collapse
Affiliation(s)
| | - Jeremiah Olalekan Taiwo
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Benjamin Obukowho Emikpe
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Stephen Owarioro Akpavie
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| |
Collapse
|
18
|
Liu F, Zhang Y, Li L, Zuo Y, Sun C, Xiaodong W, Wang Z. Rescue of eGFP-expressing small ruminant morbillivirus for identifying susceptibilities of eight mammalian cell lines to its infection. Virus Res 2019; 261:60-64. [DOI: 10.1016/j.virusres.2018.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 12/14/2022]
|
19
|
Development of reverse genetics system for small ruminant morbillivirus: Rescuing recombinant virus to express Echinococcus granulosus EG95 antigen. Virus Res 2019; 261:50-55. [DOI: 10.1016/j.virusres.2018.12.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/02/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023]
|
20
|
Comparison of the Immunogenicities and Cross-Lineage Efficacies of Live Attenuated Peste des Petits Ruminants Virus Vaccines PPRV/Nigeria/75/1 and PPRV/Sungri/96. J Virol 2018; 92:JVI.01471-18. [PMID: 30258008 PMCID: PMC6258957 DOI: 10.1128/jvi.01471-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 09/14/2018] [Indexed: 01/14/2023] Open
Abstract
Despite the widespread use of live attenuated PPRV vaccines, this is the first systematic analysis of the immune response elicited in small ruminants. These data will help in the establishment of the immunological determinants of protection, an important step in the development of new vaccines, especially DIVA vaccines using alternative vaccination vectors. This study is also the first controlled test of the ability of the two major vaccines used against virulent PPRV strains from all genetic lineages of the virus, showing conclusively the complete cross-protective ability of these vaccines. Peste des petits ruminants (PPR) is a severe disease of goats and sheep that is widespread in Africa, the Middle East, and Asia. Several effective vaccines exist for the disease, based on attenuated strains of the virus (PPRV) that causes PPR. While the efficacy of these vaccines has been established by use in the field, the nature of the protective immune response has not been determined. In addition, while the vaccine derived from PPRV/Nigeria/75/1 (N75) is used in many countries, those developed in India have never been tested for their efficacy outside that country. We have studied the immune response in goats to vaccination with either N75 or the main Indian vaccine, which is based on isolate PPRV/India/Sungri/96 (S96). In addition, we compared the ability of these two vaccines, in parallel, to protect animals against challenge with pathogenic viruses from the four known genetic lineages of PPRV, representing viruses from different parts of Africa, as well as Asia. These studies showed that, while N75 elicited a stronger antibody response than S96, as measured by both enzyme-linked immunosorbent assay and virus neutralization, S96 resulted in more pronounced cellular immune responses, as measured by virus antigen-induced proliferation and interferon gamma production. While both vaccines induced comparable numbers of PPRV-specific CD8+ T cells, S96 induced a higher number of CD4+ T cells specifically responding to virus. Despite these quantitative and qualitative differences in the immune responses following vaccination, both vaccines gave complete clinical protection against challenge with all four lineages of PPRV. IMPORTANCE Despite the widespread use of live attenuated PPRV vaccines, this is the first systematic analysis of the immune response elicited in small ruminants. These data will help in the establishment of the immunological determinants of protection, an important step in the development of new vaccines, especially DIVA vaccines using alternative vaccination vectors. This study is also the first controlled test of the ability of the two major vaccines used against virulent PPRV strains from all genetic lineages of the virus, showing conclusively the complete cross-protective ability of these vaccines.
Collapse
|
21
|
Reverse Genetics for Peste des Petits Ruminants Virus: Current Status and Lessons to Learn from Other Non-segmented Negative-Sense RNA Viruses. Virol Sin 2018; 33:472-483. [PMID: 30456658 PMCID: PMC6335227 DOI: 10.1007/s12250-018-0066-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/11/2018] [Indexed: 11/20/2022] Open
Abstract
Peste des petits ruminants (PPR) is a highly contagious transboundary animal disease with a severe socio-economic impact on the livestock industry, particularly in poor countries where it is endemic. Full understanding of PPR virus (PPRV) pathobiology and molecular biology is critical for effective control and eradication of the disease. To achieve these goals, establishment of stable reverse genetics systems for PPRV would play a key role. Unfortunately, this powerful technology remains less accessible and poorly documented for PPRV. In this review, we discussed the current status of PPRV reverse genetics as well as the recent innovations and advances in the reverse genetics of other non-segmented negative-sense RNA viruses that could be applicable to PPRV. These strategies may contribute to the improvement of existing techniques and/or the development of new reverse genetics systems for PPRV.
Collapse
|
22
|
Yang B, Qi X, Chen Z, Chen S, Xue Q, Jia P, Wang T, Wang J. Binding and entry of peste des petits ruminants virus into caprine endometrial epithelial cells profoundly affect early cellular gene expression. Vet Res 2018; 49:8. [PMID: 29368634 PMCID: PMC5784595 DOI: 10.1186/s13567-018-0504-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 12/18/2017] [Indexed: 12/22/2022] Open
Abstract
Peste des petits ruminants virus (PPRV), the etiological agent of peste des petits ruminants (PPR), causes an acute or subacute disease in small ruminants. Although abortion is observed in an unusually large proportion of pregnant goats during outbreaks of PPR, the pathogenic mechanism underlying remains unclear. Here, the gene expression profile of caprine endometrial epithelial cells (EECs) infected with PPRV Nigeria 75/1 was determined by DNA microarray to investigate the cellular response immediately after viral entry. The microarray analysis revealed that a total of 146 genes were significantly dysregulated by PPRV internalization within 1 h post-infection (hpi). Of these, 85 genes were upregulated and 61 genes were downregulated. Most of these genes, including NFKB1A, JUNB, and IL1A, have not previously been reported in association with PPRV infection in goats. Following viral replication (24 hpi), the expression of 307 genes were significantly upregulated and that of 261 genes were downregulated. The data for the genes differentially expressed in EECs were subjected to a time sequence profile analysis, gene network analysis and pathway analysis. The gene network analysis showed that 13 genes (EIF2AK3, IL10, TLR4, ZO3, NFKBIB, RAC1, HSP90AA1, SMAD7, ARG2, JUNB, ZFP36, APP, and IL1A) were located in the core of the network. We clearly demonstrate that PPRV infection upregulates the expression of nectin-4 after 1 hpi, which peaked at 24 hpi in EECs. In conclusion, this study demonstrates the early cellular gene expression in the caprine endometrial epithelial cells after the binding and entry of PPRV.
Collapse
Affiliation(s)
- Bo Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhijie Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shuying Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, 100000, China
| | - Peilong Jia
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ting Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
23
|
Liu F, Li J, Li L, Liu Y, Wu X, Wang Z. Peste des petits ruminants in China since its first outbreak in 2007: A 10-year review. Transbound Emerg Dis 2018; 65:638-648. [PMID: 29322642 DOI: 10.1111/tbed.12808] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Indexed: 11/30/2022]
Abstract
Peste des petits ruminants (PPR) is a highly infectious disease of small ruminants and caused by small ruminant morbillivirus (SRMV), formerly called peste-des-petits-ruminants virus (PPRV). This disease is circulating in Africa (except most countries in southern Africa), the Arabian Peninsula, the Middle East, and Central, East and South-East Asia. Peste des petits ruminants is still regarded as an exotic disease in China, where its first outbreak was reported in the Ngari region of Tibet in 2007, but effectively controlled by slaughter, vaccination and animal movement restriction in PPR-infected areas. However, PPR re-emerged in Xinjiang of China in December 2013, rapidly spread into much of China in the first half of 2014, but since then was substantially inhibited countrywide. Phylogenetic analysis shows that SRMVs from China share the highest homology with others from its neighbouring countries, possibly indicating the transboundary transmission of SRMVs. In 2015, a national eradication program for PPR was issued and has been being implemented in China, expecting to achieve a PPR-eradicating aim countrywide by 2020. Here, we reviewed a 10-year history (2007-2017) of PPR in China, including two major outbreaks, its infection in wild species, development of diagnostics and vaccines, and implementation of the national eradication program.
Collapse
Affiliation(s)
- F Liu
- OIE Reference Laboratory for Peste des Petits Ruminants, National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, Shandong, China
| | - J Li
- OIE Reference Laboratory for Peste des Petits Ruminants, National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, Shandong, China
| | - L Li
- OIE Reference Laboratory for Peste des Petits Ruminants, National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, Shandong, China
| | - Y Liu
- OIE Reference Laboratory for Peste des Petits Ruminants, National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, Shandong, China
| | - X Wu
- OIE Reference Laboratory for Peste des Petits Ruminants, National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, Shandong, China
| | - Z Wang
- OIE Reference Laboratory for Peste des Petits Ruminants, National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, Shandong, China
| |
Collapse
|
24
|
Baron MD, Diop B, Njeumi F, Willett BJ, Bailey D. Future research to underpin successful peste des petits ruminants virus (PPRV) eradication. J Gen Virol 2017; 98:2635-2644. [PMID: 29022862 PMCID: PMC5845661 DOI: 10.1099/jgv.0.000944] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is a significant pathogen of small ruminants and is prevalent in much of Africa, the Near and Middle East and Asia. Despite the availability of an efficacious and cheap live-attenuated vaccine, the virus has continued to spread, with its range stretching from Morocco in the west to China and Mongolia in the east. Some of the world's poorest communities rely on small ruminant farming for subsistence and the continued endemicity of PPRV is a constant threat to their livelihoods. Moreover, PPRV's effects on the world's population are felt broadly across many economic, agricultural and social situations. This far-reaching impact has prompted the Food and Agriculture Organization of the United Nations (FAO) and the World Organisation for Animal Health (OIE) to develop a global strategy for the eradication of this virus and its disease. PPRV is a morbillivirus and, given the experience of these organizations in eradicating the related rinderpest virus, the eradication of PPRV should be feasible. However, there are many critical areas where basic and applied virological research concerning PPRV is lacking. The purpose of this review is to highlight areas where new research could be performed in order to guide and facilitate the eradication programme. These areas include studies on disease transmission and epidemiology, the existence of wildlife reservoirs and the development of next-generation vaccines and diagnostics. With the support of the international virology community, the successful eradication of PPRV can be achieved.
Collapse
Affiliation(s)
- Michael D Baron
- The Pirbright Institute, Ash Rd Pirbright, Surrey GU24 0NF, UK
| | - Bouna Diop
- Food and Agriculture Organization of the United Nation, FAO, 00153 Rome, Italy
| | - Felix Njeumi
- Food and Agriculture Organization of the United Nation, FAO, 00153 Rome, Italy
| | - Brian J Willett
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow, G61 1QH, UK
| | - Dalan Bailey
- College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,The Pirbright Institute, Ash Rd Pirbright, Surrey GU24 0NF, UK
| |
Collapse
|
25
|
Sanz Bernardo B, Goodbourn S, Baron MD. Control of the induction of type I interferon by Peste des petits ruminants virus. PLoS One 2017; 12:e0177300. [PMID: 28475628 PMCID: PMC5419582 DOI: 10.1371/journal.pone.0177300] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/25/2017] [Indexed: 12/24/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is a morbillivirus that produces clinical disease in goats and sheep. We have studied the induction of interferon-β (IFN-β) following infection of cultured cells with wild-type and vaccine strains of PPRV, and the effects of such infection with PPRV on the induction of IFN-β through both MDA-5 and RIG-I mediated pathways. Using both reporter assays and direct measurement of IFN-β mRNA, we have found that PPRV infection induces IFN-β only weakly and transiently, and the virus can actively block the induction of IFN-β. We have also generated mutant PPRV that lack expression of either of the viral accessory proteins (V&C) to characterize the role of these proteins in IFN-β induction during virus infection. Both PPRV_ΔV and PPRV_ΔC were defective in growth in cell culture, although in different ways. While the PPRV V protein bound to MDA-5 and, to a lesser extent, RIG-I, and over-expression of the V protein inhibited both IFN-β induction pathways, PPRV lacking V protein expression can still block IFN-β induction. In contrast, PPRV C bound to neither MDA-5 nor RIG-I, but PPRV lacking C protein expression lost the ability to block both MDA-5 and RIG-I mediated activation of IFN-β. These results shed new light on the inhibition of the induction of IFN-β by PPRV.
Collapse
Affiliation(s)
| | - Stephen Goodbourn
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | | |
Collapse
|
26
|
Kumar N, Barua S, Riyesh T, Tripathi BN. Advances in peste des petits ruminants vaccines. Vet Microbiol 2017; 206:91-101. [PMID: 28161212 PMCID: PMC7130925 DOI: 10.1016/j.vetmic.2017.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/13/2016] [Accepted: 01/12/2017] [Indexed: 11/27/2022]
Abstract
Peste des petits ruminants (PPR) is a highly contagious disease of small ruminants that leads to high morbidity and mortality thereby results in devastating economic consequences to the livestock industry. PPR is currently endemic across most parts of Asia and Africa, the two regions with the highest concentration of poor people in the world. Sheep and goats in particularly contribute significantly towards the upliftment of livelihood of the poor and marginal farmers in these regions. In this context, PPR directly affecting the viability of sheep and goat husbandry has emerged as a major hurdle in the development of these regions. The control of PPR in these regions could significantly contribute to poverty alleviation, therefore, the Office International des Epizooties (OIE) and Food and Agricultural Organization (FAO) have targeted the control and eradication of PPR by 2030 a priority. In order to achieve this goal, a potent, safe and efficacious live-attenuated PPR vaccine with long-lasting immunity is available for immunoprophylaxis. However, the live-attenuated PPR vaccine is thermolabile and needs maintenance of an effective cold chain to deliver into the field. In addition, the infected animals cannot be differentiated from vaccinated animals. To overcome these limitations, some recombinant vaccines have been developed. This review comprehensively describes about the latest developments in PPR vaccines.
Collapse
Affiliation(s)
- Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.
| | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.
| | - Thachamvally Riyesh
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| | - Bhupendra N Tripathi
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| |
Collapse
|
27
|
Zhang J, Liu W, Chen W, Li C, Xie M, Bu Z. Development of an Immunoperoxidase Monolayer Assay for the Detection of Antibodies against Peste des Petits Ruminants Virus Based on BHK-21 Cell Line Stably Expressing the Goat Signaling Lymphocyte Activation Molecule. PLoS One 2016; 11:e0165088. [PMID: 27768770 PMCID: PMC5074545 DOI: 10.1371/journal.pone.0165088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/26/2016] [Indexed: 11/19/2022] Open
Abstract
From 2013 to 2015, peste des petits ruminants (PPR) broke out in more than half of the provinces of China; thus, the application and development of diagnostic methods are very important for the control of PPR. Here, an immunoperoxidase monolayer assay (IPMA) was developed to detect antibodies against PPR. However, during IPMA development, we found that Vero cells were not the appropriate choice because staining results were not easily observed. Therefore, we first established a baby hamster kidney-goat signaling lymphocyte activation molecule (BHK-SLAM) cell line that could stably express goat SLAM for at least 20 generations. Compared with Vero cells, the PPR-mediated cytopathic effect occurred earlier in BHK-SLAM cells, and large syncytia appeared after virus infection. Based on this cell line and recombinant PPR virus expressing the green fluorescent protein (GFP) (rPPRV-GFP), an IPMA for PPR diagnosis was developed. One hundred and ninety-eight PPR serum samples from goats or sheep were tested by the IPMA and virus neutralization test (VNT). Compared with the VNT, the sensitivity and specificity of the IPMA were 91% and 100%, respectively, and the coincidence rate of the two methods was 95.5%. The IPMA assay could be completed in 4 h, compared with more than 6 d for the VNT using rPPRV-GFP, and it is easily performed, as the staining results can be observed under a microscope. Additionally, unlike the VNT, the IPMA does not require antigen purification, which will reduce its cost. In conclusion, the established IPMA will be an alternative method that replaces the VNT for detecting antibodies against PPRV in the field.
Collapse
Affiliation(s)
- Jialin Zhang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin, China
| | - Wenxing Liu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin, China
| | - Weiye Chen
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin, China
- * E-mail: (WYC); (ZGB)
| | - Cuicui Li
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin, China
| | - Meimei Xie
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin, China
| | - Zhigao Bu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin, China
- * E-mail: (WYC); (ZGB)
| |
Collapse
|
28
|
Applications of Replicating-Competent Reporter-Expressing Viruses in Diagnostic and Molecular Virology. Viruses 2016; 8:v8050127. [PMID: 27164126 PMCID: PMC4885082 DOI: 10.3390/v8050127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/31/2016] [Accepted: 04/29/2016] [Indexed: 12/13/2022] Open
Abstract
Commonly used tests based on wild-type viruses, such as immunostaining, cannot meet the demands for rapid detection of viral replication, high-throughput screening for antivirals, as well as for tracking viral proteins or virus transport in real time. Notably, the development of replicating-competent reporter-expressing viruses (RCREVs) has provided an excellent option to detect directly viral replication without the use of secondary labeling, which represents a significant advance in virology. This article reviews the applications of RCREVs in diagnostic and molecular virology, including rapid neutralization tests, high-throughput screening systems, identification of viral receptors and virus-host interactions, dynamics of viral infections in vitro and in vivo, vaccination approaches and others. However, there remain various challenges associated with RCREVs, including pathogenicity alterations due to the insertion of a reporter gene, instability or loss of the reporter gene expression, or attenuation of reporter signals in vivo. Despite all these limitations, RCREVs have become powerful tools for both basic and applied virology with the development of new technologies for generating RCREVs, the inventions of novel reporters and the better understanding of regulation of viral replication.
Collapse
|
29
|
Holzer B, Taylor G, Rajko-Nenow P, Hodgson S, Okoth E, Herbert R, Toye P, Baron MD. Determination of the minimum fully protective dose of adenovirus-based DIVA vaccine against peste des petits ruminants virus challenge in East African goats. Vet Res 2016; 47:20. [PMID: 26796101 PMCID: PMC4721059 DOI: 10.1186/s13567-016-0306-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/05/2016] [Indexed: 11/10/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) causes an economically important disease of sheep and goats, primarily in developing countries. It is becoming the object of intensive international control efforts. Current vaccines do not allow vaccinated and infected animals to be distinguished (no DIVA capability). We have previously shown that recombinant, replication-defective, adenovirus expressing the PPRV H glycoprotein (AdH) gives full protection against wild type PPRV challenge. We have now tested lower doses of the vaccine, as well as AdH in combination with a similar construct expressing the PPRV F glycoprotein (AdF). We show here that, in a local breed of goat in a country where PPR disease is common (Kenya), as little as 10(7) pfu of AdH gives significant protection against PPRV challenge, while a vaccine consisting of 10(8) pfu of each of AdH and AdF gives apparently sterile protection. These findings underline the utility of these constructs as DIVA vaccines for use in PPR control.
Collapse
Affiliation(s)
- Barbara Holzer
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| | - Geraldine Taylor
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| | | | - Sophia Hodgson
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| | - Edward Okoth
- International Livestock Research Institute, Nairobi, Kenya.
| | - Rebecca Herbert
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| | - Philip Toye
- International Livestock Research Institute, Nairobi, Kenya.
| | - Michael D Baron
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| |
Collapse
|
30
|
Abstract
Peste des petits ruminants virus (PPRV) causes a severe contagious disease of sheep and goats and has spread extensively through the developing world. Because of its disproportionately large impact on the livelihoods of low-income livestock keepers, and the availability of effective vaccines and good diagnostics, the virus is being targeted for global control and eventual eradication. In this review we examine the origin of the virus and its current distribution, and the factors that have led international organizations to conclude that it is eradicable. We also review recent progress in the molecular and cellular biology of the virus and consider areas where further research is required to support the efforts being made by national, regional, and international bodies to tackle this growing threat.
Collapse
Affiliation(s)
- M D Baron
- The Pirbright Institute, Surrey, United Kingdom.
| | - A Diallo
- CIRAD, UMR Contrôle des maladies animales exotiques et émergentes (CMAEE), Montpellier, France; INRA, UMR CMAEE 1309, Montpellier, France
| | - R Lancelot
- CIRAD, UMR Contrôle des maladies animales exotiques et émergentes (CMAEE), Montpellier, France; INRA, UMR CMAEE 1309, Montpellier, France
| | - G Libeau
- CIRAD, UMR Contrôle des maladies animales exotiques et émergentes (CMAEE), Montpellier, France; INRA, UMR CMAEE 1309, Montpellier, France
| |
Collapse
|
31
|
Logan N, McMonagle E, Drew AA, Takahashi E, McDonald M, Baron MD, Gilbert M, Cleaveland S, Haydon DT, Hosie MJ, Willett BJ. Efficient generation of vesicular stomatitis virus (VSV)-pseudotypes bearing morbilliviral glycoproteins and their use in quantifying virus neutralising antibodies. Vaccine 2015; 34:814-22. [PMID: 26706278 PMCID: PMC4742518 DOI: 10.1016/j.vaccine.2015.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 11/20/2015] [Accepted: 12/06/2015] [Indexed: 12/18/2022]
Abstract
Morbillivirus neutralising antibodies are traditionally measured using either plaque reduction neutralisation tests (PRNTs) or live virus microneutralisation tests (micro-NTs). While both test formats provide a reliable assessment of the strength and specificity of the humoral response, they are restricted by the limited number of viral strains that can be studied and often present significant biological safety concerns to the operator. In this study, we describe the adaptation of a replication-defective vesicular stomatitis virus (VSVΔG) based pseudotyping system for the measurement of morbillivirus neutralising antibodies. By expressing the haemagglutinin (H) and fusion (F) proteins of canine distemper virus (CDV) on VSVΔG pseudotypes bearing a luciferase marker gene, neutralising antibody titres could be measured rapidly and with high sensitivity. Further, by exchanging the glycoprotein expression construct, responses against distinct viral strains or species may be measured. Using this technique, we demonstrate cross neutralisation between CDV and peste des petits ruminants virus (PPRV). As an example of the value of the technique, we demonstrate that UK dogs vary in the breadth of immunity induced by CDV vaccination; in some dogs the neutralising response is CDV-specific while, in others, the neutralising response extends to the ruminant morbillivirus PPRV. This technique will facilitate a comprehensive comparison of cross-neutralisation to be conducted across the morbilliviruses.
Collapse
Affiliation(s)
- Nicola Logan
- MRC-University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow G61 1QH, United Kingdom.
| | - Elizabeth McMonagle
- MRC-University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow G61 1QH, United Kingdom.
| | - Angharad A Drew
- MRC-University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow G61 1QH, United Kingdom.
| | - Emi Takahashi
- Royal Veterinary College, University of London, London NW1 0TU, United Kingdom.
| | - Michael McDonald
- Veterinary Diagnostic Services, University of Glasgow, Garscube Estate, Glasgow G61 1QH, United Kingdom.
| | - Michael D Baron
- The Pirbright Institute, Pirbright, Surrey GU24 0NF, United Kingdom.
| | - Martin Gilbert
- Wildlife Conservation Society, Bronx, NY, USA; Boyd Orr Centre for Population and Ecosystem Health, Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, United Kingdom.
| | - Sarah Cleaveland
- Boyd Orr Centre for Population and Ecosystem Health, Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, United Kingdom.
| | - Daniel T Haydon
- Boyd Orr Centre for Population and Ecosystem Health, Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, United Kingdom.
| | - Margaret J Hosie
- MRC-University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow G61 1QH, United Kingdom.
| | - Brian J Willett
- MRC-University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow G61 1QH, United Kingdom.
| |
Collapse
|
32
|
Abstract
Peste des petits ruminants virus causes a highly infectious disease of small ruminants that is endemic across Africa, the Middle East and large regions of Asia. The virus is considered to be a major obstacle to the development of sustainable agriculture across the developing world and has recently been targeted by the World Organisation for Animal Health (OIE) and the Food and Agriculture Organisation (FAO) for eradication with the aim of global elimination of the disease by 2030. Fundamentally, the vaccines required to successfully achieve this goal are currently available, but the availability of novel vaccine preparations to also fulfill the requisite for differentiation between infected and vaccinated animals (DIVA) may reduce the time taken and the financial costs of serological surveillance in the later stages of any eradication campaign. Here, we overview what is currently known about the virus, with reference to its origin, updated global circulation, molecular evolution, diagnostic tools and vaccines currently available to combat the disease. Further, we comment on recent developments in our knowledge of various recombinant vaccines and on the potential for the development of novel multivalent vaccines for small ruminants.
Collapse
Affiliation(s)
- S Parida
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, United Kingdom; National Institute for Animal Biotechnology, Miyapur, Hyderabad, India.
| | - M Muniraju
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, United Kingdom
| | - M Mahapatra
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, United Kingdom
| | | | - H Buczkowski
- Animal and Plant Health Agency, Weybridge, Surrey, KT15 3NB United Kingdom
| | - A C Banyard
- Animal and Plant Health Agency, Weybridge, Surrey, KT15 3NB United Kingdom
| |
Collapse
|
33
|
Singh RP, Bandyopadhyay SK. Peste des petits ruminants vaccine and vaccination in India: sharing experience with disease endemic countries. Virusdisease 2015; 26:215-24. [PMID: 26645031 DOI: 10.1007/s13337-015-0281-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/16/2015] [Indexed: 11/26/2022] Open
Abstract
Peste des petits ruminants, a viral disease of small ruminants, the control of which is important for poverty alleviation and to ensure livelihood security in Asia, Middle East and Africa. In recognition of these issues, we developed and applied vaccine and diagnostics to demonstrate effective control of PPR during preceding 6 years in a sub-population of small ruminants in India. Two south Indian states, namely Andhra Pradesh and Karnataka, strongly indicated possibility of PPR control with more than 90 % reduction in number of reported outbreaks of PPR, mostly through mass vaccination. Similarly, the situation at the national level also demonstrated a decline of more than 75 % in the number of reported outbreaks. Sharing these experiences may motivate other countries for similar initiatives leading to progressive control of PPR, which is in line with the initiatives of the organizations like FAO/OIE and the recent platforms on global PPR research alliance.
Collapse
Affiliation(s)
- R P Singh
- Division of Biological Products, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 India
| | - S K Bandyopadhyay
- Agricultural Scientist Recruitment Board, KAB-1, Pusa, New Delhi, 110012 India
| |
Collapse
|
34
|
Baron J, Baron MD. Development of a helper cell-dependent form of peste des petits ruminants virus: a system for making biosafe antigen. Vet Res 2015; 46:101. [PMID: 26396073 PMCID: PMC4579661 DOI: 10.1186/s13567-015-0231-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/29/2015] [Indexed: 11/10/2022] Open
Abstract
Peste des petits ruminants (PPR) is a viral disease of sheep and goats that is spreading through many countries in the developing world. Work on the virus is often restricted to studies of attenuated vaccine strains or to work in laboratories that have high containment facilities. We have created a helper cell dependent form of PPR virus by removing the entire RNA polymerase gene and complementing it with polymerase made constitutively in a cell line. The resultant L-deleted virus grows efficiently in the L-expressing cell line but not in other cells. Virus made with this system is indistinguishable from normal virus when used in diagnostic assays, and can be grown in normal facilities without the need for high level biocontainment. The L-deleted virus will thus make a positive contribution to the control and study of this important disease.
Collapse
Affiliation(s)
- Jana Baron
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| | - Michael D Baron
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| |
Collapse
|
35
|
Tang HB, Lu ZL, Wei XK, Zhong YZ, Zhong TZ, Pan Y, Luo Y, Liao SH, Minamoto N, Luo TR. A recombinant rabies virus expressing a phosphoprotein-eGFP fusion is rescued and applied to the rapid virus neutralization antibody assay. J Virol Methods 2015; 219:75-83. [PMID: 25845623 DOI: 10.1016/j.jviromet.2015.03.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 02/12/2015] [Accepted: 03/28/2015] [Indexed: 12/25/2022]
Abstract
Rabies remains a worldwide concern, and dogs are a major vector for rabies virus (RABV) transmission. Vaccination is used in China to control the spread of rabies in dogs, a practice which necessitates effective, efficient, and high-throughput methods to confirm vaccination. The current rapid fluorescent focus inhibition test (RFFIT) method to measure virus-neutralizing antibody titers in the serum involves multiple steps, and more efficient methods are needed to match the increasing demand for this type of monitoring. In this study, based on the parental rRC-HL strain, a recombinant RABV rRV-eGFP expressing enhanced green fluorescent protein (eGFP) fused with RABV P protein was generated by a reverse genetic technique. The rRV-eGFP grew stably and successfully expressed P-eGFP fusion in Neuro-2A (NA) host cells. Furthermore, the P protein was shown to co-localize with eGFP in rRV-eGFP-infected NA cells. Since eGFP is easily detected in infected cells under a fluorescence microscope, rRV-eGFP could be used to establish a more rapid virus-neutralizing antibody titers assay based on RFFIT, designated as the RFFIT-eGFP method. From 69 canine serum samples, the RFFIT-eGFP method was shown to be as specific and as sensitive as the RFFIT method, suggesting that it might represent a faster tool than conventional RFFIT for measuring RABV virus-neutralizing antibody titers in canine sera without sacrificing accuracy.
Collapse
Affiliation(s)
- Hai-Bo Tang
- The Key Laboratory of Ministry of Education for Microbial and Plant Genetic Engineering, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresourses, Guangxi University, Nanning 530004, Guangxi, China; Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Zhuan-Ling Lu
- The Key Laboratory of Ministry of Education for Microbial and Plant Genetic Engineering, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresourses, Guangxi University, Nanning 530004, Guangxi, China; Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Xian-Kai Wei
- The Key Laboratory of Ministry of Education for Microbial and Plant Genetic Engineering, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresourses, Guangxi University, Nanning 530004, Guangxi, China; Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Yi-Zhi Zhong
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Tao-Zhen Zhong
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Yan Pan
- The Key Laboratory of Ministry of Education for Microbial and Plant Genetic Engineering, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresourses, Guangxi University, Nanning 530004, Guangxi, China; Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Yang Luo
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Su-Huan Liao
- The Key Laboratory of Ministry of Education for Microbial and Plant Genetic Engineering, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresourses, Guangxi University, Nanning 530004, Guangxi, China; Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Nobuyuki Minamoto
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Ting Rong Luo
- The Key Laboratory of Ministry of Education for Microbial and Plant Genetic Engineering, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresourses, Guangxi University, Nanning 530004, Guangxi, China; Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
36
|
|
37
|
Muniraju M, Mahapatra M, Buczkowski H, Batten C, Banyard AC, Parida S. Rescue of a vaccine strain of peste des petits ruminants virus: In vivo evaluation and comparison with standard vaccine. Vaccine 2014; 33:465-71. [PMID: 25444790 PMCID: PMC4315076 DOI: 10.1016/j.vaccine.2014.10.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 10/17/2014] [Accepted: 10/24/2014] [Indexed: 11/17/2022]
Abstract
Rescue of a vaccine strain of peste des petits ruminants virus. In vivo evaluation of rescued vaccine strain and comparison with standard vaccine. 1SStrategy for Differentiating Infected from Vaccinated Animals (DIVA).
Across the developing world peste des petits ruminants virus places a huge disease burden on agriculture, primarily affecting the production of small ruminant. The disease is most effectively controlled by vaccinating sheep and goats with live attenuated vaccines that provide lifelong immunity. However, the current vaccines and serological tests are unable to enable Differentiation between naturally Infected and Vaccinated Animals (DIVA). This factor precludes meaningful assessment of vaccine coverage and epidemiological surveillance based on serology, in turn reducing the efficiency of control programmes. The availability of a recombinant PPRV vaccine with a proven functionality is a prerequisite for the development of novel vaccines that may enable the development of DIVA tools for PPRV diagnostics. In this study, we have established an efficient reverse genetics system for PPRV Nigeria 75/1 vaccine strain and, further rescued a version of PPRV Nigeria 75/1 vaccine strain that expresses eGFP as a novel transcription cassette and a version of PPRV Nigeria 75/1 vaccine strain with mutations in the haemagglutinin (H) gene to enable DIVA through disruption of binding to H by the C77 monoclonal antibody used in the competitive (c) H-ELISA. All three rescued viruses showed similar growth characteristics in vitro in comparison to parent vaccine strain and, following in vivo assessment the H mutant provided full protection in goats. Although the C77 monoclonal antibody used in the cH-ELISA was unable to bind to the mutated form of H in vitro, the mutation was not sufficient to enable DIVA in vivo.
Collapse
Affiliation(s)
- Murali Muniraju
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK
| | - Mana Mahapatra
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK
| | | | - Carrie Batten
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK
| | | | - Satya Parida
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
| |
Collapse
|
38
|
Liu F, Wu X, Liu W, Li L, Wang Z. Current perspectives on conventional and novel vaccines against peste des petits ruminants. Vet Res Commun 2014; 38:307-22. [DOI: 10.1007/s11259-014-9618-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/05/2014] [Indexed: 10/24/2022]
|
39
|
Kumar N, Maherchandani S, Kashyap SK, Singh SV, Sharma S, Chaubey KK, Ly H. Peste des petits ruminants virus infection of small ruminants: a comprehensive review. Viruses 2014; 6:2287-327. [PMID: 24915458 PMCID: PMC4074929 DOI: 10.3390/v6062287] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/26/2014] [Accepted: 05/28/2014] [Indexed: 12/14/2022] Open
Abstract
Peste des petits ruminants (PPR) is caused by a Morbillivirus that belongs to the family Paramyxoviridae. PPR is an acute, highly contagious and fatal disease primarily affecting goats and sheep, whereas cattle undergo sub-clinical infection. With morbidity and mortality rates that can be as high as 90%, PPR is classified as an OIE (Office International des Epizooties)-listed disease. Considering the importance of sheep and goats in the livelihood of the poor and marginal farmers in Africa and South Asia, PPR is an important concern for food security and poverty alleviation. PPR virus (PPRV) and rinderpest virus (RPV) are closely related Morbilliviruses. Rinderpest has been globally eradicated by mass vaccination. Though a live attenuated vaccine is available against PPR for immunoprophylaxis, due to its instability in subtropical climate (thermo-sensitivity), unavailability of required doses and insufficient coverage (herd immunity), the disease control program has not been a great success. Further, emerging evidence of poor cross neutralization between vaccine strain and PPRV strains currently circulating in the field has raised concerns about the protective efficacy of the existing PPR vaccines. This review summarizes the recent advancement in PPRV replication, its pathogenesis, immune response to vaccine and disease control. Attempts have also been made to highlight the current trends in understanding the host susceptibility and resistance to PPR.
Collapse
Affiliation(s)
- Naveen Kumar
- Virology Laboratory, Division of Animal Health, Central Institute for Research on Goats, Makhdoom, P.O. Farah, Mathura, UP 281122, India.
| | - Sunil Maherchandani
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, Rajasthan 334001, India.
| | - Sudhir Kumar Kashyap
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, Rajasthan 334001, India.
| | - Shoor Vir Singh
- Virology Laboratory, Division of Animal Health, Central Institute for Research on Goats, Makhdoom, P.O. Farah, Mathura, UP 281122, India.
| | - Shalini Sharma
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India.
| | - Kundan Kumar Chaubey
- Virology Laboratory, Division of Animal Health, Central Institute for Research on Goats, Makhdoom, P.O. Farah, Mathura, UP 281122, India.
| | - Hinh Ly
- Veterinary and Biomedical Sciences Department, University of Minnesota, 1988 Fitch Ave., Ste 295, Saint Paul, MN 55108, USA.
| |
Collapse
|
40
|
Yin C, Chen W, Hu Q, Wen Z, Wang X, Ge J, Yin Q, Zhi H, Xia C, Bu Z. Induction of protective immune response against both PPRV and FMDV by a novel recombinant PPRV expressing FMDV VP1. Vet Res 2014; 45:62. [PMID: 24898430 PMCID: PMC4059095 DOI: 10.1186/1297-9716-45-62] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 05/27/2014] [Indexed: 11/17/2022] Open
Abstract
Peste des petits ruminants (PPR) and foot-and-mouth disease (FMD) are both highly contagious diseases of small domestic and wild ruminants caused by the PPR virus (PPRV) and the FMD virus (FMDV). In this study, a recombinant PPRV expressing the FMDV VP1 gene (rPPRV/VP1) was generated and FMDV VP1 expression did not impair replication of the recombinant virus in vitro and immunogenicity in inducing neutralizing antibody against PPR in goats. Vaccination with one dose of rPPRV/VP1 induced FMDV neutralizing antibody in goats and protected them from challenge with virulent FMDV. Our results suggest that the recombinant PPRV expressing the FMDV VP1 protein is a potential dual live vectored vaccine against PPRV and FMDV.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chun Xia
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China.
| | | |
Collapse
|
41
|
Xue X, Zheng X, Liang H, Feng N, Zhao Y, Gao Y, Wang H, Yang S, Xia X. Generation of recombinant rabies Virus CVS-11 expressing eGFP applied to the rapid virus neutralization test. Viruses 2014; 6:1578-89. [PMID: 24714411 PMCID: PMC4014711 DOI: 10.3390/v6041578] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 02/26/2014] [Accepted: 03/13/2014] [Indexed: 12/25/2022] Open
Abstract
The determination of levels of rabies virus-neutralizing antibody (VNA) provides the foundation for the quantitative evaluation of immunity effects. The traditional fluorescent antibody virus neutralization test (FAVN) using a challenge virus standard (CVS)-11 strain as a detection antigen and staining infected cells with a fluorescein isothiocyanate (FITC)-labeled monoclonal antibody, is expensive and high-quality reagents are often difficult to obtain in developing countries. Indeed, it is essential to establish a rapid, economical, and specific rabies virus neutralization test (VNT). Here, we describe a recombinant virus rCVS-11-eGFP strain that stably expresses enhanced green fluorescent protein (eGFP) based on a reverse genetic system of the CVS-11 strain. Compared to the rCVS-11 strain, the rCVS-11-eGFP strain showed a similar growth property with passaging stability in vitro and pathogenicity in vivo. The rCVS-11-eGFP strain was utilized as a detection antigen to determine the levels of rabies VNAs in 23 human and 29 canine sera; this technique was termed the FAVN-eGFP method. The good reproducibility of FAVN-eGFP was tested with partial serum samples. Neutralization titers obtained from FAVN and FAVN-eGFP were not significantly different. The FAVN-eGFP method allows rapid economical, specific, and high-throughput assessment for the titration of rabies VNAs.
Collapse
Affiliation(s)
- Xianghong Xue
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Xuexing Zheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Hongru Liang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Na Feng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Yongkun Zhao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Yuwei Gao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Hualei Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Songtao Yang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| | - Xianzhu Xia
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun 130122, China.
| |
Collapse
|
42
|
Buczkowski H, Muniraju M, Parida S, Banyard AC. Morbillivirus vaccines: recent successes and future hopes. Vaccine 2014; 32:3155-61. [PMID: 24703852 PMCID: PMC7115685 DOI: 10.1016/j.vaccine.2014.03.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/05/2014] [Accepted: 03/13/2014] [Indexed: 01/21/2023]
Abstract
Morbilliviruses cause severe disease in both human and animal populations. Morbilliviruses are recognised targets for eradication. Live attenuated vaccines are available for some morbilliviruses. DIVA vaccines may be important for future morbillivirus eradication attempts.
The impact of morbilliviruses on both human and animal populations is well documented in the history of mankind. Indeed, prior to the development of vaccines for these diseases, morbilliviruses plagued both humans and their livestock that were heavily relied upon for food and motor power within communities. Measles virus (MeV) was responsible for the death of millions of people annually across the world and those fortunate enough to escape the disease often faced starvation where their livestock had died following infection with rinderpest virus (RPV) or peste des petits ruminants virus (PPRV). Canine distemper virus has affected dog populations for centuries and in the past few decades appears to have jumped species, now causing disease in a number of non-canid species, some of which are been pushed to the brink of extinction by the virus. During the age of vaccination, the introduction and successful application of vaccines against rinderpest and measles has led to the eradication of the former and the greater control of the latter. Vaccines against PPR and canine distemper have also been generated; however, the diseases still pose a threat to susceptible species. Here we review the currently available vaccines against these four morbilliviruses and discuss the prospects for the development of new generation vaccines.
Collapse
Affiliation(s)
- Hubert Buczkowski
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, Weybridge, Surrey, KT15 3NB, United Kingdom
| | - Murali Muniraju
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, United Kingdom
| | - Satya Parida
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, United Kingdom
| | - Ashley C Banyard
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, Weybridge, Surrey, KT15 3NB, United Kingdom.
| |
Collapse
|
43
|
Eschbaumer M, Law S, Solis C, Chernick A, van der Meer F, Czub M. Rapid detection of neutralizing antibodies against bovine viral diarrhoea virus using quantitative high-content screening. J Virol Methods 2014; 198:56-63. [DOI: 10.1016/j.jviromet.2013.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 11/26/2013] [Accepted: 12/17/2013] [Indexed: 11/29/2022]
|
44
|
Falzarano D, Groseth A, Hoenen T. Development and application of reporter-expressing mononegaviruses: current challenges and perspectives. Antiviral Res 2014; 103:78-87. [PMID: 24462694 DOI: 10.1016/j.antiviral.2014.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 01/06/2014] [Accepted: 01/12/2014] [Indexed: 12/19/2022]
Abstract
Reverse genetics allows the generation of recombinant viruses entirely from cDNA. One application of this technology is the creation of reporter-expressing viruses, which greatly increase the detail and ease with which these viruses can be studied. However, there are a number of challenges when working with reporter-expressing viruses. Both the reporter protein itself as well as the genetic manipulations within the viral genome required for expression of this reporter can result in altered biological properties of the recombinant virus, and lead to attenuation in vitro and/or in vivo. Further, instability of reporter expression and purging of the genetic information encoding for the reporter from the viral genome can be an issue. Finally, a practical challenge for in vivo studies lies in the attenuation of light signals when traversing tissues. Novel expression strategies and the continued development of brighter, red and far-red shifted reporters and the increased use of bioluminescent reporters for in vivo applications promise to overcome some of these limitations in future. However, a "one size fits all" approach to the design of reporter-expressing viruses has thus far not been possible. Rather, a reporter suited to the intended application must be selected and an appropriate expression strategy and location for the reporter in the viral genome chosen. Still, attenuating effects of the reporter on viral fitness are difficult to predict and have to be carefully assessed with respect to the intended application. Despite these limitations the generation of suitable reporter-expressing viruses will become more common as technology and our understanding of the intricacies of viral gene expression and regulation improves, allowing deeper insight into virus biology both in living cells and in animals.
Collapse
Affiliation(s)
- Darryl Falzarano
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Allison Groseth
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Thomas Hoenen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
45
|
A novel porcine reproductive and respiratory syndrome virus vector system that stably expresses enhanced green fluorescent protein as a separate transcription unit. Vet Res 2013; 44:104. [PMID: 24176053 PMCID: PMC4176086 DOI: 10.1186/1297-9716-44-104] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 10/21/2013] [Indexed: 11/10/2022] Open
Abstract
Here we report the rescue of a recombinant porcine reproductive and respiratory syndrome virus (PRRSV) carrying an enhanced green fluorescent protein (EGFP) reporter gene as a separate transcription unit. A copy of the transcription regulatory sequence for ORF6 (TRS6) was inserted between the N protein and 3′-UTR to drive the transcription of the EGFP gene and yield a general purpose expression vector. Successful recovery of PRRSV was obtained using an RNA polymerase II promoter to drive transcription of the full-length virus genome, which was assembled in a bacterial artificial chromosome (BAC). The recombinant virus showed growth replication characteristics similar to those of the wild-type virus in the infected cells. In addition, the recombinant virus stably expressed EGFP for at least 10 passages. EGFP expression was detected at approximately 10 h post infection by live-cell imaging to follow the virus spread in real time and the infection of neighbouring cells occurred predominantly through cell-to-cell-contact. Finally, the recombinant virus generated was found to be an excellent tool for neutralising antibodies and antiviral compound screening. The newly established reverse genetics system for PRRSV could be a useful tool not only to monitor virus spread and screen for neutralising antibodies and antiviral compounds, but also for fundamental research on the biology of the virus.
Collapse
|
46
|
Abstract
We have created a completely helper cell-dependent morbillivirus by modifying the genome to remove the coding sequence of the phosphoprotein (P) and recovering the recombinant virus in a cell line constitutively expressing the P protein. The P protein-deleted virus (P−) grew very inefficiently unless both of the viral accessory proteins (V and C) were also expressed. Growth of the virus was restricted to the P-expressing cell line. The P− virus grew more slowly than the parental virus and expressed much less viral protein in infected cells. The technique could be used to create virus-like particles for use as a vaccine or as antigen in immunological or serological assays.
Collapse
Affiliation(s)
- Jana Baron
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK
| | - Michael Baron
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK
| |
Collapse
|
47
|
Zhang Y, Wu S, Lv J, Feng C, Deng J, Wang C, Yuan X, Zhang T, Lin X. Peste des petits ruminants virus exploits cellular autophagy machinery for replication. Virology 2013; 437:28-38. [PMID: 23318276 DOI: 10.1016/j.virol.2012.12.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/10/2012] [Accepted: 12/17/2012] [Indexed: 12/19/2022]
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. Although PPRV is known to induce apoptosis in infected cells, the interaction between PPRV and permissive cells requires further elucidation. Here, we provide the first evidence that PPRV infection triggered autophagy in Vero cells based on the appearance of abundant double- and single-membrane vesicles, the accumulation of LC3 fluorescent puncta, the enhancement of LC3-I/-II conversion, and autophagic flux. We further demonstrated that induction of autophagy with rapamycin significantly increased PPRV progeny yield and nucleocapsid (N) protein expression, while inhibition of autophagy with siRNA targeting ATG7 resulted in diametrically opposite results. Our data indicate that PPRV exploits the autophagy machinery to facilitate its own replication in host cells, thus the production efficiency of live attenuated PPRV vaccines may be improved by targeting the autophagic pathway.
Collapse
Affiliation(s)
- Yongning Zhang
- Institute of Animal Quarantine, Chinese Academy of Inspection and Quarantine, Beijing 100029, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Albina E, Kwiatek O, Minet C, Lancelot R, Servan de Almeida R, Libeau G. Peste des Petits Ruminants, the next eradicated animal disease? Vet Microbiol 2012; 165:38-44. [PMID: 23313537 DOI: 10.1016/j.vetmic.2012.12.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 12/03/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
Abstract
Peste des Petits Ruminants (PPR) is a widespread viral disease caused by a Morbillivirus (Paramyxoviridae). There is a single serotype of PPR virus, but four distinct genetic lineages. Morbidity and mortality are high when occurring in naive sheep and goats populations. Cattle and African buffaloes (Syncerus caffer) are asymptomatically infected. Other wild ruminants and camels may express clinical signs and mortality. PPR has recently spread in southern and northern Africa, and in central and far-east Asia. More than one billion sheep and goats worldwide are at risk. PPR is also present in Europe through western Turkey. Because of its clinical incidence and the restrictions on animal movements, PPR is a disease of major economic importance. A live attenuated vaccine was developed in the 1980s, and has been widely used in sheep and goats. Current researches aim (i) to make it more thermotolerant for use in countries with limited cold chain, and (ii) to add a DIVA mark to shorten and reduce the cost of final eradication. Rinderpest virus-another Morbillivirus-was the first animal virus to be eradicated from Earth. PPRV has been proposed as the next candidate. Considering its wide distribution and its multiple target host species which have an intense mobility, it will be a long process that cannot exclusively rely on mass vaccination. PPR specific epidemiological features and socio-economic considerations will also have to be taken into account, and sustained international, coordinated, and funded strategy based on a regional approach of PPR control will be the guarantee toward success.
Collapse
|