1
|
Wei X, Lu K, Chang Z, Guo H, Li Q, Yuan B, Liu C, Yang Z, Liu H. Genetic analyses and functional validation of ruminant SLAMs reveal potential hosts for PPRV. Vet Res 2025; 56:57. [PMID: 40103005 PMCID: PMC11916873 DOI: 10.1186/s13567-025-01489-w] [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: 09/05/2024] [Accepted: 12/05/2024] [Indexed: 03/20/2025] Open
Abstract
Peste des petits ruminants (PPR), caused by the peste des petits ruminants virus (PPRV), is a highly contagious disease affecting ruminants. While goats and sheep are well-known hosts, PPRV has also spread to wild ruminants, and it remains unclear which ruminant species can be infected. SLAM (Signaling lymphocytic activation molecule) acts as the primary receptor for PPRV, playing a crucial role in the viral infection process. Identifying which ruminant SLAMs can mediate PPRV infection is essential for understanding the potential hosts of PPRV, which is vital for effective eradication efforts. In this study, we first extracted 77 ruminant species' SLAM sequences from ruminant genome database. Based on these sequences, we predicted the structures of ruminant SLAMs. The analysis revealed that SLAM conformation is similar across ruminant species, and the potential PPRV H protein binding domain residues were conserved among SLAMs of these 77 species. Phylogenetic analysis of SLAM grouped ruminants into six families. We then selected representative SLAMs from each ruminant family to assess their role in PPRV infection. Our findings demonstrated that ruminant SLAMs efficiently mediated PPRV infection, with enhanced viral amplification observed in cells expressing SLAM from java mouse deer (Tragulidae) and goat (Bovidae), compared to cells expressing SLAM from white tailed deer (Cervidae) and giraffe (Giraffidae). These results underscore the need to consider a broader range of potential host populations beyond goat and sheep in efforts to prevent and eradicate PPRV.
Collapse
Affiliation(s)
- Xi Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhengwu Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hanwei Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Qinfeng Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Binxuan Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chen Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China.
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China.
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| |
Collapse
|
2
|
Arun PM, Rajasekhar R, Ravishankar C, Palekkodan H, Kanjirakkuzhiyil S, Somasekhar S, Maneesh KM. Molecular epidemiology of small ruminant morbillivirus (SRMV) isolates from field outbreaks in Kerala, India based on fusion (F) and nucleoprotein (N) gene. Virusdisease 2025; 36:104-113. [PMID: 40290771 PMCID: PMC12021742 DOI: 10.1007/s13337-024-00902-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/27/2024] [Indexed: 04/30/2025] Open
Abstract
Small ruminants contribute significantly to the animal husbandry economy. Peste des petits ruminants (PPR) is one of the major infectious diseases of small ruminants caused by small ruminant morbillivirus (SRMV) previously known as PPR virus-PPRV, a member of the genus Morbillivirus, which causes significant morbidity and mortality in affected population thereby disturb the economy of rural poor. The present study describes the molecular characterization and phylogenetic analysis of SRMV with complete nucleocapsid (N) and fusion (F) gene sequence. Phylogenetic analysis of the SRMV isolates revealed that, all the isolates shared a common ancestor with Tamil Nadu isolate and were grouped under lineage IV. Phylogenetic analysis also revealed that two genetic groups are circulating in Kerala and have recently evolved. Analysis of the F protein of SRMV showed two unique mutations (A18E and S430I) in Kerala isolates. Amino acid analysis of nucleoprotein revealed that most of the changes were in the C-C-terminal region. Four unique mutations were also observed in the nucleoprotein (NP) of the present SRMV isolates (I153V, A431V, R458M, and G461K). Among the 19 B cell epitopes identified on nucleoprotein, at least one amino acid variation was detected in four epitopes. These changes may affect the monoclonal antibody-based diagnostic assays. These changes in the F and N genes indicate the continuous emergence and circulation of new variants of the virus within the same geographical area. This is the first report on the molecular characterization of PPRV isolates based on full N and F genes from the Kerala state of India.
Collapse
Affiliation(s)
- P. M. Arun
- Department of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India
| | - Ravindran Rajasekhar
- Department of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India
| | - Chintu Ravishankar
- Department of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India
| | - Hamza Palekkodan
- Department of Veterinary Pathology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India
| | - Sumod Kanjirakkuzhiyil
- Department of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India
| | - Shashank Somasekhar
- Department of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India
| | - K. M. Maneesh
- Department of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India
| |
Collapse
|
3
|
Arun PM, Rajasekhar R, Ravishankar C, Palekkodan H, Kanjirakkuzhiyil S, Somasekhar S. Development of a SYBR Green-based RT-qPCR assay for the rapid detection of peste-des-petits-ruminants virus. J Vet Diagn Invest 2025; 37:278-283. [PMID: 39866047 PMCID: PMC11773506 DOI: 10.1177/10406387241311514] [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] [Indexed: 01/28/2025] Open
Abstract
Peste-des-petits-ruminants (PPR) is primarily a disease of small ruminants caused by peste-des-petits-ruminants virus (PPRV; Paramyxoviridae, Morbillivirus caprinae), formerly the small ruminant morbillivirus. PPRV can cause significant morbidity and mortality in small ruminants and a significant economic impact. Conventional reverse-transcription PCR (RT-PCR), and probe-based and SYBR Green-based RT quantitative real-time PCR (RT-qPCR), are employed for the molecular detection of PPRV. Here we describe a SYBR Green-based RT-qPCR for rapid and sensitive detection of PPRV. We designed the specific primers from the conserved region of the fusion gene (F) of PPRV. The standard curve of the established RT-qPCR assay had a good linear relationship. The developed assay was also 3 log units more sensitive than the conventional RT-PCR, with a detection limit of 13.6 copies and an efficiency of 98.2%. There was no cross-reactivity with other caprine respiratory viruses, namely bluetongue virus, goatpox virus, and orf virus. The positive detection rate of clinical samples was 11 of 64 (17.2%) versus 10 of 64 (15.6%) by conventional RT-PCR. We confirmed our results by sequencing the full F and N genes of the isolates. Our SYBR Green RT-qPCR can be used as a fast, economical, and sensitive alternative to RT-PCR for the detection of PPRV.
Collapse
Affiliation(s)
- Paravalappil Muraleedharan Arun
- Departments of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Kerala, India
| | - Ravindran Rajasekhar
- Departments of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Kerala, India
| | - Chintu Ravishankar
- Departments of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Kerala, India
| | - Hamza Palekkodan
- Veterinary Pathology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Kerala, India
| | - Sumod Kanjirakkuzhiyil
- Departments of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Kerala, India
| | - Shashank Somasekhar
- Departments of Veterinary Microbiology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Pookode, Kerala, India
| |
Collapse
|
4
|
Khulape SA, Choudhary SS, Jyotsana B, Prakash V, Rakshit S, Sahoo A. Synonymous codon usage influences the transmission of peste des petits ruminants (PPR) virus in camels. Vet Res Commun 2024; 48:4043-4049. [PMID: 39167257 DOI: 10.1007/s11259-024-10503-z] [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: 01/09/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Peste des petits ruminants virus (PPRV) is an infectious pathogen; causing highly contagious, acute febrile, and economically important disease of small ruminants. The virus is known to have intrinsic ability to adapt new hosts and to cross the species barrier. The incidence of PPR has already been reported in unusual host species such as camels, bovines, and wild animals from spill-over or natural infection. Still, there are elementary gaps in our knowledge of the extent of susceptibility of camel to PPRV and the adaptability of PPRV to camel. The present study delineates the potential role of preferential codon usage patterns responsible for adaptation, host immune evasion, and transmission of PPRV to unusual hosts like old world camel species namely, dromedary and bactrian camel. The results indicate codon usage of the PPRV genome is functioned by an interplay of mutational pressure and natural selection to exhort the adaptation and fitness of PPRV in probable hosts. The indices of natural selection like the relative codon deoptimization index (RCDI) and codon adaptation index (CAI) predict the ability of PPRV to adapt and evolve in camel species. The analysis also depicts the potential role of the CpG depletion mechanism employed by PPRV to evade host adaptive immune response. The report emphasizes the need for a comprehensive national PPR surveillance plan in unusual hosts like camels for the successful implementation of the PPR Global Eradication Programme (PPR- GEP).
Collapse
Affiliation(s)
| | | | - Basanti Jyotsana
- ICAR-National Research Centre on Camel, Bikaner, 334 001, Rajasthan, India
| | - Ved Prakash
- ICAR-National Research Centre on Camel, Bikaner, 334 001, Rajasthan, India
| | - Shantanu Rakshit
- ICAR-National Research Centre on Camel, Bikaner, 334 001, Rajasthan, India
| | - Artabandhu Sahoo
- ICAR-National Research Centre on Camel, Bikaner, 334 001, Rajasthan, India
| |
Collapse
|
5
|
Shi Y, Han D, Li J, Ye L, Ji X, Nie F, Song Z, Chen C, Ai J, Xin J. A novel quantitative real-time PCR with the GAPDH reference gene for peste des petits ruminants. VET MED-CZECH 2024; 69:234-242. [PMID: 39221119 PMCID: PMC11359978 DOI: 10.17221/123/2023-vetmed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 06/20/2024] [Indexed: 09/04/2024] Open
Abstract
Peste des petits ruminants (PPR) is a serious acute, highly contagious disease caused by the peste des petits ruminants virus (PPRV). This study aims to establish a qRT-PCR assay with an internal amplification control for the rapid and accurate detection of PPRV. The primers and probes for PPRV N were based on the national standard of the diagnostic techniques for PPR of China, and a pair of primers and TaqMan probes for the internal reference gene of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was designed. Optimisation of the reaction conditions, specificity, sensitivity and reproducibility tests, and clinical sample detection were conducted. The results showed that the optimal primers and probe concentrations of PPRV were 0.4 μmol/l and 0.4 μmol/l, respectively, and were 0.4 μmol/l and 0.2 μmol/l for the reference gene GAPDH, respectively. The established method has no cross-reaction with other viruses. The minimum detection limit was 6.8 copies/μl for PPRV and 190 copies/μl for GAPDH. The coefficients of variation (CV%) of PPRV and GAPDH were both lower than 2%. The results suggest that the PPRV qRT-PCR method containing internal reference genes has strong specificity, high sensitivity, and good reproducibility. The addition of internal reference genes for the sample quality control improves the accuracy of the detection.
Collapse
Affiliation(s)
- Yaling Shi
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, P.R. China
| | - Diangang Han
- Animal Quarantine Laboratory, Technology Center of Kunming Customs, Kunming, P.R. China
| | - Jing Li
- Animal Quarantine Laboratory, Technology Center of Kunming Customs, Kunming, P.R. China
| | - Lingling Ye
- Animal Quarantine Laboratory, Technology Center of Kunming Customs, Kunming, P.R. China
| | - Xincheng Ji
- Research Center for International Inspection and Quarantine Standard and Technical Regulation, General Administration of Customs, Beijing, P.R. China
| | - Fuping Nie
- Animal and Plant Quarantine Laboratory, Technology Center of Chongqing Customs, Chongqing, P.R. China
| | - Zhigang Song
- Research Center for International Inspection and Quarantine Standard and Technical Regulation, General Administration of Customs, Beijing, P.R. China
| | - Chaolin Chen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, P.R. China
| | - Jun Ai
- Animal Quarantine Laboratory, Technology Center of Kunming Customs, Kunming, P.R. China
| | - Jige Xin
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, P.R. China
| |
Collapse
|
6
|
Xu L, Ren J, Li L, Wang M, Zhu G, Zheng H, Zeng Q, Shang Y, Li D. Vimentin inhibits peste des petits ruminants virus replication by interaction with nucleocapsid protein. Virology 2024; 595:110056. [PMID: 38552409 DOI: 10.1016/j.virol.2024.110056] [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: 10/20/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 05/18/2024]
Abstract
The Peste des petits ruminant virus (PPRV) is a member of the Paramyxoviridae family and is classified into the genus Measles virus. PPRV predominantly infects small ruminants, leading to mortality rates of nearly 100%, which have caused significant economic losses in developing countries. Host proteins are important in virus replication, but the PPRV nucleocapsid (N) protein-host interacting partners for regulating PPRV replication remain unclear. The present study confirmed the interaction between PPRV-N and the host protein vimentin by co-immunoprecipitation and co-localization experiments. Overexpression of vimentin suppressed PPRV replication, whereas vimentin knockdown had the opposite effect. Mechanistically, N was subjected to degradation via the ubiquitin/proteasome pathway, where vimentin recruits the E3 ubiquitin ligase NEDD4L to fulfill N-ubiquitination, resulting in the degradation of the N protein. These findings suggest that the host protein vimentin and E3 ubiquitin ligase NEDD4L have an anti-PPRV effect.
Collapse
Affiliation(s)
- Long Xu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Jingjing Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Lingxia Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Mengyi Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Guoqiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Youjun Shang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China.
| | - Dan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China.
| |
Collapse
|
7
|
Zhang R, Hu Z, Wei D, Li R, Li Y, Zhang Z. Carboplatin restricts peste des petits ruminants virus replication by suppressing the STING-mediated autophagy. Front Vet Sci 2024; 11:1383927. [PMID: 38812563 PMCID: PMC11133560 DOI: 10.3389/fvets.2024.1383927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Peste des petits ruminants virus (PPRV) is a morbillivirus that causes the acute and highly pathogenic infectious disease peste des petits ruminants (PPR) in small ruminants and poses a major threat to the goat and sheep industries. Currently, there is no effective treatment for PPRV infection. Here, we propose Carboplatin, a platinum-based regimen designed to treat a range of malignancies, as a potential antiviral agent. We showed that Carboplatin exhibits significant antiviral activity against PPRV in a cell culture model. The mechanism of action of Carboplatin against PPRV is mainly attributed to its ability to block STING mediated autophagy. Together, our study supports the discovery of Carboplatin as an antiviral against PPRV and potentially other closely related viruses, sheds light on its mode of action, and establishes STING as a valid and attractive target to counteract viral infection.
Collapse
Affiliation(s)
| | | | | | | | - Yanmin Li
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, China
| | - Zhidong Zhang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Ayaz Kök S, Üstün S, Taşkent Sezgin H. Diagnosis of Ruminant Viral Diseases with Loop-Mediated Isothermal Amplification. Mol Biotechnol 2023; 65:1228-1241. [PMID: 36719638 PMCID: PMC9888337 DOI: 10.1007/s12033-023-00674-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023]
Abstract
Infectious diseases in livestock industry are major problems for animal health, food safety, and the economy. Zoonotic diseases from farm animals are significant threat to human population as well. These are notifiable diseases listed by the World Organization for Animal Health (OIE). Rapid diagnostic methods can help keep infectious diseases under control in herds. Loop-mediated isothermal amplification (LAMP) is a simple and rapid nucleic acid amplification method that is studied widely for detection of many infectious diseases in the field. LAMP allows biosensing of target DNA or RNA under isothermal conditions with high specificity in a short period of time. An untrained user can analyze results based on color change or turbidity. Here we review LAMP assays to diagnose OIE notifiable ruminant viral diseases in literature highlighting properties of LAMP method considering what is expected from an efficient, field usable diagnostic test.
Collapse
Affiliation(s)
- Sanem Ayaz Kök
- Biotechnology Interdisciplinary Program, İzmir Institute of Technology, Gülbahçe, Urla, İzmir, Turkey, 35430
- New Era Biotechnology, Teknopark İzmir, Gülbahçe, Urla, İzmir, Turkey, 35430
| | - Selcen Üstün
- Bioengineering Department, İzmir Institute of Technology, Gülbahçe, Urla, İzmir, Turkey, 35430
| | - Hümeyra Taşkent Sezgin
- Biotechnology Interdisciplinary Program, İzmir Institute of Technology, Gülbahçe, Urla, İzmir, Turkey, 35430.
- New Era Biotechnology, Teknopark İzmir, Gülbahçe, Urla, İzmir, Turkey, 35430.
- Bioengineering Department, İzmir Institute of Technology, Gülbahçe, Urla, İzmir, Turkey, 35430.
| |
Collapse
|
9
|
Rahman MM, Islam MS, Sabuj AAM, Hossain MG, Islam MA, Alam J, Ershaduzzaman M, Saha S. Molecular Epidemiology and Phylogenetic Analysis of Peste des Petits Ruminants Virus Circulating in Sheep in Bangladesh. Transbound Emerg Dis 2023; 2023:1175689. [PMID: 40303772 PMCID: PMC12016724 DOI: 10.1155/2023/1175689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 05/02/2025]
Abstract
Peste des petits ruminants (PPR) is a viral disease of small ruminants that is highly contagious, severe, reportable, and economically important. The present study was conducted to detect the PPR virus (PPRV) circulating in sheep in Bangladesh to determine its association with epidemiological risk factors and the degree of relationship between the F and H genes of the PPRV of sheep with those of other sheep and goat isolates. A cross-sectional study was conducted in five selected districts of Bangladesh to collect data on locations, ecological zones, breeds, age, sex, sources, time period, and farming systems using a structured questionnaire accompanied by face-to-face interviews. During sampling, 250 nasal swab samples were collected from live sheep with the typical clinical signs of PPR. Thereafter, a reverse-transcriptase polymerase chain reaction (RT-PCR) assay was employed to detect PPRV using the F and H genes. Risk factors were determined using bivariable and multivariable logistic regression analyses. Phylogenetic analysis of the detected PPRV was performed using MEGA software after sequencing both F and H genes. Using RT-PCR, 35.6% (89/250, 95% CI: 29.6%-41.6%) of the samples were found to be positive for PPRV. Locations, breeds, sources, and feeding systems were identified as potential molecular epidemiological risk factors for PPRV infection in a multivariate logistic regression model. Nucleotide sequencing and phylogenetic analysis showed that the PPRV strain was genetically related to the lineage IV virus isolates. For the F gene, the sequence divergence of our gene and other selected genes ranged from 0.01% to 0.018% within lineage IV, and the similarity ranged from 98.2% to 99.0%. In the case of the H gene, similar results were also observed in divergence, ranging from 0.017% to 0.083% among lineage IV and others, and similarity varied from 91.7% to 98.3%. To the best of our knowledge, this is the first study in Bangladesh conducted to determine the RT-PCR-based molecular epidemiology of PPRV in sheep. This study highlights the importance of establishing successful interventions for managing PPRV infections in small ruminants in Bangladesh.
Collapse
Affiliation(s)
- Mohammad Mojibur Rahman
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
- Bangladesh Civil Service Livestock Academy, Savar, Dhaka 1349, Bangladesh
| | - Md. Saiful Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Abdullah Al Momen Sabuj
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Md. Golzar Hossain
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Md. Alimul Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Jahangir Alam
- Animal Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Md. Ershaduzzaman
- Krishi Gobeshona Foundation, Bangladesh Agricultural Research Council, Farmgate, Dhaka 1215, Bangladesh
| | - Sukumar Saha
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| |
Collapse
|
10
|
Bisht D, Sajjanar BK, Saxena S, Kakodia B, Dighe V, Thakuria D, Kharayat NS, Chanu KV, Kumar S. Identification and characterization of phage display-selected peptides having affinity to Peste des petits ruminants virus. J Immunol Methods 2023; 515:113455. [PMID: 36893896 DOI: 10.1016/j.jim.2023.113455] [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: 10/13/2022] [Revised: 01/23/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
Phage display is a well-established technique used for selecting novel ligands having affinity to a plethora of targets including proteins, viruses, whole bacterial and mammalian cells as well as lipid targets. In the present study, phage display technology was used to identify peptides having affinity to PPRV. The binding capacity of these peptides was characterized through various formats of ELISA using phage clones, linear and multiple antigenic peptides. The whole PPRV was used as an immobilized target in a surface biopanning process using a 12-mer phage display random peptide library. After five rounds of biopanning, forty colonies were picked and amplified followed by DNA isolation and amplification for sequencing. Sequencing suggested 12 different clones expressing different peptide sequence Phage-ELISA was performed using all 12 phage clones. Results indicated that four phage clones i.e., P4, P8, P9 and P12 had a specific binding activity to PPR virus. Linear peptides displayed by all 12 clones were synthesized using solid phase peptide synthesis and subjected to virus capture ELISA. No significant binding of the linear peptides with PPRV was evident which may be due to loss of conformation of linear peptide after coating. When the four selected phage clones displayed peptide sequences were synthesized in Multiple antigenic peptide (MAP) format and used in virus capture ELISA, the results indicated significant binding of PPRV to the MAPs. It may be due to increased avidity and/or better projection of binding residues in 4-armed MAPs as compared to linear peptides. MAP-peptides were also conjugated on gold nanoparticles (AuNPs). Visual colour change from wine red to purple was observed on addition of PPRV in MAP-conjugated AuNPs solution. This colour change may be attributable to the networking of PPRV with MAP -conjugated AuNPs resulting in aggregation of AuNPs. All these results supported the hypothesis that the phage display selected peptides were capable of binding to the PPRV. The potential of these peptides to develop novel diagnostic or therapeutic agents remains to be investigated.
Collapse
Affiliation(s)
- Deepika Bisht
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; Division of Virology, ICAR-Indian Veterinary Research Institute, Mukteswar, Nainital, Uttarakhand 263138, India.
| | - B K Sajjanar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India.
| | - Shikha Saxena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India.
| | - Bhuvna Kakodia
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Vikas Dighe
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India.
| | - Dimpal Thakuria
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; ICAR-Directorate of Coldwater Fisheries Research, Bhimtal, Nainital, Uttarakhand 263136, India.
| | - Nitish S Kharayat
- Temperate Animal Husbandry Division, ICAR-Indian Veterinary Research Institute, Mukteswar Campus, Nainital, Uttarakhand 263138, India.
| | | | - Satish Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India.
| |
Collapse
|
11
|
Peste Des Petits Ruminants Virus Nucleocapsid Protein Interacts with Protein Kinase R-Activating Protein and Induces Stress Granules To Promote Viral Replication. J Virol 2023; 97:e0171222. [PMID: 36651745 PMCID: PMC9972914 DOI: 10.1128/jvi.01712-22] [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: 01/19/2023] Open
Abstract
The pathogenic mechanisms of peste des petits ruminants virus (PPRV) infection remain poorly understood, leaving peste des petits ruminants (PPR) control and eradication especially difficult. Here, we determined that PPRV nucleocapsid (N) protein triggers formation of stress granules (SGs) to benefit viral replication. A mass spectrometry-based profiling of the interactome of PPRV N protein revealed that PPRV N protein interacted with protein kinase R (PKR)-activating protein (PACT), and this interaction was confirmed in the context of PPRV infection. PACT was essential for PPRV replication. Besides, the ectopic expression of N activated the PKR/eIF2α (α subunit of eukaryotic initiation factor 2) pathway through induction of PKR phosphorylation, but it did not induce PKR phosphorylation in PACT-deficient (PACT-/-) cells. PPRV N interacted with PACT, impairing the interaction between PACT and a PKR inhibitor, transactivation response RNA-binding protein (TRBP), which subsequently enhanced the interaction between PACT and PKR and thus promoted the activation of PKR and eIF2α phosphorylation, resulting in formation of stress granules (SGs). Consistently, PPRV infection induced SG formation through activation of the PKR/eIF2α pathway, and knockdown of N impaired PPRV-induced SG formation. PPRV-induced SG formation significantly decreased in PACT-/- cells as well. The role of SG formation in PPRV replication was subsequently investigated, which showed that SG formation plays a positive role in PPRV replication. By using an RNA fluorescence in situ hybridization assay, we found that PPRV-induced SGs hid cellular mRNA rather than viral mRNA. Altogether, our data provide the first evidence that PPRV N protein plays a role in modulating the PKR/eIF2α/SG axis and promotes virus replication through targeting PACT. IMPORTANCE Stress granule (SG) formation is a conserved cellular strategy to reduce stress-related damage regulating cell survival. A mass spectrometry-based profiling of the interactome of PPRV N protein revealed that PPRV N interacted with PACT to regulate the assembly of SGs. N protein inhibited the interaction between PACT and a PKR inhibitor, TRBP, through binding to the M1 domain of PACT, which enhanced the interaction between PACT and PKR and thus promoted PKR activation and subsequent eIF2α phosphorylation as well as SG formation. The regulatory function of N protein was strikingly abrogated in PACT-/- cells. SGs induced by PPRV infection through the PKR/eIF2α pathway are PACT dependent. The loss-of-function assay indicated that PPRV-induced SGs were critical for PPRV replication. We concluded that the PPRV N protein manipulates the host PKR/eIF2α/SG axis to favor virus replication.
Collapse
|
12
|
Zhao Z, Huang C, Zhu X, Qi Z, Cao Y, Li P, Bao H, Sun P, Bai X, Fu Y, Li K, Zhang J, Ma X, Wang J, Yuan H, Li D, Liu Z, Zhang Q, Lu Z. Creation of poxvirus expressing foot-and-mouth and peste des petits ruminant disease virus proteins. Appl Microbiol Biotechnol 2023; 107:639-650. [PMID: 36586016 DOI: 10.1007/s00253-022-12351-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Foot-and-mouth disease (FMD) and Peste des petits ruminant disease (PPR) are acute and severe infectious diseases of sheep and are listed as animal diseases for compulsory immunization. However, there is no dual vaccine to prevent these two diseases. The Modified Vaccinia virus Ankara strain (MVA) has been widely used in the construction of recombinant live vector vaccine because of its large capacity of foreign gene, wide host range, high safety, and immunogenicity. In this study, MVA-GFP recombinant virus skeleton was used to construct dual live vector vaccines against FMD and PPR. METHODS The recombinant plasmid pUC57-FMDV P1-2A3CPPRV FH was synthesized and transfected into MVA-GFP infected CEF cells for homologous recombination. RESULTS The results showed that a recombinant virus without fluorescent labeling was obtained after multiple rounds of plaque screening. The recombinant virus successfully expressed the target proteins, and the empty capsid of FMDV could be observed by transmission electron microscope (TME), and the expression levels of foreign proteins (VP1 and VP3) detected by ELISA were like those detected in FMDV-infected cells. This study laid the foundation for the successful construction of a live vector vaccine against FMD and PPR. KEY POINTS • A recombinant MVA expressing FMDVP12A3C and PRRV HF proteins • Both the FMDV and PRRV proteins inserted into the virus were expressed • The proteins expressed by the recombinant poxvirus were assembled into VLPs.
Collapse
Affiliation(s)
- Zhixun Zhao
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Caiyun Huang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Xueliang Zhu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Zheng Qi
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Yimei Cao
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Pinghua Li
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Huifang Bao
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Pu Sun
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Xingwen Bai
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Yuanfang Fu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Kun Li
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Xueqing Ma
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Jian Wang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Hong Yuan
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Dong Li
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| | - Zaixin Liu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China.
| | - Qiang Zhang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China.
| | - Zengjun Lu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
13
|
Ishag HZA, Terab AMA, Eltahir YM, El Tigani-Asil ETA, Khalil NAH, Gasim EFM, Yuosf MF, Al Yammahi SMS, Al Mansoori AMA, Al Muhairi SSM, Al Hammadi ZMAH, Shah AAM, Alherbawi MMAN, Al Nuaimat MMH, Bensalah OK, Khalafalla AI. A Clinical, Pathological, Epidemiological and Molecular Investigation of Recent Outbreaks of Peste des Petits Ruminants Virus in Domestic and Wild Small Ruminants in the Abu Dhabi Emirate, United Arab Emirates. Vet Sci 2023; 10:56. [PMID: 36669056 PMCID: PMC9862675 DOI: 10.3390/vetsci10010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
(1) Background: Peste des petits ruminants (PPR) is a highly contagious animal disease affecting small ruminants, leading to significant economic losses. There has been little published data on PPR virus (PPRV) infection in the United Arab Emirates (UAE); (2) Methods: four outbreaks reported in goats and Dama gazelle in 2021 were investigated using pathological and molecular testing; (3) Results: The infected animals showed symptoms of dyspnea, oculo-nasal secretions, cough, and diarrhea. Necropsy findings were almost similar in all examined animals and compliant to the classical forms of the disease. Phylogenetic analysis based on N gene and F gene partial sequences revealed a circulation of PPRV Asian lineage IV in the UAE, and these sequences clustered close to the sequences of PPRV from United Arab Emirates, Pakistan, Tajikistan and Iran; (4) Conclusions: PPRV Asian lineage IV is currently circulating in the UAE. To the best of our knowledge, this is a first study describing PPRV in domestic small ruminant in the UAE.
Collapse
Affiliation(s)
- Hassan Zackaria Ali Ishag
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Abdelnasir Mohammed Adam Terab
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Yassir Mohammed Eltahir
- Animal Health Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - El Tigani Ahmed El Tigani-Asil
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Nasereldien Altaib Hussein Khalil
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Esamt Faisal Malik Gasim
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Mohd Farouk Yuosf
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Saeed Mohamed Saeed Al Yammahi
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Asma Mohammed Amer Al Mansoori
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | | | - Zulaikha Mohamed Abdel Hameed Al Hammadi
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Asma Abdi Mohamed Shah
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | | | | | - Oum Keltoum Bensalah
- Animal Health Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| | - Abdelmalik Ibrahim Khalafalla
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi P.O. Box 52150, United Arab Emirates
| |
Collapse
|
14
|
Aziz MH, Shabbir MZ, Ali MM, Asif Z, Ijaz MU. Immunoinformatics Approach for Epitope Mapping of Immunogenic Regions (N, F and H Gene) of Small Ruminant Morbillivirus and Its Comparative Analysis with Standard Vaccinal Strains for Effective Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10122179. [PMID: 36560589 PMCID: PMC9785197 DOI: 10.3390/vaccines10122179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Outbreaks of small ruminant morbillivirus (SRMV) are regularly occurring in Pakistan despite vaccine availability. This study was designed to identify substitutions within the immunogenic structural and functional regions of the nucleocapsid, fusion, and hemagglutinin genes of SRMV and their comparison with vaccinal strains of Nigerian and Indian origin. METHODS Swabs and tissue samples were collected from diseased animals. RT-PCR was used to characterize selected genes encoded by viral RNA. The study's N, F, and H protein sequences and vaccinal strains were analyzed for B and T cell epitope prediction using ABCpred, Bipred, and IEDB, respectively. RESULTS Significant substitutions were found on the C terminus of the nucleocapsid, within the fusion motif region of the fusion gene and in the immunoreactive region of the hemagglutinin gene. CONCLUSION Our results emphasize the need for the development of effective vaccines that match the existing variants of SRMV strains circulating in Pakistan.
Collapse
Affiliation(s)
- Muhammad Hasaan Aziz
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore 54600, Pakistan
| | - Muhammad Zubair Shabbir
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore 54600, Pakistan
- Correspondence: (M.Z.S.); (M.M.A.)
| | - Muhammad Muddassir Ali
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore 54600, Pakistan
- Correspondence: (M.Z.S.); (M.M.A.)
| | - Zian Asif
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore 54600, Pakistan
| | - Muhammad Usman Ijaz
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore 54600, Pakistan
| |
Collapse
|
15
|
Free ISG15 Inhibits the Replication of Peste des Petits Ruminants Virus by Breaking the Interaction of Nucleoprotein and Phosphoprotein. Microbiol Spectr 2022; 10:e0103122. [PMID: 36036587 PMCID: PMC9603952 DOI: 10.1128/spectrum.01031-22] [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] [Indexed: 12/30/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) causes a highly contagious disease in small ruminants and severe economic losses in developing countries. PPRV infection can stimulate high levels of interferon (IFN) and many IFN-stimulated genes (ISGs), such as ISG15, which may play a key role in the process of viral infection. However, the role of ISG15 in PPRV infection and replication has not yet been reported. In this study, we found ISG15 expression to be significantly upregulated after PPRV infection of caprine endometrial epithelial cells (EECs), and ISG15 inhibits the proliferation of PPRV. Further analysis showed that free ISG15 could inhibit PPRV proliferation. Moreover, ISG15 does not affect the binding, entry, and transcription but does suppress the replication of PPRV. A detailed analysis revealed that ISG15 interacts and colocalizes with both viral N and P proteins and that its interactive regions are all located in the N-terminal domain. Further studies showed that ISG15 can competitively interact with N and P proteins and significantly interfere with their binding. Finally, through the construction of the C-terminal mutants of ISG15 with different lengths, it was found that amino acids (aa) 77 to 101 play a key role in inhibiting the binding of N and P proteins and that interaction with the P protein disappears after the deletion of 77 to 101 aa. The present study revealed a novel mechanism of ISG15 in disrupting the activity of the N0-P complex to inhibit viral replication. IMPORTANCE PPRV, a widespread and fatal disease of small ruminants, is one of the most devastating animal diseases in Africa, the Middle East, and Asia, causing severe economic losses. IFNs play an important role as a component of natural immunity against pathogens, yet the role of ISG15, an IFN-stimulated gene, in protecting against PPRV infection is currently unknown. We demonstrated, for the first time, that free ISG15 inhibits PPRV proliferation by disrupting the activity of the N0-P complex, a finding that has not been reported in other viruses. Our results provide important insights that can further understand the pathogenesis and innate immune mechanisms of PPRV.
Collapse
|
16
|
Expression and Evaluation of a Novel PPRV Nanoparticle Antigen Based on Ferritin Self-Assembling Technology. Pharmaceutics 2022; 14:pharmaceutics14091902. [PMID: 36145650 PMCID: PMC9500948 DOI: 10.3390/pharmaceutics14091902] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Peste des Petits Ruminants (PPR) is a highly pathogenic disease that is classified as a World Organization for Animal Health (OIE)-listed disease. PPRV mainly infects small ruminants such as goats and sheep. In view of the global and high pathogenicity of PPRV, in this study, we proposed a novel nanoparticle vaccine strategy based on ferritin (Fe) self-assembly technology. Using Helicobacter pylori (H. pylori) ferritin as an antigen delivery vector, a PPRV hemagglutinin (H) protein was fused with ferritin and then expressed and purified in both Escherichia coli (E. coli) and silkworm baculovirus expression systems. Subsequently, the nanoparticle antigens’ expression level, immunogenicity and protective immune response were evaluated. Our results showed that the PPRV hemagglutinin–ferritin (H-Fe) protein was self-assembled in silkworms, while it was difficult to observe the correctly folded nanoparticle in E. coli. Meanwhile, the expression level of the H-Fe protein was higher than that of the H protein alone. Furthermore, the immunogenicity and protective immune response of H-Fe nanoparticle antigens expressed by silkworms were improved compared with the H antigen alone. Particularly, the protective immune response of H-Fe antigens expressed in E. coli did not change, as opposed to the H antigen, which was probably due to the incomplete nanoparticle structure in E. coli. This study indicated that the use of ferritin nanoparticles as antigen delivery carriers could increase the expression of antigen proteins and improve the immunogenicity and immune effect of antigens.
Collapse
|
17
|
Wen B, Yang L, Guo J, Chang W, Wei S, Yu S, Qi X, Xue Q, Wang J. Peste des petits ruminants virus induces ERS-mediated autophagy to promote virus replication. Vet Microbiol 2022; 270:109451. [PMID: 35594636 DOI: 10.1016/j.vetmic.2022.109451] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/25/2022]
Abstract
Peste des petits ruminants virus (PPRV) has long been a significant threat to small ruminant productivity worldwide. Virus infection-induced endoplasmic reticulum (ER) stress (ERS) and the subsequently activated unfolded protein response (UPR) play significant roles in viral replication and pathogenesis. However, the relationship between ERS and PPRV infection is unknown. In this study, we demonstrated that ERS was induced during PPRV infection in caprine endometrial epithelial cells (EECs). Importantly, we demonstrated that the induction of autophagy by PPRV was mediated by ERS. Furthermore, we found that the PERK/eIF2α pathway but not the ATF6 or IRE1 pathway was activated and that the activated PERK/eIF2α pathway participated in regulating ERS-mediated autophagy. Moreover, virus replication was required for PPRV infection-induced ERS-mediated autophagy and PERK pathway activation. Additionally, we revealed that either the viral nucleocapsid (N) or nonstructural protein C was sufficient to elicit ERS and activate the PERK/eIF2α pathway, which further increased autophagy. Taken together, these results suggest that PPRV N and C protein-induced autophagy enhances viral replication through the induction of ERS and that the PERK pathway may be involved in the activation of ERS-mediated autophagy during PPRV infection.
Collapse
Affiliation(s)
- Bo Wen
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Lulu Yang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Jiaona Guo
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Wenchi Chang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Shaopeng Wei
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Shengmeng Yu
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China.
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing 100000, China.
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
18
|
Peste Des Petits Ruminants Virus N Protein Is a Critical Proinflammation Factor That Promotes MyD88 and NLRP3 Complex Assembly. J Virol 2022; 96:e0030922. [PMID: 35502911 DOI: 10.1128/jvi.00309-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inflammatory responses play a central role in host defense against invading pathogens. Peste des petits ruminants virus (PPRV) causes highly contagious acute or subacute disease of small ruminants. However, the precise mechanism by which PPRV regulates inflammatory responses remains unknown. Here, we revealed a novel mechanism by which PPRV induces inflammation. Our study showed that PPRV induced the secretion of interleukin 1β (IL-1β) by activating the NF-κB signaling pathway and the NLRP3 inflammasome. Moreover, PPRV replication and protein synthesis were essential for NLRP3 inflammasome activation. Importantly, PPRV N protein promoted NF-κB signaling pathway and NLRP3 inflammasome via direct binding of MyD88 and NLPR3, respectively, and induced caspase-1 cleavage and IL-1β maturation. Biochemically, N protein interacted with MyD88 to potentiate the assembly of MyD88 complex and interacted with NLPR3 to facilitate NLRP3 inflammasome complex assembly by forming an N-NLRP3-ASC ring-like structure, leading to IL-1β secretion. These findings demonstrate a new function of PPRV N protein as an important proinflammation factor and identify a novel underlying mechanism modulating inflammasome assembly and function induced by PPRV. IMPORTANCE An important part of the innate immune response is the activation of NF-κB signaling pathway and NLPR3 inflammasome, which is induced upon exposure to pathogens. Peste des petits ruminants virus (PPRV) is a highly contagious virus causing fever, stomatitis, and pneumoenteritis in goats by inducing many proinflammatory cytokines. Although the NF-κB signaling pathway and NLRP3 inflammasome play an important role in regulating host immunity and viral infection, the precise mechanism by which PPRV regulates inflammatory responses remains unknown. This study demonstrates that PPRV induces inflammatory responses. Mechanistically, PPRV N protein facilitates the MyD88 complex assembly by directly binding to MyD88 and promotes the NLRP3 inflammasome complex assembly by directly binding to NLRP3 to form ring-like structures of N-NLRP3-ASC. These findings provide insights into the prevention and treatment of PPRV infection.
Collapse
|
19
|
Siering O, Cattaneo R, Pfaller CK. C Proteins: Controllers of Orderly Paramyxovirus Replication and of the Innate Immune Response. Viruses 2022; 14:v14010137. [PMID: 35062341 PMCID: PMC8778822 DOI: 10.3390/v14010137] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 01/07/2023] Open
Abstract
Particles of many paramyxoviruses include small amounts of proteins with a molecular weight of about 20 kDa. These proteins, termed “C”, are basic, have low amino acid homology and some secondary structure conservation. C proteins are encoded in alternative reading frames of the phosphoprotein gene. Some viruses express nested sets of C proteins that exert their functions in different locations: In the nucleus, they interfere with cellular transcription factors that elicit innate immune responses; in the cytoplasm, they associate with viral ribonucleocapsids and control polymerase processivity and orderly replication, thereby minimizing the activation of innate immunity. In addition, certain C proteins can directly bind to, and interfere with the function of, several cytoplasmic proteins required for interferon induction, interferon signaling and inflammation. Some C proteins are also required for efficient virus particle assembly and budding. C-deficient viruses can be grown in certain transformed cell lines but are not pathogenic in natural hosts. C proteins affect the same host functions as other phosphoprotein gene-encoded proteins named V but use different strategies for this purpose. Multiple independent systems to counteract host defenses may ensure efficient immune evasion and facilitate virus adaptation to new hosts and tissue environments.
Collapse
Affiliation(s)
- Oliver Siering
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, 63225 Langen, Germany;
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55906, USA
- Correspondence: (R.C.); (C.K.P.)
| | - Christian K. Pfaller
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, 63225 Langen, Germany;
- Correspondence: (R.C.); (C.K.P.)
| |
Collapse
|
20
|
Nucleotide amplification and sequencing of the GC-rich region between matrix and fusion protein genes of peste des petits ruminants virus. J Virol Methods 2021; 300:114390. [PMID: 34848280 DOI: 10.1016/j.jviromet.2021.114390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/08/2021] [Accepted: 11/26/2021] [Indexed: 01/05/2023]
Abstract
Peste des petits ruminants virus (PPRV) causes a highly devastating disease of sheep and goats, that threatens the conservation of small wild ruminants. The development of PPRV vaccines, diagnostics and therapeutics, greatly depends on in-depth genomic data. Yet, high guanine-cytosine (GC) content between matrix (M) and fusion (F) genes of PPRV poses difficulty for both primer design and nucleotide amplification. In turn, this has led into absence or low nucleotide sequence coverage in this region. This poses a risk of missing important part of the genome that could help to infer viral evolution. Here, an overlapping long-read primer-based amplification strategy was developed to amplify the GC-rich fragments between M-F gene junction using nexus gradient polymerase chain reaction (PCR). The resulting amplicons were sequenced by dideoxynucleotide cycle sequencing and compared with other PPRV nucleotide sequences available at GenBank. Our findings indicate clear PCR amplification products with expected size of the GC-rich fragments on agarose gel electrophoresis. The sequencing results of these fragments indicate 99.5 % nucleotide identity with PPRV strain KY628761. An extremely difficult PCR target of 67.4 % GC contents was successfully amplified and sequenced using this long-read primer approach. The long-read primer set may be used in tiling multiplex PCR for complete genome sequencing of PPRV.
Collapse
|
21
|
Nooruzzaman M, Akter MN, Begum JA, Begum S, Parvin R, Giasuddin M, Islam MR, Lamien CE, Cattoli G, Dundon WG, Chowdhury EH. Molecular insights into peste des petits ruminants virus identified in Bangladesh between 2008 and 2020. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 96:105163. [PMID: 34848354 DOI: 10.1016/j.meegid.2021.105163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 06/13/2023]
Abstract
An in-depth knowledge of the molecular evolution of the peste des petits ruminants virus (PPRV) is critical for the success of the current global eradication program. For this reason, a molecular evolutionary analysis of PPRVs circulating in Bangladesh over a decade (2008-2020) was performed. The complete genome sequencing of three PPRV isolates from 2008 (BD2), 2015 (BD12) and 2017 (BD17) as well as full length nucleocapsid (N), matrix (M) and fusion (F) gene sequencing of seven more samples from 2015 to 2020 was performed. Phylogenetic analysis classified all ten PPRVs from Bangladesh as members of lineage IV and showed that they were closely related to PPRV strains detected in China and Tibet during 2007-2008, and India during 2014-2018. Time scale Bayesian Maximum Clade Credibility (MCC) phylogenetic analysis of the three complete genomes revealed a mean Time to Most Recent Common Ancestor (TMRCA) of 2000. Comparative deduced amino acid residue analysis at various functional motifs of PPRVs related to virus structure and function, virulence and host adaptation, receptor binding sites and polymerase activity revealed conserved residues among the PPRVs from Bangladesh. In total sixteen epitopes were predicted from four immunogenic proteins i.e. N, M, F and haemagglutinin (H). Interestingly, the predicted epitopes from the N and M proteins shared conserved epitopes with two vaccine strains currently being used, indicating that the strains from Bangladesh could be potentially used as alternative local vaccines.
Collapse
Affiliation(s)
- Mohammed Nooruzzaman
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Mst Nazia Akter
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Jahan Ara Begum
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Shahana Begum
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; Department of Physiology, Faculty of Veterinary, Animal & Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Rokshana Parvin
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Md Giasuddin
- Animal Health Division, Bangladesh Livestock Research Institute, Savar, Dhaka, Bangladesh
| | - Mohammad Rafiqul Islam
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Charles E Lamien
- Animal Production and Health Laboratory, Joint FAO/IAEA Division, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO/IAEA Division, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - William G Dundon
- Animal Production and Health Laboratory, Joint FAO/IAEA Division, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | | |
Collapse
|
22
|
Ongoing Assessment of the Molecular Evolution of Peste Des Petits Ruminants Virus Continues to Question Viral Origins. Viruses 2021; 13:v13112144. [PMID: 34834951 PMCID: PMC8619268 DOI: 10.3390/v13112144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Understanding the evolution of viral pathogens is critical to being able to define how viruses emerge within different landscapes. Host susceptibility, which is spread between different species and is a contributing factor to the subsequent epidemiology of a disease, is defined by virus detection and subsequent characterization. Peste des petits ruminants virus is a plague of small ruminant species that is a considerable burden to the development of sustainable agriculture across Africa and much of Asia. The virus has also had a significant impact on populations of endangered species in recent years, highlighting its significance as a pathogen of high concern across different regions of the globe. Here, we have re-evaluated the molecular evolution of this virus using novel genetic data to try and further resolve the molecular epidemiology of this disease. Viral isolates are genetically characterized into four lineages (I-IV), and the historic origin of these lineages is of considerable interest to the molecular evolution of the virus. Our re-evaluation of viral emergence using novel genome sequences has demonstrated that lineages I, II and IV likely originated in West Africa, in Senegal (I) and Nigeria (II and IV). Lineage III sequences predicted emergence in either East Africa (Ethiopia) or in the Arabian Peninsula (Oman and/or the United Arab Emirates), with a paucity of data precluding a more refined interpretation. Continual refinements of evolutionary emergence, following the generation of new data, is key to both understanding viral evolution from a historic perspective and informing on the ongoing genetic emergence of this virus.
Collapse
|
23
|
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: 1.3] [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
|
24
|
Gallo G, Conceicao C, Tsirigoti C, Willett B, Graham SC, Bailey D. Application of error-prone PCR to functionally probe the morbillivirus Haemagglutinin protein. J Gen Virol 2021; 102. [PMID: 33739251 PMCID: PMC8290269 DOI: 10.1099/jgv.0.001580] [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] [Indexed: 12/04/2022] Open
Abstract
The enveloped morbilliviruses utilise conserved proteinaceous receptors to enter host cells: SLAMF1 or Nectin-4. Receptor binding is initiated by the viral attachment protein Haemagglutinin (H), with the viral Fusion protein (F) driving membrane fusion. Crystal structures of the prototypic morbillivirus measles virus H with either SLAMF1 or Nectin-4 are available and have served as the basis for improved understanding of this interaction. However, whether these interactions remain conserved throughout the morbillivirus genus requires further characterisation. Using a random mutagenesis approach, based on error-prone PCR, we targeted the putative receptor binding site for SLAMF1 interaction on peste des petits ruminants virus (PPRV) H, identifying mutations that inhibited virus-induced cell-cell fusion. These data, combined with structural modelling of the PPRV H and ovine SLAMF1 interaction, indicate this region is functionally conserved across all morbilliviruses. Error-prone PCR provides a powerful tool for functionally characterising functional domains within viral proteins.
Collapse
Affiliation(s)
- Giulia Gallo
- The Pirbright Institute, Guildford, Surrey, GU24 0NF, UK
| | | | | | - Brian Willett
- MRC University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Stephen C Graham
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Dalan Bailey
- The Pirbright Institute, Guildford, Surrey, GU24 0NF, UK
| |
Collapse
|
25
|
Kinimi E, Muyldermans S, Vincke C, Odongo S, Kock R, Parida S, Mahapatra M, Misinzo G. Development of Nanobodies Targeting Peste des Petits Ruminants Virus: The Prospect in Disease Diagnosis and Therapy. Animals (Basel) 2021; 11:ani11082206. [PMID: 34438664 PMCID: PMC8388416 DOI: 10.3390/ani11082206] [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: 06/16/2021] [Revised: 07/10/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Peste des petits ruminants virus (PPRV) causes a highly devastating disease, peste des petits ruminants (PPR) of sheep and goats, that threatens food security, small ruminant production, and the conservation of wild small ruminants. Current efforts are directed towards the global control and eradication of PPRV, an initiative of the World Organisation for Animal Health and Food and the Agriculture Organisation of the United Nations. A plethora of diagnostic tools for PPR were primarily developed for livestock. New innovative diagnostic tools are needed to detect PPRV in atypical hosts (e.g., Camelidae, Suidae, and Bovinae), in wildlife ecosystems, and in complex field situations. Recent studies confirmed that single-domain antigen binding fragments (nanobodies) derived from heavy-chain-only camelid antibodies have proven to be a powerful tool in diagnostics and therapeutics due to their unique properties, such as small size and strong antigen-binding affinity. Therefore, the main objective of this study was to generate PPRV-reactive nanobodies in order to set a pace for the development of diagnostic and possibly therapeutic nanobodies in the future. Initially, a strategy was developed whereby an alpaca was immunized with PPRV in order to raise an affinity-matured immune response, from which an immune nanobody library was constructed. Following phage display, nine nanobodies that specifically recognise PPRV were identified on enzyme-linked immunosorbent assay. This study has generated PPRV-reactive nanobodies and have significant implications in the development of cost-effective diagnostic tools in context with the planned eradication of PPR in the world. Abstract Peste des petits ruminants virus (PPRV) causes a highly devastating disease, peste des petits ruminants (PPR) of sheep and goats, that threatens food security, small ruminant production, and the conservation of wild small ruminants in many developing countries, especially in Africa. Robust serological and molecular diagnostic tools are available to detect PPRV infection, but they were mainly developed for domestic sheep and goats. The presence of a wide host range for PPRV does present serological diagnostic challenges. New innovative diagnostic tools are needed to detect PPRV in atypical hosts (e.g., Camelidae, Suidae, and Bovinae), in wildlife ecosystems and in complex field situations. Interestingly, single-domain antigen binding fragments (nanobodies) derived from heavy-chain-only camelid antibodies have emerged as a new hope in the development of accurate, rapid, and cost-effective diagnostic tools in veterinary and biomedical fields that are suitable for low-income countries. The main objective of this study was to construct an immune nanobody library to retrieve PPRV-reactive nanobodies that enable the development of diagnostic and therapeutic nanobodies in the future. Here, a strategy was developed whereby an alpaca (Vicugna pacos) was immunized with a live attenuated vaccine strain (PPRV/N/75/1) to raise an affinity-matured immune response in the heavy-chain-only antibody classes. The nanobody gene repertoire was engineered in pMECS-GG phagemid, whereby a ccdB gene (encoding a lethal protein) was substituted by the nanobody gene. An immune nanobody library with approximately sixty-four million independent transformants was constructed, of which 100% contained an insert with the proper size of nanobody gene. Following phage display and biopanning, nine nanobodies that specifically recognise completely inactivated PPRV were identified on enzyme-linked immunosorbent assay. They showed superb potency in rapidly identifying PPRV, which is likely to open a new perspective in the diagnosis and possible treatment of PPR infection.
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 25523, Tanzania;
- Department of Veterinary Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3017, Morogoro 25523, Tanzania
- Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3019, Morogoro 25523, Tanzania
- Correspondence: (E.K.); (G.M.)
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (S.M.); (C.V.)
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (S.M.); (C.V.)
| | - Steven Odongo
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine, Animal Resources and Biosecurity (COVAB), Makerere University, Kampala 7062, Uganda;
| | - Richard Kock
- The Royal Veterinary College, University of London, Hawkshead Lane, North Mymms, 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 25523, Tanzania;
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK;
| | - Mana Mahapatra
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK;
| | - Gerald Misinzo
- SACIDS Africa Centre of Excellence for Infectious Diseases, SACIDS Foundation for One Health, Sokoine University of Agriculture, P.O. Box 3297, Morogoro 25523, Tanzania;
- Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3019, Morogoro 25523, Tanzania
- Correspondence: (E.K.); (G.M.)
| |
Collapse
|
26
|
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
|
27
|
Zinc finger antiviral protein (ZAP) inhibits small ruminant morbillivirus replication in vitro. Vet Microbiol 2021; 260:109163. [PMID: 34311269 DOI: 10.1016/j.vetmic.2021.109163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/18/2021] [Indexed: 11/23/2022]
Abstract
Small ruminant morbillivirus (SRMV) is a highly contagious and economically important viral disease of small domestic and wild ruminants. Difficulty with its stable proliferation in ovis aries-derived cells has led to a relative lag in the study of its natural immunity and pathogenesis. Here we report the antiviral properties of ZAP against SRMV, a single-stranded negative-stranded RNA virus of the genus Morbillivirus. ZAP expression was significantly induced in sheep endometrial epithelial cells following SRMV infection. ZAP inhibited SRMV replication in cells after infection, while its overexpression in Vero-SLAM cells significantly increased their resistance to SRMV replication. The ZAP protein co-localized with SRMV RNA in the cytoplasm and ZAP-responsive elements were mapped to the 5' untranslated region of SRMV nucleocapsid, phosphoprotein, matrix, and fusion. In summary, ZAP confers resistance to SRMV infection by directly targeting viral RNA and inhibiting viral replication. Our findings further extend the ranges of viral targets of ZAP and help elucidate the mechanism of SRMV replication.
Collapse
|
28
|
Balamurugan V, Varghese B, SowjanyaKumari S, Vinod Kumar K, Muthuchelvan D, Nagalingam M, Hemadri D, Roy P, Shome BR. Avidin-Biotin recombinant nucleoprotein competitive ELISA for the detection of peste des petits ruminants virus antibodies in sheep and goats. J Virol Methods 2021; 295:114213. [PMID: 34119607 DOI: 10.1016/j.jviromet.2021.114213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/08/2021] [Indexed: 01/21/2023]
Abstract
The present study describes the development of a truncated recombinant peste des petits ruminants virus (PPRV) nucleoprotein (rPPRV-NPN) and its polyclonal antibodies-based immuno-diagnostic assay, Avidin-Biotin (AB) recombinant nucleoprotein competitive ELISA (ABrC-ELISA) for the detection of PPRV antibodies in the sheep and goats. The PPRV N-terminal immunogenic region (1-266 aa) of nucleoprotein (NPN) coding sequence was amplified and cloned into the pETite vector. The rPPRV-NPN with a molecular weight of ∼ 30 kDa was expressed in E. coli, purified, and characterized by SDS-PAGE and immunoblot using standard PPRV specific sera. The Ni-NTA affinity-purified rPPRV-NPN as coating antigen and its hyperimmune serum as competitive antibodies raised in guinea pigs were evaluated as diagnostic reagents in ABrC-ELISA using the known standard panel of sera. The threshold (cut-off) Percentage Inhibition (PI) value was determined as 45 (mean ± 3 SD) based on the reactivity of the known sheep and goats sera to PPRV antibodies [negative (n = 140) and positive (n = 98)] and the assay had a sensitivity of 97 % (95 % Confidence Interval (CI): 91.3-99.4 %) and specificity of 100 % (95 % CI: 97.4-100 %) with an excellent Area under curve (AUC) of 0.997 (95 % CI: 0.99-1.0). On evaluation of diagnostic performance of the assay using the sheep and goats sera (n = 391) from vaccinated, infected, and non-vaccinated animals, the ABrC-ELISA showed the relative diagnostic sensitivity of 95.88 % (95 % CI: 92.56-98.01 %) & 98.77 % (95 % CI: 96.43-99.74 %) and diagnostic specificity of 97.97 % (95 % CI: 94.19-99.58 %) & 90.54 % (95 % CI: 84.64-94.73 %) against indigenous PPR competitive ELISA kit & IDvet Screen® PPR Competition kit, respectively. The study showed that ABrC-ELISA is rapid, sensitive, and specific and can be a better alternative assay for the detection of the PPRV antibodies in the sera of small ruminants for serosurveillance / seromonitoring of PPR not only at the eradication and post-eradication phases in the disease-controlled endemic countries but also in the PPR non-endemic countries.
Collapse
Affiliation(s)
- V Balamurugan
- Indian Council of Agricultural Research -National Institute of Veterinary Epidemiology and Disease Informatics (ICAR-NIVEDI), Yelahanka, Bengaluru, 560 064, Karnataka, India.
| | - Bibitha Varghese
- Indian Council of Agricultural Research -National Institute of Veterinary Epidemiology and Disease Informatics (ICAR-NIVEDI), Yelahanka, Bengaluru, 560 064, Karnataka, India
| | - S SowjanyaKumari
- Indian Council of Agricultural Research -National Institute of Veterinary Epidemiology and Disease Informatics (ICAR-NIVEDI), Yelahanka, Bengaluru, 560 064, Karnataka, India
| | - K Vinod Kumar
- Indian Council of Agricultural Research -National Institute of Veterinary Epidemiology and Disease Informatics (ICAR-NIVEDI), Yelahanka, Bengaluru, 560 064, Karnataka, India
| | - D Muthuchelvan
- Division of Virology, ICAR-Indian Veterinary Research Institute, Campus Mukteswar-263 138, Nainital, Uttarakhand, India
| | - M Nagalingam
- Indian Council of Agricultural Research -National Institute of Veterinary Epidemiology and Disease Informatics (ICAR-NIVEDI), Yelahanka, Bengaluru, 560 064, Karnataka, India
| | - D Hemadri
- Indian Council of Agricultural Research -National Institute of Veterinary Epidemiology and Disease Informatics (ICAR-NIVEDI), Yelahanka, Bengaluru, 560 064, Karnataka, India
| | - Parimal Roy
- Centre for Animal Health Studies, TANUVAS, Madhavaram Milk Colony, Chennai, 600 051, Tamil Nadu, India
| | - B R Shome
- Indian Council of Agricultural Research -National Institute of Veterinary Epidemiology and Disease Informatics (ICAR-NIVEDI), Yelahanka, Bengaluru, 560 064, Karnataka, India
| |
Collapse
|
29
|
Ahmed S, Hosny WAEW, Mahmoud M, Mahmoud MAEF. Isolation and identification of peste des petits ruminants virus from goats in Egyptian governorates. Vet World 2021; 14:926-932. [PMID: 34083942 PMCID: PMC8167518 DOI: 10.14202/vetworld.2021.926-932] [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: 11/27/2020] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Aim The peste des petits ruminants (PPR) is a highly contagious disease of small ruminants which negatively affects animal production and the socioeconomic status of farmers. Peste des petits ruminants virus (PPRV) encodes eight proteins, with the viral fusion protein (F) playing a role in virus virulence and stimulating an effective protective immune response. This study aimed to isolate and complete the identification of PPRV circulating in goats in different Egyptian governorates and perform molecular characterization of the PPRV F gene. Materials and Methods Samples were collected from unvaccinated animals with clinical signs suggestive of PPR. A total of 256 sera were tested for the detection of PPRV antibodies using a competitive enzyme-linked immunosorbent assay (c-ELISA) kit, while 214 samples of blood buffy coat preparation, animal swabs (nasal, ocular, and saliva), and fecal and tissue samples were tested for the detection of the PPRV antigen using an antigen-capture ELISA kit. Molecular diagnosis, gene cloning, blast analysis, and phylogenetic analysis were performed for the molecular characterization of PPRV. Results The seroprevalence results of PPRV antibodies in the tested sera showed a total of 67.9% positive samples. The rates of PPR antigen recorded by the antigen-capture ELISA in the swabs (nasal and ocular) and tissue samples were 44.3%, 46.8%, and 43.5%, respectively, with saliva swabs having the highest rate of PPRV positivity (76.4%) and fecal samples having the lowest (33.3%). Molecular characterization of the PPRV Vero cell culture revealed that the circulating PPRV strain belongs to the IV lineage. Blast analysis of the PPRV F gene showed 96.7% identity with the PPRV strain Egypt-2014 fusion protein (F) gene, KT006589.1, differing by 43 single-nucleotide polymorphisms. Conclusion The results of this study indicate that the emerging PPRV belongs to the IV lineage among small ruminant animals. The findings also indicate the need for an innovative strategy to control and eliminate this disease based on a regularly administered and effective vaccine, a test to distinguish between infected and vaccinated animals, and the need for further study on the protein structure and PPRV F gene expression, which should help us to understand the molecular evolution of the virus and control and eliminate PPR disease.
Collapse
Affiliation(s)
- Sahar Ahmed
- Department of Cell Biology, Genetic Engineering and Biotechnology Research Dvision, National Research Centre, 12622 Dokki, Giza, Egypt
| | - Wafaa Abd El Wahab Hosny
- ELISA Unit and Virus Strains Bank, Animal Health Research Institute, Agriculture Research Centre, Dokki, Giza, Egypt
| | - Mervat Mahmoud
- ELISA Unit and Virus Strains Bank, Animal Health Research Institute, Agriculture Research Centre, Dokki, Giza, Egypt
| | - Mohammed Abd El-Fatah Mahmoud
- Department of Parasitology and Animal Diseases, Veterinary Research Division, National Research Centre. Dokki Giza, Egypt
| |
Collapse
|
30
|
Begum S, Nooruzzaman M, Islam MR, Chowdhury EH. A Sequential Study on the Pathology of Peste Des Petits Ruminants and Tissue Distribution of the Virus Following Experimental Infection of Black Bengal Goats. Front Vet Sci 2021; 8:635671. [PMID: 33681333 PMCID: PMC7933573 DOI: 10.3389/fvets.2021.635671] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
We studied the sequential pathology of peste des petits ruminants (PPR) in Black Bengal goats and analyzed virus distribution in tissues and virus shedding following experimental infection with a Bangladeshi isolate of lineage IV PPR virus (PPRV). The early clinical signs like fever, depression, and ocular and nasal discharges first appeared at 4-7 days post-infection (dpi). Three out of eight inoculated goats died at 13, 15, and 18 dpi, and the rest were killed at different time points from 5 to 18 dpi. Initially, the virus multiplied mostly in the lymphoid organs of the pharyngeal region and caused extensive lymphoid destruction and hemorrhages. This was followed by viremia, massive virus replication in the lungs, and pneumonia along with the appearance of the clinical signs. Subsequently, the virus spread to other organs causing necrotic and hemorrhagic lesions, as well as the virus localized in the upper respiratory, oral and intestinal mucosa resulting in catarrhal, erosive, and ulcerative lesions. On hematological and biochemical investigation progressive leukopenia and hypoproteinemia, a gradual increase of serum metabolites and enzymes associated with liver and kidney damage, and electrolyte imbalance were observed. Seroconversion started at 7 dpi and all the surviving animals had serum antibodies at 14 dpi. Virus shedding was observed in nasal and ocular secretions at 4 dpi and in feces and urine at 14 dpi, which gradually increased and continued till the end of the experiment (18 dpi) despite seroconversion. Therefore, the virus shedding of naturally infected seroconverted goats should be monitored for effective control strategies.
Collapse
Affiliation(s)
- Shahana Begum
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mohammed Nooruzzaman
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mohammad Rafiqul Islam
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Emdadul Haque Chowdhury
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| |
Collapse
|
31
|
Yirsaw A, Baldwin CL. Goat γδ T cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103809. [PMID: 32795585 DOI: 10.1016/j.dci.2020.103809] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/06/2020] [Accepted: 07/25/2020] [Indexed: 06/11/2023]
Abstract
Goats are important food animals and are disseminated globally because of their high adaptability to varying environmental conditions and feeding regimes that provide them with a comparative advantage. Productivity is impacted by infectious diseases; this then contributes to societal poverty, food insecurity, and international trade restrictions. Since γδ T cells have been shown to have vital roles in immune responses in other mammals we reviewed the literature regarding what is known about their functions, distribution in tissues and organs and their responses to a variety of infections in goats. It has been shown that caprine γδ T cells produce interferon-γ and IL-17, are found in a variety of lymphoid and nonlymphoid tissues and constitute a significant population of blood mononuclear cells. Their representation in tissues and their functional responses may be altered concomitant with infection. This review summarizes caprine γδ T cell responses to Brucella melitensis, Fasciola hepatica, Mycobacterium avium paratuberculosis, caprine arthritis encephalitis virus (CAEV), and Schistosoma bovis in infected or vaccinated goats. Caprine γδ T cells have also been evaluated in goats infected with M. caprae, Ehrilichia ruminantium, Haemonchus contortus and peste des petits ruminants (PPR) virus but found to have an unknown or limited response or role in either protective immunity or immunopathogenesis in those cases.
Collapse
Affiliation(s)
- Alehegne Yirsaw
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, 661 N. Pleasant St, University of Massachusetts, Amherst, MA, 01003, USA.
| | - Cynthia L Baldwin
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, 661 N. Pleasant St, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
32
|
Roy R, Mishra A, Poddar S, Nayak D, Kar P. Investigating the mechanism of recognition and structural dynamics of nucleoprotein-RNA complex from Peste des petits ruminants virus via Gaussian accelerated molecular dynamics simulations. J Biomol Struct Dyn 2020; 40:2302-2315. [DOI: 10.1080/07391102.2020.1838327] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Rajarshi Roy
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Anurag Mishra
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Sayan Poddar
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Debasis Nayak
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Parimal Kar
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| |
Collapse
|
33
|
Peste des petits ruminants in Africa: a review of currently available molecular epidemiological data, 2020. Arch Virol 2020; 165:2147-2163. [PMID: 32653984 PMCID: PMC7497342 DOI: 10.1007/s00705-020-04732-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/11/2020] [Indexed: 12/03/2022]
Abstract
Small ruminants (e.g., sheep and goats) contribute considerably to the cash income and nutrition of small farmers in most countries in Africa and Asia. Their husbandry is threatened by the highly infectious transboundary viral disease peste des petits ruminants (PPR) caused by peste-des-petits-ruminants virus (PPRV). Given its social and economic impact, PPR is presently being targeted by international organizations for global eradication by 2030. Since its first description in Côte d’Ivoire in 1942, and particularly over the last 10 years, a large amount of molecular epidemiological data on the virus have been generated in Africa. This review aims to consolidate these data in order to have a clearer picture of the current PPR situation in Africa, which will, in turn, assist authorities in global eradication attempts.
Collapse
|
34
|
Comerlato J, Albina E, Puech C, Franco AC, Minet C, Eloiflin RJ, Rodrigues V, Servan de Almeida R. Identification of a murine cell line that distinguishes virulent from attenuated isolates of the morbillivirus Peste des Petits Ruminants, a promising tool for virulence studies. Virus Res 2020; 286:198035. [PMID: 32461190 DOI: 10.1016/j.virusres.2020.198035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 11/18/2022]
Abstract
Comprehensive pathogenesis studies on Peste des Petits Ruminants virus (PPRV) have been delayed so far by the absence of a small animal model reproducing the disease or an in vitro biological system revealing virulence differences. In this study, a mouse 10T1/2 cell line has been identified as presenting different susceptibility to virulent and attenuated PPRV strains. As evidenced by immunofluorescence test and RT-PCR, both virulent and attenuated PPR viruses penetrated and initiated the replication cycle in 10T1/2 cells, independently of the presence of the SLAM goat receptor. However, only virulent strains successfully completed their replication cycle while the vaccine strains did not. Since 10T1/2 cells are interferon-producing cells, the role of the type I interferon (type I IFN) response on this differentiated replication between virulent and attenuated strains was verified by stimulation or repression. Modulation of the type I IFN response did not improve the replication of the vaccine strains, indicating that other cell factor(s) not yet established may hinder the replication of attenuated PPRV in 10T1/2. This 10T1/2 cell line can be proposed as a new in vitro tool for PPRV-host interaction and virulence studies.
Collapse
Affiliation(s)
- Juliana Comerlato
- CIRAD, UMR ASTRE, F-34398, Montpellier, France; Laboratório de Virologia, Departamento de Microbiologia, Imunologia e Parasitologia, ICBS UFRGS. Rua Sarmento Leite, 500, Porto Alegre. CEP 90050-170, RS, Brazil
| | - Emmanuel Albina
- CIRAD, UMR ASTRE, F-97170, Petit-Bourg, Guadeloupe, France; ASTRE, CIRAD, INRA, Univ Montpellier, Montpellier, France
| | - Carinne Puech
- INRA, UMR ASTRE, F-34398 Montpellier, France; ASTRE, CIRAD, INRA, Univ Montpellier, Montpellier, France
| | - Ana C Franco
- Laboratório de Virologia, Departamento de Microbiologia, Imunologia e Parasitologia, ICBS UFRGS. Rua Sarmento Leite, 500, Porto Alegre. CEP 90050-170, RS, Brazil
| | - Cécile Minet
- CIRAD, UMR ASTRE, F-34398, Montpellier, France; INTERTRYP, Univ Montpellier, CIRAD, IRD, Montpellier, France
| | | | - Valérie Rodrigues
- CIRAD, UMR ASTRE, F-34398, Montpellier, France; ASTRE, CIRAD, INRA, Univ Montpellier, Montpellier, France
| | - Renata Servan de Almeida
- CIRAD, UMR ASTRE, F-34398, Montpellier, France; ASTRE, CIRAD, INRA, Univ Montpellier, Montpellier, France.
| |
Collapse
|
35
|
Hemida MG, Alghadeer HM, Alhammadi M, Ali S. Prevalence and molecular characterization of some circulating strains of the peste-des-petits-ruminants virus in Saudi Arabia between 2014-2016. PeerJ 2020; 8:e9035. [PMID: 32518716 PMCID: PMC7261128 DOI: 10.7717/peerj.9035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background The peste-des-petits-ruminants virus (PPRV) is a highly devastating virus of small ruminants in many parts of the world, including the Kingdome of Saudi Arabia. Therefore, our objectives were (1) to conduct a molecular prevalence study of PPRV in sheep and goat across the KSA and (2) to isolate and identify currently circulating PPRV lineages. Swabs and tissue specimens were collected from 97 herds suspected to be infected with PPRV across the Kingdome of Saudi Arabia (KSA). Testing for the presence of the virus was done by the real-time PCR. Confirmation of the identity of the reactions was done by the gel-based-PCR then by sequencing of the partial PPRV genome. Results Our results revealed that 24.1% of the tested specimens were PPRV-positive. Isolation of PPRV was successful from samples using the Vero cell line. Sequence analysis of some partial PPRV genes (N, F, M, L, P, and H) revealed that these strains were belonging to lineage IV of the PPRV. Conclusions This is the first study to conduct both the nationwide prevalence, isolation, and molecular characterizations of the PPRV in the KSA. Continuous surveillance and monitoring of the circulating strains of PPRV among sheep and goats will contribute substantially to the global eradication campaign of such a virus.
Collapse
Affiliation(s)
- Maged Gomaa Hemida
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Hufuf, Al-Hasa, Saudi Arabia.,Department of Virology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Kafrelsheikh, Egypt
| | - Hussain Mohammed Alghadeer
- Department of Virology, Central Veterinary Diagnostic Laboratory, Minstry of Water, Enviroment and Agriculture, Riyadh, Riyadh, Saudi Arabia
| | - Mohammed Alhammadi
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Hufuf, Al-Hasa, Saudi Arabia
| | - Sayed Ali
- Department of Family and Community Medicine, College of Medicine, King Faisal University, Al-Hufuf, Al-Hasa, Saudi Arabia
| |
Collapse
|
36
|
Murr M, Hoffmann B, Grund C, Römer-Oberdörfer A, Mettenleiter TC. A Novel Recombinant Newcastle Disease Virus Vectored DIVA Vaccine against Peste des Petits Ruminants in Goats. Vaccines (Basel) 2020; 8:vaccines8020205. [PMID: 32354145 PMCID: PMC7348985 DOI: 10.3390/vaccines8020205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Peste des petits ruminants virus (PPRV, species: small ruminant morbillivirus) is the causative agent of the eponymous notifiable disease, the peste des petits ruminants (PPR) in wild and domestic sheep and goats. Mortality rates vary between 50% and 100%, causing significant losses of estimated 1.5 to 2 billion US Dollars per year. Live-attenuated PPRV vaccine strains are used in the field for disease prevention, but the application of a more thermostable vaccine enabling differentiation between infected and vaccinated animals (DIVA) would be highly desirable to achieve the goal of global disease eradication. We generated a recombinant Newcastle disease virus (rNDV) based on the live-attenuated NDV Clone 30 that expresses the surface protein hemagglutinin (H) of PPRV strain Kurdistan/11 (rNDV_HKur). In vitro analyses confirmed transgene expression as well as virus replication in avian, caprine, and ovine cells. Two consecutive subcutaneous vaccinations of German domestic goats with rNDV_HKur prevented clinical signs and hematogenic dissemination after an intranasal challenge with virulent PPRV Kurdistan/11. Virus shedding by different routes was reduced to a similar extent as after vaccination with the live-attenuated PPRV strain Nigeria 75/1. Goats that were either not vaccinated or inoculated with parental rNDV were used as controls. In summary, we demonstrate in a proof-of-concept study that an NDV vectored vaccine can protect against PPR. Furthermore, it provides DIVA-applicability and a high thermal tolerance.
Collapse
Affiliation(s)
- Magdalena Murr
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
- Correspondence: ; Tel.: +49-38351-7-1629
| | - Bernd Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Christian Grund
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Angela Römer-Oberdörfer
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Thomas C. Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| |
Collapse
|
37
|
Zhao L, Du M, Liu X, Zhang Z, Zhang Z, Meng X, Li Y. Interaction with the Receptor SLAM and Baculovirus Surface Display of Peste des petits ruminants Virus Hemagglutinin. DNA Cell Biol 2020; 39:992-999. [PMID: 32326732 DOI: 10.1089/dna.2020.5414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute, highly infectious, and highly pathogenic disease, which mainly damages small ruminants such as goats and sheep. Hemagglutinin protein (H), the main antigenic protein of peste des petits ruminants virus (PPRV), has been a hot spot in the research of genetic engineering vaccine for PPRV. In this study, the silkworm baculovirus surface display technology is combined with the transmembrane structure of the silkworm baculovirus envelope protein GP64 and different characteristics of the promoters to display four kinds of fusion proteins, which contain Pph-H, Pph-HJ, Pie1-H, and Pie1-HJ. The fusion proteins displayed on baculovirus surface have been detected by western blotting, cell surface immunofluorescence, and immunogold electron microscopy. In addition, the dominant form of PPR H displayed on baculovirus surface has been determined which is fusion protein mediated by Pph containing the hemagglutinin protein and full-length GP64, Pph-H. Furthermore, by comparing the fluorescence intensity of binding of hemagglutinin protein and signaling lymphocyte activation molecules (SLAM) in Vero-SLAM cells by immunocytochemistry, Pph-H can be combined with the receptor protein of PPRV, SLAM. It provides technical support for displaying the different structure of hemagglutinin and exploring the key sites of hemagglutinin and SLAM binding. Meanwhile, it is important for exploring the pathogenesis and immune mechanism of PPRV.
Collapse
Affiliation(s)
- Lulu Zhao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mengtan Du
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xingjian Liu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhidong Zhang
- Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xuelian Meng
- Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yinü Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
38
|
Yan F, Li E, Li L, Schiffman Z, Huang P, Zhang S, Li G, Jin H, Wang H, Zhang X, Gao Y, Feng N, Zhao Y, Wang C, Xia X. Virus-Like Particles Derived From a Virulent Strain of Pest des Petits Ruminants Virus Elicit a More Vigorous Immune Response in Mice and Small Ruminants Than Those From a Vaccine Strain. Front Microbiol 2020; 11:609. [PMID: 32390966 PMCID: PMC7190788 DOI: 10.3389/fmicb.2020.00609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/19/2020] [Indexed: 11/13/2022] Open
Abstract
Peste des petits ruminants (PPRs) is highly contagious, acute or subacute disease of small ruminants caused by peste des petits ruminants virus (PPRV). To date, several studies have designed and evaluated PPRV-like particles (VLPs) as a vaccine candidate for the prevention and control of PPR, with the majority of these VLPs constructed using sequences derived from a PPRV vaccine strain due to its high immunogenicity. However, because of the lack of available genetic material and certain structural proteins and/or the alteration of posttranslational glycosylation modifications, the immunogenicity of VLPs derived from a vaccine strain is not always optimal. In this study, two PPRV VLP candidates, derived from either the lineage IV Tibet/30 virulent strain or the lineage II Nigeria 75/1 vaccine strain, were generated using a baculovirus system through the coexpression of the PPRV matrix (M), hemagglutinin (H), and fusion (F) proteins in the high expression level cell line High Five. These VLPs were then used to immunize mice, goats, and sheep followed by two boosts after primary immunization. Both VLPs were found to induce a potent humoral immune response as demonstrated by the high ratio of immunoglobulin G1 (IgG1) to IgG2a. In all animals, both VLPs induced high titers of virus-neutralizing antibodies (VNAs), as well as H- and F-specific antibodies, with the Tibet/30 VLPs yielding higher antibody titers by comparison to the Nigeria 75/1 VLPs. Studies in mice also demonstrated that the Tibet/30 VLPs induced a more robust interleukin 4 and interferon γ response than the Nigeria 75/1 VLPs. Goats and sheep immunized with both VLPs exhibited a robust humoral and cell-mediated immune response. Furthermore, our results demonstrated that the VLPs derived from the virulent lineage IV Tibet/30 strain were more immunogenic, inducing a more potent and robust humoral and cell-mediated immune response in vaccinated animals by comparison to the lineage II Nigeria 75/1 vaccine strain VLPs. In addition, VNA titers were significantly higher among animals vaccinated with the Tibet/30 VLPs by comparison to the Nigeria 75/1 VLPs. Taken together, these findings suggest that VLPs derived from the virulent lineage IV Tibet/30 strain are more immunogenic by comparison to those derived from the lineage II Nigeria 75/1 vaccine strain and thus represent a promising vaccine candidate for the control and eradication of PPR.
Collapse
Affiliation(s)
- Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Entao Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ling Li
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Zachary Schiffman
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,National Microbiology Laboratory, Special Pathogens Program, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Pei Huang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Shengnan Zhang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Wildlife Resources, Northeast Forestry University, Harbin, China
| | - Guohua Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Hongli Jin
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hualei Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xinghai Zhang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Chengyu Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| |
Collapse
|
39
|
Complete Genome Sequence of a Lineage IV Peste des Petits Ruminants Virus from Turkey, 2018. Microbiol Resour Announc 2020; 9:9/15/e01446-19. [PMID: 32273369 PMCID: PMC7380515 DOI: 10.1128/mra.01446-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We report the whole-genome sequence of a peste des petits ruminants virus (PPRV) from a lamb exhibiting clinical signs in Turkey in September 2018. The genome of PPRV/Turkey/Central_Anatolia/2018 shows the highest nucleotide sequence identity (97.63%) to PPRV isolated in Turkey in 2000.
Collapse
|
40
|
Influence of mutation in nucleoprotein of Peste-des-petits-ruminants virus (PPRV) isolated from 2016 Indian outbreak. Small Rumin Res 2020. [DOI: 10.1016/j.smallrumres.2020.106048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
41
|
Kinimi E, Odongo S, Muyldermans S, Kock R, Misinzo G. Paradigm shift in the diagnosis of peste des petits ruminants: scoping review. Acta Vet Scand 2020; 62:7. [PMID: 31996243 PMCID: PMC6988203 DOI: 10.1186/s13028-020-0505-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/18/2020] [Indexed: 11/10/2022] Open
Abstract
Peste des petits ruminants virus causes a highly contagious disease, which poses enormous economic losses in domestic animals and threatens the conservation of wild herbivores. Diagnosis remains a cornerstone to the Peste des petits ruminants Global Control and Eradication Strategy, an initiative of the World Organisation for Animal Health and the Food and Agriculture Organisation. The present review presents the peste des petits ruminants diagnostic landscape, including the practicality of commercially available diagnostic tools, prototype tests and opportunities for new technologies. The most common peste des petits ruminants diagnostic tools include; agar gel immunodiffusion, counter-immunoelectrophoresis, enzyme-linked immunosorbent assays, reverse transcription polymerase chain reaction either gel-based or real-time, reverse transcription loop-mediated isothermal amplification, reverse transcription recombinase polymerase amplification assays, immunochromatographic lateral flow devices, luciferase immunoprecipitation system and pseudotype-based assays. These tests vary in their technical demands, but all require a laboratory with exception of immunochromatographic lateral flow and possibly reverse transcription loop-mediated isothermal amplification and reverse transcription recombinase polymerase amplification assays. Thus, we are proposing an efficient integration of diagnostic tests for rapid and correct identification of peste des petits ruminants in endemic zones and to rapidly confirm outbreaks. Deployment of pen-side tests will improve diagnostic capacity in extremely remote settings and susceptible wildlife ecosystems, where transportation of clinical samples in the optimum cold chain is unreliable.
Collapse
Affiliation(s)
- Edson Kinimi
- SACIDS Africa Centre of Excellence for Infectious Diseases of Humans and Animals in East and Southern Africa (SACIDS-ACE), SACIDS Foundation for One Health, Sokoine University of Agriculture, P.O. Box 3297, Morogoro, Tanzania.
| | - Steven Odongo
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine, Animal Resources and Biosecurity (COVAB), Makerere University, P.O. Box 7962, Kampala, Uganda
| | - Serge Muyldermans
- Department 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, Hatfield, Hertfordshire, AL9 7TA, UK
| | - Gerald Misinzo
- SACIDS Africa Centre of Excellence for Infectious Diseases of Humans and Animals in East and Southern Africa (SACIDS-ACE), SACIDS Foundation for One Health, Sokoine University of Agriculture, P.O. Box 3297, Morogoro, Tanzania
| |
Collapse
|
42
|
Dong D, Zhu S, Miao Q, Zhu J, Tang A, Qi R, Liu T, Yin D, Liu G. Nucleolin (NCL) inhibits the growth of peste des petits ruminants virus. J Gen Virol 2020; 101:33-43. [PMID: 31794379 PMCID: PMC7414435 DOI: 10.1099/jgv.0.001358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 01/05/2023] Open
Abstract
Peste des petits ruminants (PPR) is a highly contagious disease of small ruminants that is caused by peste des petits ruminants virus (PPRV). To date, the molecular mechanism of PPRV infection is still unclear. It is well known that host proteins might be involved in the pathogenesis process for many viruses. In this study, we first proved that nucleolin (NCL), a highly conserved host factor, interacts with the core domain of PPRV N protein through its C terminus and co-locates with the N protein in the nucleus of cells. To investigate the role of NCL in PPRV infection, the expression level of NCL was inhibited with small interfering RNAs of NCL, and the results showed that PPRV growth was improved. However, the proliferation of PPRV was inhibited when the expression level of NCL was improved. Further analysis indicated that the inhibitory effect of NCL on the PPRV was caused by stimulating the interferon (IFN) pathways in host cells. In summary, our results will help us to understand the mechanism of PPRV infection.
Collapse
Affiliation(s)
- Dandan Dong
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Shiqiang Zhu
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Qiuhong Miao
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Jie Zhu
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Aoxing Tang
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Ruibin Qi
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Teng Liu
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Dongdong Yin
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| | - Guangqing Liu
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, PR China
| |
Collapse
|
43
|
Li L, Wu J, Liu D, Du G, Liu Y, Shang Y, Liu X. Transcriptional Profiles of Murine Bone Marrow-Derived Dendritic Cells in Response to Peste des Petits Ruminants Virus. Vet Sci 2019; 6:vetsci6040095. [PMID: 31795377 PMCID: PMC6958494 DOI: 10.3390/vetsci6040095] [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: 11/04/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 11/16/2022] Open
Abstract
Background: Peste des petits ruminants virus (PPRV) is the causative agent of PPR, which can cause an acute, highly contagious and fatal disease of sheep and goats, resulting in significant economic losses for commercial animal husbandry due to its high mortality and morbidity. As professional antigen-presenting cells, dendritic cells (DCs) play a unique role in innate immunity. This study aimed to gain a deeper understanding of the transcriptional response of bone marrow-derived dendritic cells (BMDCs) stimulated with PPRV. Results: Transcriptional profiling was performed using RNA sequencing. Herein, we reported that compared to untreatedBMDCs, 4492 differentially expressed genes (DEGs) were identified following PPRV stimulation, out of these DEGs 2311 were upregulated and 2181 were downregulated, respectively. A total of three gene ontology (GO) term clusters of biological process, cell component and molecular function were significantly enriched in 963 GO terms in the PPRV-stimulated BMDCs. These GO clusters were related to inflammatory response, cell division and vacuole, anchoring junction, positive regulation of cellular component and nucleoside binding. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of DEGs were enriched in a chemokine signaling pathway, protein processing in endoplasmic reticulum, cell cycle and mTOR signaling pathway. Additionally, identified DEGs of BMDCs were further validated by qRT-PCR and the results were in accordance with the change of the genes. This study suggested the effects of PPRV stimulation on the maturation and function of BMDCs. Conclusion: We found that the dramatic BMDCs transcriptome changes triggered were predominantly related to an inflammatory response and chemokine signaling pathway.
Collapse
|
44
|
Evidence of Peste des petits Ruminants' Virus in Dromedary Camels in the Kingdom of Saudi Arabia between 2014 and 2016. Vet Med Int 2019; 2019:4756404. [PMID: 32089813 PMCID: PMC7012203 DOI: 10.1155/2019/4756404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/04/2019] [Indexed: 11/18/2022] Open
Abstract
Infection with the Peste des petits ruminants virus (PPRV) is a highly devastating viral infection of small ruminants. Dromedary camels live in close proximity of small ruminants in Arabian Peninsula (AP) and many other regions in the world. Little is known about the reasons behind continuous PPRV emergence in Saudi Arabia (KSA). Our objective was to test some dromedary camel population across the kingdom for the presence of specific PPRV antibodies. Our results show detection of specific PPRV antibodies (2.92%) in sera of tested dromedary camels from the eastern and south regions of the KSA. Our results suggest the exposure of dromedary camels to PPRV infection. Thus, dromedary camels may play some important roles in the sustainability of PPRV in the small ruminants across the AP. This is the first study examined the nationwide prevalence of the PPRV in dromedary camels in the KSA.
Collapse
|
45
|
Yang Y, Qin X, Meng X, Zhu X, Zhang X, Li Y, Zhang Z. MicroRNA Expression Profile in Peripheral Blood Lymphocytes of Sheep Vaccinated with Nigeria 75/1 Peste Des Petits Ruminants Virus. Viruses 2019; 11:v11111025. [PMID: 31694166 PMCID: PMC6893480 DOI: 10.3390/v11111025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/25/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Peste des petits ruminants (PPR) is one of the highly contagious transboundary viral diseases of small ruminants. Host microRNA (miRNA) expression patterns may change in response to virus infection, and it mainly works as a post-transcriptional moderator in gene expression and affects viral pathogenesis and replication. In this study, the change of miRNA expression profile in peripheral blood lymphocyte (PBMC) from sheep inoculated with PPR vaccine virus in vivo as well as primary sheep testicular (ST) cells inoculated with PPR vaccine virus in vitro were determined via deep sequencing technology. In PBMC cells, 373 and 115 differentially expressed miRNAs (DEmiRNAs) were identified 3 days and 5 days post inoculated (dpi), respectively. While, 575 DEmiRNAs were identified when comparing miRNA profiles on 5 dpi with 3 dpi. Some of the DEmiRNAs were found to change significantly via time-course during PPR vaccine virus inoculated. Similarly, in ST cells, 136 DEmiRNAs were identified at 3 dpi in comparison with mock-inoculation. A total of 12 DEmiRNAs were validated by real-time quantitative PCR (RT-qPCR). The oar-miR-150, oar-miR-370-3p and oar-miR-411b-3p were found common differentially expressed in both PPR vaccine virus-inoculated PBMC cells and ST cells. Targets prediction and functional analysis of the DEmiRNAs uncovered mainly gathering in antigen processing and presentation pathways, protein processing in endoplasmic reticulum pathways and cell adhesion molecules pathways. Our study supplies information about the DEmiRNAs in PPR vaccine virus-inoculated PBMC cells and ST cells, and provides clues for further understanding the function of miRNAs in PPR vaccine virus replication.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanmin Li
- Correspondence: ; Tel.: +86-0931-8374622
| | | |
Collapse
|
46
|
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
|
47
|
Yan F, Banadyga L, Zhao Y, Zhao Z, Schiffman Z, Huang P, Li E, Wang C, Gao Y, Feng N, Wang T, Wang H, Xia X, Wang C, Yang S, Qiu X. Peste des Petits Ruminants Virus-Like Particles Induce a Potent Humoral and Cellular Immune Response in Goats. Viruses 2019; 11:v11100918. [PMID: 31590353 PMCID: PMC6833106 DOI: 10.3390/v11100918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/19/2022] Open
Abstract
Peste des petits ruminants is a highly contagious acute or subacute disease of small ruminants caused by the peste des petits ruminants virus (PPRV), and it is responsible for significant economic losses in animal husbandry. Vaccination represents the most effective means of controlling this disease, with virus-like particle (VLP) vaccines offering promising vaccine candidates. In this study, a PPRV VLP-based vaccine was developed using a baculovirus expression system, allowing for the simultaneous expression of the PPRV matrix (M), hemagglutinin (H), fusion (F) and nucleocapsid (N) proteins in insect cells. Immunization of mice and goats with PPRV VLPs elicited a robust neutralization response and a potent cellular immune response. Mouse studies demonstrated that VLPs induced a more robust IFN-γ response in CD4+ and CD8+ T cells than PPRV Nigeria 75/1 and recruited and/or activated more B cells and dendritic cells in inguinal lymph nodes. In addition, PPRV VLPs induced a strong Th1 class response in mice, as indicated by a high IgG2a to IgG1 ratio. Goat studies demonstrated that PPRV VLPs can induce the production of antibodies specific for F and H proteins and can also stimulate the production of virus neutralizing antibodies to the same magnitude as the PPRV Nigeria 75/1 vaccine. Higher amounts of IFN-γ in VLP-immunized animal serum suggested that VLPs also elicited a cellular immune response in goats. These results demonstrated that VLPs elicit a potent immune response against PPRV infection in small ruminants, making PPRV VLPs a potential candidate for PPRV vaccine development.
Collapse
Affiliation(s)
- Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E 3R2, Manitoba, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Logan Banadyga
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E 3R2, Manitoba, Canada.
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
| | - Ziqi Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, Jilin, China.
| | - Zachary Schiffman
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E 3R2, Manitoba, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Pei Huang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, Jilin, China.
| | - Entao Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Huanan Agricultural University, Guangzhou 510642, Guangdong, China.
| | - Cuiling Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- Xinxiang medical university, Xinxiang 453003, Henan, China.
| | - Yuwei Gao
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E 3R2, Manitoba, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
| | - Hualei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Jilin University, Changchun 130122, Jilin, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Huanan Agricultural University, Guangzhou 510642, Guangdong, China.
- College of Veterinary Medicine, Jilin University, Changchun 130122, Jilin, China.
| | - Chengyu Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, Jilin, China.
- College of Veterinary Medicine, Jilin University, Changchun 130122, Jilin, China.
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E 3R2, Manitoba, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
48
|
Ahamed S, Nazir KHMNH, Yousuf MA, Islam MM, Arafat MY, Islam MA, Mahmud MM, Islam MR. Seromonitoring of Peste des Petits Ruminants in goats and molecular characterization of PPR virus from field cases. J Adv Vet Anim Res 2019; 6:416-424. [PMID: 31583240 PMCID: PMC6760515 DOI: 10.5455/javar.2019.f362] [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: 07/15/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 11/03/2022] Open
Abstract
Objectives The study was undertaken with the objectives to perform seromonitoring of Peste des Petits Ruminants (PPR) antibodies in goats vaccinated with PPR vaccine and molecular characterization of PPR virus (PPRV) from field cases in Bangladesh. Materials and Methods Seromonitoring work was conducted in Char Kalibari, Mymensingh Sadar, Mymensingh. For this, a total of 50 goats were randomly selected and were divided into two groups; vaccinated (Group A; n = 25) and non-vaccinated (Group B; n = 25). The goats of both groups were again sub-divided into four age groups; (i) 0-6 months (n = 5), (ii) 6-12 months (n = 5), (iii) 12-24 months (n = 10), and (iv) >24 months (n = 5). Blood samples were collected on Day-0 and after 21 days of post-vaccination (DPV), and the sera were prepared. The sera were examined for the presence of antibodies against PPRV by competitive enzyme-linked immunosorbent assay. For molecular characterization, nasal swabs (n = 10) were collected from PPR infected goats in Jessore during PPR outbreak (February 2016). The causative agent, PPRV isolated from field cases were confirmed by N gene based on reverse transcription polymerase chain reaction (RT-PCR), followed by sequencing, phylogenetic analysis, and multiple sequence alignment analyses. Results In the case of seromonitoring, the results revealed that before vaccination (at Day-0), overall, 44% (n = 22/50) goats were seropositive for PPRV. In Group A, 48% (n = 12/25) goats were seropositive, but after 21 DPV, 96% (n = 24/25) goats become seropositive. On the other hand, in Group B, 40% (n = 10/25) and 16% (n = 04/25) seropositive goats found at Day-0 and after 21 DPV, respectively, indicating that the antibody titer was increasing after vaccination and decreasing in convalescent goats. Out of 10 nasal swab samples, 40% (n = 4/10) was confirmed by RT-PCR targeting nucleocapsid (N gene). Phylogenetically, our isolate (KY039156/PPRV/BDG/Jes/2016) was similar to the other strains of PPRV under lineage IV. However, there was a unique amino acid substitution, where glycine (G) was recorded in place of arginine (R). The strain is closely related with other Chinese or Indian strains. The nucleotide sequence homology by NCBI BLAST search of the isolated strain ranged from 95% to 99% with other strains circulating in Bangladesh. Conclusion The PPRV is prevailing in the Mymensingh and Jessore regions of Bangladesh. Effective control of PPR in goats may depend on vaccination with PPR vaccine. Molecular characterization of PPRV in Jessore reveals that the virus is differing from the strain prevalent in other regions of Bangladesh and the world.
Collapse
Affiliation(s)
- Shamim Ahamed
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - K H M Nazmul Hussain Nazir
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Abu Yousuf
- Animal Health Research Division, Bangladesh Livestock Research Institute (BLRI) Savar, Dhaka, Bangladesh
| | - Md Monowarul Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Yeasin Arafat
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Ariful Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Muket Mahmud
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Rafiqul Islam
- Livestock Division, Bangladesh Agricultural Research Council, Dhaka, Bangladesh
| |
Collapse
|
49
|
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: 20] [Impact Index Per Article: 3.3] [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
|
50
|
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
|