1
|
Zou H, Niu Z, Cheng P, Wu C, Li W, Luo G, Huang S. Structure, Attachment and Transmembrane Internalisation of Peste Des Petits Ruminants Virus. Vet Med Sci 2025; 11:e70182. [PMID: 39739994 DOI: 10.1002/vms3.70182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/19/2024] [Accepted: 12/08/2024] [Indexed: 01/02/2025] Open
Abstract
Peste des petits ruminants virus (PPRV), a single-stranded negative-sense RNA virus with an envelope, belongs to the Morbillivirus in the Paramyxoviridae family and is prevalent worldwide. PPRV infection causes fever, stomatitis, diarrhoea, pneumonia, abortion and other symptoms in small ruminants, with a high mortality rate that poses a significant threat to the sustainability and productivity of the small ruminant livestock sector. The PPRV virus particles have a diameter of approximately 400-500 nm and are composed of six structural proteins: nucleocapsid protein (N), phosphoprotein (P), envelope matrix protein (M), fusion protein (F), haemagglutinin protein (H) and large protein (L). Each protein has a distinct role in the virus's life cycle. Although the life cycle activities of PPRV have been widely reported, they are still limited. Research has demonstrated that PPRV has distinct adhesion factors on various cell surfaces, such as the epithelial cell adhesion factor nectin-4 or the lymphocyte adhesion factor SLAM. After attaching to the cell, the F and H proteins on the PPRV membrane interact with each other, resulting in a conformational change in the F protein. This change allows the F protein to enter the cell through direct fusion with the host cell membrane. The virus enters the host cell via the outer vesicle endocytosis strategy and replicates and proliferates through the role of caveolin, actin, dynein and cholesterol on the host cell membrane. This review summarises the viral structure, attachment mechanism and transmembrane internalisation mechanism of PPRV. The aim of this review is to provide theoretical support for the development of PPRV inhibitors and the prevention and control of PPR.
Collapse
Affiliation(s)
- Hong Zou
- Chongqing Three Gouges Vocational College, College of Animal Science & Technology, Wanzhou, China
| | - Zheng Niu
- College of Veterinary Medicine, Northwest A & F University, Yangling, China
| | - Peng Cheng
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Chunxia Wu
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Wenjie Li
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Gan Luo
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Shilei Huang
- Chongqing Three Gouges Vocational College, College of Animal Science & Technology, Wanzhou, China
| |
Collapse
|
2
|
Li L, Li S, Han S, Li P, Du G, Wu J, Cao X, Shang Y. Inhibition of caspase-1-dependent apoptosis suppresses peste des petits ruminants virus replication. J Vet Sci 2023; 24:e55. [PMID: 37638708 PMCID: PMC10556287 DOI: 10.4142/jvs.22288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/10/2023] [Accepted: 05/26/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Peste des petits ruminants (PPR), caused by the PPR virus (PPRV), is an acute and fatal contagious disease that mainly infects goats, sheep, and other artiodactyls. Peripheral blood mononuclear cells (PBMCs) are considered the primary innate immune cells. OBJECTIVES PBMCs derived from goats were infected with PPRV and analyzed to detect the relationship between PPRV replication and apoptosis or the inflammatory response. METHODS Quantitative real-time polymerase chain reaction was used to identify PPRV replication and cytokines expression. Flow cytometry was conducted to detect apoptosis and the differentiation of CD4+ and CD8+ T cells after PPRV infection. RESULTS PPRV stimulated the differentiation of CD4+ and CD8+ T cells. In addition, PPRV induced apoptosis in goat PBMCs. Furthermore, apoptosis and the inflammatory response induced by PPRV could be suppressed by Z-VAD-FMK and Z-YVAD-FMK, respectively. Moreover, the virus titer of PPRV was attenuated by inhibiting caspase-1-dependent apoptosis and inflammation. CONCLUSIONS This study showed that apoptosis and the inflammatory response play an essential role in PPR viral replication in vitro, providing a new mechanism related to the cell host response.
Collapse
Affiliation(s)
- Lingxia Li
- College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China
- College of Animal Science and Veterinary Science, Qinghai University; Qinghai Key Laboratory of Pathogen Diagnosis and Green Prevention and Control Technology for Animal Diseases, Xining 810016, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Shengqing Li
- College of Animal Science and Veterinary Science, Qinghai University; Qinghai Key Laboratory of Pathogen Diagnosis and Green Prevention and Control Technology for Animal Diseases, Xining 810016, China
| | - Shengyi Han
- College of Animal Science and Veterinary Science, Qinghai University; Qinghai Key Laboratory of Pathogen Diagnosis and Green Prevention and Control Technology for Animal Diseases, Xining 810016, China
| | - Pengfei Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Guoyu Du
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Jinyan Wu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Xiaoan Cao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Youjun Shang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China.
| |
Collapse
|
3
|
Kumar A, Sodhi M, Mukesh M, Kaur A, Bhakri G, Chaudhary V, Swami P, Sharma V, Mohanty AK, Kataria RS. Identification of stably expressed Internal Control Genes (ICGs) for normalization of expression data in liver of C57BL/6 mice injected with beta casomorphins. PLoS One 2023; 18:e0282994. [PMID: 37145997 PMCID: PMC10162558 DOI: 10.1371/journal.pone.0282994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/28/2023] [Indexed: 05/07/2023] Open
Abstract
In recent years, beta-casomorphin peptides (BCM7/BCM9) derived from the digestion of cow milk have drawn a lot of attention world over because of their proposed impact on human health. In order to evaluate the transcriptional modulation of target genes through RT-qPCR in response to these peptides, availability of appropriate reference or internal control genes (ICGs) will be the key. The present study was planned to identify a panel of stable ICGs in the liver tissue of C57BL/6 mice injected with BCM7/BCM9 cow milk peptides for 3 weeks. A total of ten candidate genes were evaluated as potential ICGs by assessing their expression stability using software suites; geNorm, NormFinder and BestKeeper. The suitability of the identified ICGs was validated by assessing the relative expression levels of target genes, HP and Cu/Zn SOD. Based on geNorm, PPIA and SDHA gene pair was identified to be most stably expressed in liver tissue during the animal trials. Similarly, NormFinder analysis also identified PPIA as the most stable gene. BestKeeper analysis showed crossing point SD value for all the genes in the acceptable range that is closer to 1. Overall, the study identified a panel of stable ICGs for reliable normalization of target genes expression data in mice liver tissues during BCM7/9 peptides trial.
Collapse
Affiliation(s)
- Anurag Kumar
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - Monika Sodhi
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Manishi Mukesh
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Amandeep Kaur
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Gaurav Bhakri
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Vipul Chaudhary
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Preeti Swami
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Vishal Sharma
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Ashok Kumar Mohanty
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - Ranjit S Kataria
- Animal Biotechnology Division, National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| |
Collapse
|
4
|
Li L, Wu J, Cao X, He J, Liu X, Shang Y. Analysis and Sequence Alignment of Peste des Petits Ruminants Virus ChinaSX2020. Vet Sci 2021; 8:vetsci8110285. [PMID: 34822658 PMCID: PMC8623451 DOI: 10.3390/vetsci8110285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
The peste des petits ruminants virus (PPRV) mainly infects goats and sheep and causes a highly contagious disease, PPR. Recently, a PPRV strain named ChinaSX2020 was isolated and confirmed following an indirect immunofluorescence assay and PCR using PPRV-specific antibody and primers, respectively. A sequencing of the ChinaSX2020 strain showed a genome length of 15,954 nucleotides. A phylogenetic tree analysis showed that the ChinaSX2020 genome was classified into lineage IV of the PRRV genotypes. The genome of the ChinaSX2020 strain was found to be closely related to PPRVs isolated in China between 2013 and 2014. These findings revealed that not a variety of PRRVs but similar PPRVs were continuously spreading and causing sporadic outbreaks in China.
Collapse
|
5
|
Eloiflin RJ, Auray G, Python S, Rodrigues V, Seveno M, Urbach S, El Koulali K, Holzmuller P, Totte P, Libeau G, Bataille A, Summerfield A. Identification of Differential Responses of Goat PBMCs to PPRV Virulence Using a Multi-Omics Approach. Front Immunol 2021; 12:745315. [PMID: 34671358 PMCID: PMC8521192 DOI: 10.3389/fimmu.2021.745315] [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: 07/21/2021] [Accepted: 09/14/2021] [Indexed: 12/02/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease of small ruminants, mainly sheep and goats. Host susceptibility varies considerably depending on the PPR virus (PPRV) strain, the host species and breed. The effect of strains with different levels of virulence on the modulation of the immune system has not been thoroughly compared in an experimental setting so far. In this study, we used a multi-omics approach to investigate the host cellular factors involved in different infection phenotypes. Peripheral blood mononuclear cells (PBMCs) from Saanen goats were activated with a T-cell mitogen and infected with PPRV strains of different virulence: Morocco 2008 (high virulence), Ivory Coast 1989 (low virulence) and Nigeria 75/1 (live attenuated vaccine strain). Our results showed that the highly virulent strain replicated better than the other two in PBMCs and rapidly induced cell death and a stronger inhibition of lymphocyte proliferation. However, all the strains affected lymphocyte proliferation and induced upregulation of key antiviral genes and proteins, meaning a classical antiviral response is orchestrated regardless of the virulence of the PPRV strain. On the other hand, the highly virulent strain induced stronger inflammatory responses and activated more genes related to lymphocyte migration and recruitment, and inflammatory processes. Both transcriptomic and proteomic approaches were successful in detecting viral and antiviral effectors under all conditions. The present work identified key immunological factors related to PPRV virulence in vitro.
Collapse
Affiliation(s)
- Roger-Junior Eloiflin
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Gaël Auray
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sylvie Python
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Valérie Rodrigues
- ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France.,CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Petit-Bourg, France
| | - Martial Seveno
- BCM (BioCampus Montpellier), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Serge Urbach
- IGF (Institut de Génomique Fonctionnelle), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Khadija El Koulali
- BCM (BioCampus Montpellier), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Philippe Holzmuller
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Philippe Totte
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Genevieve Libeau
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Arnaud Bataille
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Artur Summerfield
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Liu W, Feng Q, Liao W, Li E, Wu L. TUG1 promotes the expression of IFITM3 in hepatocellular carcinoma by competitively binding to miR-29a. J Cancer 2021; 12:6905-6920. [PMID: 34659578 PMCID: PMC8517998 DOI: 10.7150/jca.57477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/13/2021] [Indexed: 02/07/2023] Open
Abstract
Purpose: Numerous studies have demonstrated the important relationship of TUG1 with tumorigenesis. The present study investigated the role of TUG1 and its downstream genes miR-29a and IFITM3 in the occurrence and development of hepatocellular carcinoma (HCC). We found that both TUG1 and IFITM3 genes are highly expressed in HCC, whereas the expression of miR-29a is low in HCC. Downregulation of TUG1 reduces cell invasion, metastasis, and cell proliferation ability and promotes cell apoptosis. Simultaneous downregulation of miR-29a reverses this effect. Moreover, IFITM3, as the target gene of miR-29a, is positively regulated by TUG1. However, the adjustment relationship between these three components is still unknown and thus warrants further investigation. The objective of this study was to investigate the regulatory relationship between TUG1, miR-29a, and IFITM3 in human liver cancer. Patients and methods: The expression of TUG1 and miR-29a in tumor tissues and adjacent non-tumor tissues of 65 patients with HCC was detected by real-time quantitative polymerase chain reaction (RT-qPCR). The migration and invasion of liver cancer cells were studied by the wound healing assay and the Transwell method, respectively. The apoptosis rate of HCC cells was detected by flow cytometry, and the proliferation rate of hepatoma cells was detected by the 5-ethynyl-2'-deoxyuridine (EdU) method. Immunofluorescence was used to detect the expression of TUG1 and IFITM3 in HCC-LM3 and HL-7702 cell lines. The relationship between TUG1 and miR-29a was detected using a double luciferase reporter assay and fluorescence in situ hybridization (FISH). Tumors were established in vivo by subcutaneous injection of HCC cells into nude mice and injection of these cells into the tail vein. Western blotting was used to quantify the biomarkers. Results: The expression of TUG1 increased significantly in tumor tissues and HCC cells. Moreover, the expression of miR-29a in liver cancer tissues was significantly lower than that in normal human liver tissues. The expression of TUG1 in liver cancer tissue was negatively correlated with miR-29a. Knockdown of TUG1 weakened the invasion, migration, and proliferation of HCC cells, and enhanced their apoptosis. A simultaneous knockdown of miR-29a enhanced cell invasion, metastasis, and cell proliferation, whereas the apoptosis ability decreased. As a target gene of miR-29a, IFITM3 is not only negatively regulated by miR-29a, but also positively regulated by TUG1. Therefore, TUG1 regulates IFITM3 in HCC cells by competitively binding to miR-29a, thus affecting cell invasion, migration, proliferation, and apoptosis. Conclusion: As a CeRNA, TUG1 competitively binds to miR-29a to regulate IFITM3 and promote the development of liver cancer. Downregulation of TUG1 can significantly inhibit the migration, invasion, and proliferation of liver cancer cells. Based on these results, we conclude that TUG1 could serve as a key gene to improve the prognosis of patients with HCC.
Collapse
Affiliation(s)
| | | | | | - Enliang Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Nanchang University, 1 Mindle Road, Nanchang Jiangxi 330006, P.R. China
| | - Linquan Wu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Nanchang University, 1 Mindle Road, Nanchang Jiangxi 330006, P.R. China
| |
Collapse
|
7
|
Abstract
Immune response is a highly coordinated cascade involving all the subsets of peripheral blood mononuclear cells (PBMCs). In this study, RNA sequencing (RNA-Seq) analysis of PBMC subsets was done to delineate the systems biology behind immune protection of the vaccine in sheep and goats. The PBMC subsets studied were CD4+, CD8+, CD14+, CD21+, and CD335+ cells from day 0 and day 5 of sheep and goats vaccinated with Sungri/96 peste des petits ruminants virus. Assessment of the immune response processes enriched by the differentially expressed genes (DEGs) in all the subsets suggested a strong dysregulation toward the development of early inflammatory microenvironment, which is very much required for differentiation of monocytes to macrophages, and activation as well as the migration of dendritic cells into the draining lymph nodes. The protein-protein interaction networks among the antiviral molecules (IFIT3, ISG15, MX1, MX2, RSAD2, ISG20, IFIT5, and IFIT1) and common DEGs across PBMC subsets in both species identified ISG15 to be a ubiquitous hub that helps in orchestrating antiviral host response against peste des petits ruminants virus (PPRV). IRF7 was found to be the key master regulator activated in most of the subsets in sheep and goats. Most of the pathways were found to be inactivated in B lymphocytes of both the species, indicating that 5 days postvaccination (dpv) is too early a time point for the B lymphocytes to react. The cell-mediated immune response and humoral immune response pathways were found more enriched in goats than in sheep. Although animals from both species survived the challenge, a contrast in pathway activation was observed in CD335+ cells. IMPORTANCE Peste des petits ruminants (PPR) by PPR virus (PPRV) is an World Organisation for Animal Health (OIE)-listed acute, contagious transboundary viral disease of small ruminants. The attenuated Sungri/96 PPRV vaccine used all over India against this PPR provides long-lasting robust innate and adaptive immune response. The early antiviral response was found mediated through type I interferon-independent interferon-stimulated gene (ISG) expression. However, systems biology behind this immune response is unknown. In this study, in vivo transcriptome profiling of PBMC subsets (CD4+, CD8+, CD14+, CD21+, and CD335+) in vaccinated goats and sheep (at 5 days postvaccination) was done to understand this systems biology. Though there are a few differences in the systems biology across cells (specially the NK cells) between sheep and goats, the coordinated response that is inclusive of all the cell subsets was found to be toward the induction of a strong innate immune response, which is needed for an appropriate adaptive immune response.
Collapse
|
8
|
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: 2.2] [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
|