1
|
Xu J, Wang Y, Zhang Y, Wang S, Su N, Chang X, Ren W, Zou Y, Liu S, Li L, Li J, Bao J, Wang Z. Establishment of a RAA-CRISPR Cas12a based diagnostic method for peste des petits ruminants virus N gene and M gene. J Virol Methods 2024; 329:114971. [PMID: 38876255 DOI: 10.1016/j.jviromet.2024.114971] [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: 03/03/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
Peste des petis ruminants (PPR) is an acute, highly contagious fatal disease affecting both domestic and wild small ruminants, caused by Morbillivirus caprinae (also known as peste des petis ruminants virus (PPRV)). Herein, a rapid method based on recombinase aided amplification-clustered regularly interspaced short palindromic repeats-Cas12a (RAA-CRISPR Cas12a) to detect PPRV was developed. CRISPR RNAs and RAA primers for PPRV-N (nucleocapsid) and PPRV-M (matrix) fragments were designed. The reaction system was constructed following screening and optimization. Detection could be completed within in 50 minutes at 37°C. Detection of gradient dilutions of plasmids carrying of PPRV N and M gene fragments indicated a minimum limit of detection of 10 copies/μL. There were no cross-reactions with related viruses and all tested lineages of PPRV were detected successfully. The method also showed good repeatability. The detection of clinical samples (previously detected using reverse transcription polymerase chain reaction (RT-PCR)) indicated good consistency between the RAA-CRISPR Cas12a method and RT-PCR. Thus, the RAA-CRISPR Cas12a method for rapid PPRV diagnosis has strong specificity, high sensitivity, and stable repeatability. Moreover, the results can be observed visually under blue or UV light or using lateral flow strips without complex instruments.
Collapse
Affiliation(s)
- Jiao Xu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Yingli Wang
- China Animal Health and Epidemiology Center, Qingdao, China
| | | | - Shujuan Wang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Su
- Qingdao Agricultural University, Qingdao, China
| | - Xing Chang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Weijie Ren
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Yanli Zou
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Shan Liu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Lin Li
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Jinming Li
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Jingyue Bao
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Zhiliang Wang
- China Animal Health and Epidemiology Center, Qingdao, China.
| |
Collapse
|
2
|
Gaur SK, Jain J, Chaudhary Y, Kaul R. Insights into the mechanism of Morbillivirus induced immune suppression. Virology 2024; 600:110212. [PMID: 39232265 DOI: 10.1016/j.virol.2024.110212] [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: 04/02/2024] [Revised: 07/26/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Viruses enter the host cell, and various strategies are employed to evade the host immune system. These include overcoming the various components of the immune system, including modulation of the physical and chemical barriers, non-specific innate response and specific adaptive immune response. Morbilliviruses impose immune modulation by utilizing various approaches including hindering antigen presentation to T-Helper (TH) cells, hematopoiesis and suppression of effector molecule activities. These viruses can also impede the early stages of T cell activation. Despite the availability of effective vaccines, morbilliviruses are still a significant threat to mankind. After infection, they also induce a state of immune suppression in the host. The molecular mechanisms employed by morbilliviruses to induce the state of immune suppression in the infected host are still being investigated. This review is an attempt to summarize insights into some of the strategies adopted by morbilliviruses to mediate immune modulation in the host.
Collapse
Affiliation(s)
- Sharad Kumar Gaur
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Juhi Jain
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Yash Chaudhary
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
3
|
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 2024:10.1007/s11259-024-10503-z. [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] [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
|
4
|
Jimale YA, Jesse FFA, Paul BT, Chung ELT, Zakaria A, Azhar NA, Mohd Lila MA. Seroprevalence and contributing factors of transboundary animal diseases in sheep and goats: a study in Peninsular Malaysia. Trop Anim Health Prod 2024; 56:212. [PMID: 39002035 DOI: 10.1007/s11250-024-04061-4] [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: 07/17/2023] [Accepted: 06/20/2024] [Indexed: 07/15/2024]
Abstract
Diseases caused by small ruminant lentiviruses, Mycobacterium avium ssp. paratuberculosis (MAP), Schmallenberg virus, and peste des petits ruminants virus (PPR) is globally recognised as serious threats to the ruminant industry due to their potential to spread rapidly across boundaries. Despite their global distribution and negative impacts on ruminant production, there is a gap in knowledge of the current trends in their epidemiology among sheep and goat populations in Peninsular Malaysia. This study was therefore designed to fill the gap of knowledge concerning the seroprevalence and contributing factors of CAEV, paratuberculosis, SBV, and PPRV among small ruminants from selected flocks in Selangor, Negeri Sembilan, and Pahang states in Peninsular Malaysia. A cross-sectional study design was used to collect animal data and blood samples for serological assays simultaneously. The ID Screen (ID.VET, France) indirect ELISA screening tests were used to detect serum antibodies directed against CAEV/MVV (VISNAS Ver 0922), paratuberculosis (PARAS Ver 0516), SBV (SBVC Ver 1114) and PPRV (PPRC Ver 0821). There was 45.4% (95% CI = 40.74-50.74), 6.8% (95% CI = 4.66-9.69), 27.8% (95% CI = 23.35-32.77), and 2.6% (95% CI = 1.11-0.51) true seroprevalence for CAEV, paratuberculosis, SBV, and PPR, respectively. Geographical location and species were the risk factors for CAEV and paratuberculosis, while the management system and age of small ruminants were the risk factors for SBV. The present study is the first to document a large-scale seroprevalence of MAP and PPR infection among sheep and goat flocks in Peninsular Malaysia. The presence of PPRV and MAP antibodies among small ruminant flocks is signalling current or previous exposure to the pathogens or cross reactions with similar antigens. This finding further suggests the potential for future outbreaks of these devastating diseases among sheep and goats in Malaysia. The high seroprevalence of CAEV and SBV among small ruminants indicates high levels of exposure to the viruses in the environment, which is a potential threat to production.
Collapse
Affiliation(s)
- Yonis Ahmed Jimale
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Faez Firdaus Abdullah Jesse
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia.
| | - Bura Thlama Paul
- Department of Animal Science and Fisheries, Faculty of Agriculture and Forestry Science, Universiti Putra Malaysia Campus Bintulu Sarawak, Sarawak, Malaysia
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, University of Maiduguri 600230, Borno State, Nigeria
| | - Eric Lim Teik Chung
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Aida Zakaria
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Nur Amira Azhar
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Mohd Azmi Mohd Lila
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| |
Collapse
|
5
|
Zhang Q, Zheng X, Zhang F, Cui X, Yan N, Hu J, Guo Y, Wang X. Unveiling of the Co-Infection of Peste des Petits Ruminants Virus and Caprine Enterovirus in Goat Herds with Severe Diarrhea in China. Viruses 2024; 16:986. [PMID: 38932277 PMCID: PMC11209052 DOI: 10.3390/v16060986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Here, we report the discovery of two viruses associated with a disease characterized by severe diarrhea on a large-scale goat farm in Jilin province. Electron Microscopy observations revealed two kinds of virus particles with the sizes of 150-210 nm and 20-30 nm, respectively. Detection of 276 fecal specimens from the diseased herds showed the extensive infection of peste des petits ruminants virus (63.77%, 176/276) and caprine enterovirus (76.81%, 212/276), with a co-infection rate of 57.97% (160/276). These results were partially validated with RT-PCR, where all five PPRV-positive and CEV-positive specimens yielded the expected size of fragments, respectively, while no fragments were amplified from PPRV-negative and CEV-negative specimens. Moreover, corresponding PPRV and CEV fragments were amplified in PPRV and CEV double-positive specimens. Histopathological examinations revealed severe microscopic lesions such as degeneration, necrosis, and detachment of epithelial cells in the bronchioles and intestine. An immunohistochemistry assay detected PPRV antigens in bronchioles, cartilage tissue, intestine, and lymph nodes. Simultaneously, caprine enterovirus antigens were detected in lung, kidney, and intestinal tissues from the goats infected by the peste des petits ruminants virus. These results demonstrated the co-infection of peste des petits ruminants virus with caprine enterovirus in goats, revealing the tissue tropism for these two viruses, thus laying a basis for the future diagnosis, prevention, and epidemiological survey for these two virus infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xinping Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130012, China; (Q.Z.); (X.Z.); (F.Z.); (X.C.); (N.Y.); (J.H.); (Y.G.)
| |
Collapse
|
6
|
Mittal P, Khandelwal N, Chander Y, Verma A, Kumar R, Putatunda C, Barua S, Gulati BR, Kumar N. p38-MAPK is prerequisite for the synthesis of SARS-CoV-2 protein. Virusdisease 2024; 35:329-337. [PMID: 39071879 PMCID: PMC11269555 DOI: 10.1007/s13337-024-00873-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/15/2024] [Indexed: 07/30/2024] Open
Abstract
The inhibition of p38 mitogen-activated protein kinase (p38-MAPK) by small molecule chemical inhibitors was previously shown to impair severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, however, mechanisms underlying antiviral activity remains unexplored. In this study, reduced growth of SARS-CoV-2 in p38-α knockout Vero cells, together with enhanced viral yield in cells transfected with construct expressing p38α, suggested that p38-MAPK is essential for the propagation of SARS-CoV-2. The SARS-CoV-2 was also shown to induce phosphorylation (activation) of p38, at time when transcription/translational activities are considered to be at the peak levels. Further, we demonstrated that p38 supports viral RNA/protein synthesis without affecting viral attachment, entry, and budding in the target cells. In conclusion, we provide mechanistic insights on the regulation of SARS-CoV-2 replication by p38 MAPK.
Collapse
Affiliation(s)
- Priyasi Mittal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
- Om Sterling Global University (OSGU), Hisar, Haryana 125001 India
| | - Nitin Khandelwal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
| | - Yogesh Chander
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
| | - Assim Verma
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
| | - Ram Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
| | | | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
| | - Baldev Raj Gulati
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
| | - Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana 125001 India
| |
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
|
Xu JL, Chen JT, Hu B, Guo WW, Guo JJ, Xiong CR, Qin LX, Yu XN, Chen XM, Cai K, Li YR, Liu MQ, Chen LJ, Hou W. Discovery and genetic characterization of novel paramyxoviruses from small mammals in Hubei Province, Central China. Microb Genom 2024; 10:001229. [PMID: 38700925 PMCID: PMC11145887 DOI: 10.1099/mgen.0.001229] [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: 10/07/2023] [Accepted: 03/26/2024] [Indexed: 05/05/2024] Open
Abstract
Paramyxoviruses are a group of single-stranded, negative-sense RNA viruses, some of which are responsible for acute human disease, including parainfluenza virus, measles virus, Nipah virus and Hendra virus. In recent years, a large number of novel paramyxoviruses, particularly members of the genus Jeilongvirus, have been discovered in wild mammals, suggesting that the diversity of paramyxoviruses may be underestimated. Here we used hemi-nested reverse transcription PCR to obtain 190 paramyxovirus sequences from 969 small mammals in Hubei Province, Central China. These newly identified paramyxoviruses were classified into four clades: genera Jeilongvirus, Morbillivirus, Henipavirus and Narmovirus, with most of them belonging to the genus Jeilongvirus. Using Illumina sequencing and Sanger sequencing, we successfully recovered six near-full-length genomes with different genomic organizations, revealing the more complex genome content of paramyxoviruses. Co-divergence analysis of jeilongviruses and their known hosts indicates that host-switching occurred more frequently in the evolutionary histories of the genus Jeilongvirus. Together, our findings demonstrate the high prevalence of paramyxoviruses in small mammals, especially jeilongviruses, and highlight the diversity of paramyxoviruses and their genome content, as well as the evolution of jeilongviruses.
Collapse
Affiliation(s)
- Jia-le Xu
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Jin-tao Chen
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Bing Hu
- Institute of Health Inspection and Testing, Hubei Provincial Center for Disease Control & Prevention, 6 Zhuodaoquan Road, Wuhan, Hubei, 430079, PR China
| | - Wei-wei Guo
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Jing-jing Guo
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Chao-rui Xiong
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Ling-xin Qin
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Xin-nai Yu
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Xiao-min Chen
- Division of Virology, Wuhan Center for Disease Control & Prevention, 288 Machang Road, Wuhan, Hubei, 430015, PR China
| | - Kun Cai
- Institute of Health Inspection and Testing, Hubei Provincial Center for Disease Control & Prevention, 6 Zhuodaoquan Road, Wuhan, Hubei, 430079, PR China
| | - Yi-rong Li
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Man-qing Liu
- Division of Virology, Wuhan Center for Disease Control & Prevention, 288 Machang Road, Wuhan, Hubei, 430015, PR China
| | - Liang-jun Chen
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| | - Wei Hou
- State Key Laboratory of Virology/Department of Laboratory Medicine/Hubei Provincial Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences/Zhongnan Hospital, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
- School of Public Health, Wuhan University, 185 Donghu Road, Wuhan, Hubei, 430071, PR China
| |
Collapse
|
9
|
Wu J, Yang W, Li L, Wu J, He J, Ru Y, Ren J, Wang Y, Zheng H, Shang Y, Li D. Plasminogen activator urokinase interacts with the fusion protein and antagonizes the growth of Peste des petits ruminants virus. J Virol 2024; 98:e0014624. [PMID: 38440983 PMCID: PMC11019896 DOI: 10.1128/jvi.00146-24] [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: 01/21/2024] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Peste des petits ruminants is an acute and highly contagious disease caused by the Peste des petits ruminants virus (PPRV). Host proteins play a crucial role in viral replication. However, the effect of fusion (F) protein-interacting partners on PPRV infection is poorly understood. In this study, we found that the expression of goat plasminogen activator urokinase (PLAU) gradually decreased in a time- and dose-dependent manner in PPRV-infected goat alveolar macrophages (GAMs). Goat PLAU was subsequently identified using co-immunoprecipitation and confocal microscopy as an F protein binding partner. The overexpression of goat PLAU inhibited PPRV growth and replication, whereas silencing goat PLAU promoted viral growth and replication. Additionally, we confirmed that goat PLAU interacted with a virus-induced signaling adapter (VISA) to antagonize F-mediated VISA degradation, increasing the production of type I interferon. We also found that goat PLAU reduced the inhibition of PPRV replication in VISA-knockdown GAMs. Our results show that the host protein PLAU inhibits the growth and replication of PPRV by VISA-triggering RIG-I-like receptors and provides insight into the host protein that antagonizes PPRV immunosuppression.IMPORTANCEThe role of host proteins that interact with Peste des petits ruminants virus (PPRV) fusion (F) protein in PPRV replication is poorly understood. This study confirmed that goat plasminogen activator urokinase (PLAU) interacts with the PPRV F protein. We further discovered that goat PLAU inhibited PPRV replication by enhancing virus-induced signaling adapter (VISA) expression and reducing the ability of the F protein to degrade VISA. These findings offer insights into host resistance to viral invasion and suggest new strategies and directions for developing PPR vaccines.
Collapse
Affiliation(s)
- Junhuang Wu
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Wenping Yang
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Lingxia Li
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Jingyan Wu
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Jijun He
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Yi Ru
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Yong Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| |
Collapse
|
10
|
Courcelle M, Salami H, Tounkara K, Lo MM, Ba A, Diop M, Niang M, Sidibe CAK, Sery A, Dakouo M, Kaba L, Sidime Y, Keyra M, Diallo AOS, El Mamy AB, El Arbi AS, Barry Y, Isselmou E, Habiboullah H, Doumbia B, Gueya MB, Awuni J, Odoom T, Ababio PT, TawiahYingar DNY, Coste C, Guendouz S, Kwiatek O, Libeau G, Bataille A. Comparative evolutionary analyses of peste des petits ruminants virus genetic lineages. Virus Evol 2024; 10:veae012. [PMID: 38476867 PMCID: PMC10930206 DOI: 10.1093/ve/veae012] [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: 10/10/2023] [Revised: 01/16/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
Peste des petits ruminants virus (PPRV) causes a highly infectious disease affecting mainly goats and sheep in large parts of Africa, Asia, and the Middle East and has an important impact on the global economy and food security. Full genome sequencing of PPRV strains has proved to be critical to increasing our understanding of PPR epidemiology and to inform the ongoing global efforts for its eradication. However, the number of full PPRV genomes published is still limited and with a heavy bias towards recent samples and genetic Lineage IV (LIV), which is only one of the four existing PPRV lineages. Here, we generated genome sequences for twenty-five recent (2010-6) and seven historical (1972-99) PPRV samples, focusing mainly on Lineage II (LII) in West Africa. This provided the first opportunity to compare the evolutionary pressures and history between the globally dominant PPRV genetic LIV and LII, which is endemic in West Africa. Phylogenomic analysis showed that the relationship between PPRV LII strains was complex and supported the extensive transboundary circulation of the virus within West Africa. In contrast, LIV sequences were clearly separated per region, with strains from West and Central Africa branched as a sister clade to all other LIV sequences, suggesting that this lineage also has an African origin. Estimates of the time to the most recent common ancestor place the divergence of modern LII and LIV strains in the 1960s-80s, suggesting that this period was particularly important for the diversification and spread of PPRV globally. Phylogenetic relationships among historical samples from LI, LII, and LIII and with more recent samples point towards a high genetic diversity for all these lineages in Africa until the 1970s-80s and possible bottleneck events shaping PPRV's evolution during this period. Molecular evolution analyses show that strains belonging to LII and LIV have evolved under different selection pressures. Differences in codon usage and adaptative selection pressures were observed in all viral genes between the two lineages. Our results confirm that comparative genomic analyses can provide new insights into PPRV's evolutionary history and molecular epidemiology. However, PPRV genome sequencing efforts must be ramped up to increase the resolution of such studies for their use in the development of efficient PPR control and surveillance strategies.
Collapse
Affiliation(s)
- Maxime Courcelle
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
| | - Habib Salami
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
- Institut Sénégalais de Recherches Agricoles, Laboratoire National d’Elevage et de Recherches Vétérinaires (LNERV), Dakar-Hann BP 2057, Sénégal
| | - Kadidia Tounkara
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
- Laboratoire Central Vétérinaire (LCV), Bamako BP 2295, Mali
| | - Modou Moustapha Lo
- Institut Sénégalais de Recherches Agricoles, Laboratoire National d’Elevage et de Recherches Vétérinaires (LNERV), Dakar-Hann BP 2057, Sénégal
| | - Aminata Ba
- Institut Sénégalais de Recherches Agricoles, Laboratoire National d’Elevage et de Recherches Vétérinaires (LNERV), Dakar-Hann BP 2057, Sénégal
| | - Mariame Diop
- Institut Sénégalais de Recherches Agricoles, Laboratoire National d’Elevage et de Recherches Vétérinaires (LNERV), Dakar-Hann BP 2057, Sénégal
| | - Mamadou Niang
- Laboratoire Central Vétérinaire (LCV), Bamako BP 2295, Mali
| | | | - Amadou Sery
- Laboratoire Central Vétérinaire (LCV), Bamako BP 2295, Mali
| | - Marthin Dakouo
- Laboratoire Central Vétérinaire (LCV), Bamako BP 2295, Mali
| | - Lanceï Kaba
- Institut Supérieur des Sciences et de Médecine Vétérinaire, Dalaba BP 2201, Guinea
| | - Youssouf Sidime
- Institut Supérieur des Sciences et de Médecine Vétérinaire, Dalaba BP 2201, Guinea
| | - Mohamed Keyra
- Institut Supérieur des Sciences et de Médecine Vétérinaire, Dalaba BP 2201, Guinea
| | | | - Ahmed Bezeid El Mamy
- Office National de Recherches et de Développement de l’Elevage (ONARDEL), Nouakchott BP 167, Mauritania
| | - Ahmed Salem El Arbi
- Office National de Recherches et de Développement de l’Elevage (ONARDEL), Nouakchott BP 167, Mauritania
| | - Yahya Barry
- Office National de Recherches et de Développement de l’Elevage (ONARDEL), Nouakchott BP 167, Mauritania
| | - Ekaterina Isselmou
- Office National de Recherches et de Développement de l’Elevage (ONARDEL), Nouakchott BP 167, Mauritania
| | - Habiboullah Habiboullah
- Office National de Recherches et de Développement de l’Elevage (ONARDEL), Nouakchott BP 167, Mauritania
| | - Baba Doumbia
- Office National de Recherches et de Développement de l’Elevage (ONARDEL), Nouakchott BP 167, Mauritania
| | - Mohamed Baba Gueya
- Office National de Recherches et de Développement de l’Elevage (ONARDEL), Nouakchott BP 167, Mauritania
| | - Joseph Awuni
- Accra Veterinary Laboratory, Veterinary Services Directorate, Accra M161, Ghana
| | - Theophilus Odoom
- Accra Veterinary Laboratory, Veterinary Services Directorate, Accra M161, Ghana
| | | | | | - Caroline Coste
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
| | - Samia Guendouz
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
| | - Olivier Kwiatek
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
| | - Geneviève Libeau
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
| | - Arnaud Bataille
- ASTRE, University of Montpellier, CIRAD, INRAE, Montpellier F-34398, France
- CIRAD, UMR ASTRE, Montpellier F-34398, France
| |
Collapse
|
11
|
Yuan S, Liu Y, Mu Y, Kuang Y, Chen S, Zhao YT, Liu Y. Peste des petits ruminants virus infection induces endoplasmic reticulum stress and apoptosis via IRE1-XBP1 and IRE1-JNK signaling pathways. J Vet Sci 2024; 25:e21. [PMID: 38568823 PMCID: PMC10990917 DOI: 10.4142/jvs.23236] [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: 09/28/2023] [Revised: 12/31/2023] [Accepted: 01/10/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Peste des petits ruminants (PPR) is a contagious and fatal disease of sheep and goats. PPR virus (PPRV) infection induces endoplasmic reticulum (ER) stress-mediated unfolded protein response (UPR). The activation of UPR signaling pathways and their impact on apoptosis and virus replication remains controversial. OBJECTIVES To investigate the role of PPRV-induced ER stress and the IRE1-XBP1 and IRE1-JNK pathways and their impact on apoptosis and virus replication. METHODS The cell viability and virus replication were assessed by 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay, immunofluorescence assay, and Western blot. The expression of ER stress biomarker GRP78, IRE1, and its downstream molecules, PPRV-N protein, and apoptosis-related proteins was detected by Western blot and quantitative reverse transcription-polymerase chain reaction, respectively. 4-Phenylbutyric acid (4-PBA) and STF-083010 were respectively used to inhibit ER stress and IRE1 signaling pathway. RESULTS The expression of GRP78, IRE1α, p-IRE1α, XBP1s, JNK, p-JNK, caspase-3, caspase-9, Bax and PPRV-N were significantly up-regulated in PPRV-infected cells, the expression of Bcl-2 was significantly down-regulated. Due to 4-PBA treatment, the expression of GRP78, p-IRE1α, XBP1s, p-JNK, caspase-3, caspase-9, Bax, and PPRV-N were significantly down-regulated, the expression of Bcl-2 was significantly up-regulated. Moreover, in PPRV-infected cells, the expression of p-IRE1α, p-JNK, Bax, and PPRV-N was significantly decreased, and the expression of Bcl-2 was increased in the presence of STF-083010. CONCLUSIONS PPRV infection induces ER stress and IRE1 activation, resulting in apoptosis and enhancement of virus replication through IRE1-XBP1s and IRE1-JNK pathways.
Collapse
Affiliation(s)
- Shuyi Yuan
- College of Coastal Agricultural Science of Guangdong Ocean University, Zhanjiang 524088, China
| | - Yanfen Liu
- College of Coastal Agricultural Science of Guangdong Ocean University, Zhanjiang 524088, China
| | - Yun Mu
- College of Coastal Agricultural Science of Guangdong Ocean University, Zhanjiang 524088, China
| | - Yongshen Kuang
- College of Coastal Agricultural Science of Guangdong Ocean University, Zhanjiang 524088, China
| | - Shaohong Chen
- College of Food Science and Technology of Guangdong Ocean University, Zhanjiang 524088, China
| | - Yun-Tao Zhao
- College of Food Science and Technology of Guangdong Ocean University, Zhanjiang 524088, China
| | - You Liu
- College of Food Science and Technology of Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
12
|
Ba A, Diop GL, Ndiaye M, Dione M, Lo MM. First Report of the Emergence of Peste des Petits Ruminants Lineage IV Virus in Senegal. Viruses 2024; 16:305. [PMID: 38400080 PMCID: PMC10892897 DOI: 10.3390/v16020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/25/2023] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Peste des petits ruminants (PPR) is a highly contagious viral disease and one of the deadliest affecting wild goats, sheep, and small ruminants; however, goats are generally more sensitive. The causative agent is the Peste des Petits Ruminants virus (PPRV), which is a single-stranded RNA virus of negative polarity belonging to the Paramyxoviridae family. In February 2020, an active outbreak of PPR was reported in a herd of a transhumant farmer in the village of Gainth Pathé (department of Kounguel, Kaffrine region, Senegal). Of the ten swabs collected from the goats, eight returned a positive result through a quantitative real-time PCR. The sample that yielded the strongest signal from the quantitative real-time PCR was further analyzed with a conventional PCR amplification and direct amplicon sequencing. A phylogenetic analysis showed that the sequence of the PPR virus obtained belonged to lineage IV. These results confirm those found in the countries bordering Senegal and reinforce the hypothesis of the importance of animal mobility between these neighboring countries in the control of PPRV. In perspective, following the discovery of this lineage IV in Senegal, a study on its dispersion is underway throughout the national territory. The results that will emerge from this study, associated with detailed data on animal movements and epidemiological data, will provide appropriate and effective information to improve PPR surveillance and control strategies with a view to its eradication.
Collapse
Affiliation(s)
- Aminata Ba
- Institut Sénégalais de Recherches Agricoles, Laboratoire National de l’Elevage et de Recherches Vétérinaires (ISRA-LNERV), Dakar-Hann BP 2057, Senegal; (G.L.D.); (M.N.); (M.M.L.)
| | - Gaye Laye Diop
- Institut Sénégalais de Recherches Agricoles, Laboratoire National de l’Elevage et de Recherches Vétérinaires (ISRA-LNERV), Dakar-Hann BP 2057, Senegal; (G.L.D.); (M.N.); (M.M.L.)
| | - Mbengué Ndiaye
- Institut Sénégalais de Recherches Agricoles, Laboratoire National de l’Elevage et de Recherches Vétérinaires (ISRA-LNERV), Dakar-Hann BP 2057, Senegal; (G.L.D.); (M.N.); (M.M.L.)
| | - Michel Dione
- International Livestock Research Institute (ILRI), Dakar 24265, Senegal;
| | - Modou Moustapha Lo
- Institut Sénégalais de Recherches Agricoles, Laboratoire National de l’Elevage et de Recherches Vétérinaires (ISRA-LNERV), Dakar-Hann BP 2057, Senegal; (G.L.D.); (M.N.); (M.M.L.)
| |
Collapse
|
13
|
Lu G, Wang P, Miao S, Huang J, Ma W, Mi X, Xue J, Shayilan K, Yang X, Yan G. Prokaryotic expression of the V protein of the peste des petits ruminants virus and development of an indirect ELISA. Anim Biotechnol 2023; 34:5011-5015. [PMID: 37288766 DOI: 10.1080/10495398.2023.2221703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this study, we recombinantly expressed the V protein of the peste des petits ruminants virus (PPRV) and evaluated its diagnostic value for PPRV infection using an indirect ELISA (i-ELISA). The optimal concentration of the coated antigen of V protein was 15 ng/well at a serum dilution of 1:400, and the optimal positive threshold value was 0.233. A cross-reactivity assay showed that the V protein-based i-ELISA was specific to PPRV with consistent reproducibility and showed a specificity of 82.6% and a sensitivity of 100% with a virus neutralization test. Using the recombinant V protein as an antigen in ELISA is useful for seroepidemiological studies of PPRV infections.
Collapse
Affiliation(s)
- Guili Lu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, P. R. China
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Ping Wang
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Shukui Miao
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Jiong Huang
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Wenge Ma
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Xiaoyun Mi
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Jing Xue
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Kayizha Shayilan
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Xueyun Yang
- The Veterinary Research Institute, Animal Science Academy of Xinjiang (Animal Clinical Medicine Research Center, Animal Science Academy of Xinjiang), Urumqi, Xinjiang, P. R. China
| | - Genqiang Yan
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, P. R. China
| |
Collapse
|
14
|
Agrawal A, Varshney R, Gattani A, Hira Khan M, Gupta R, Solanki KS, Patel SK, Singh RP, Singh P. Development of Hemagglutinin-Neuraminidase Homologous Peptides as Novel Promising Therapeutic Agents Against Peste des Petits Ruminants Virus. Protein J 2023; 42:685-697. [PMID: 37421558 DOI: 10.1007/s10930-023-10134-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/10/2023]
Abstract
The lack of specific antiviral therapy and complications associated with the existing peste des petits ruminants (PPR) vaccines accentuates the search of novel antiviral blocking agents in order to curtail the PPR infection at initial level. The synthetic hemagglutinin-neuraminidase (HN) homologous peptides may compete with the natural HN protein of PPR virus for binding to signaling lymphocytic activation molecule (SLAM) receptor, consequently, may disrupt peste des petits ruminants virus (PPRV) at entry level. Therefore, insilico analysis, synthesis, purification and subsequent characterization of HN homologous peptides were conducted in this study. The HN homologous peptides were synthesized by means of solid phase chemistry and were purified by reversed-phase-high performance liquid chromatography. The mass as well as sequence of HN homologous peptides were assessed by mass spectroscopy while its secondary structure was elucidated by circular dichroism spectroscopy. The binding (interaction) efficacy of HN homologous peptides with PPRV antibodies was assessed via indirect enzyme linked immunosorbent assay, visual detection test (red wine to purple), bathochromic shift under UV-Vis spectrophotometry and lateral flow immunochromatographic strip test. The antiviral properties and cytotoxicity of these peptides were also assessed in B95a cell line with changes in cytopathic effect and titer of PPRV (Sungri/96). The presence of green fluorescein isothiocyanate over the B95a cell surface pointed towards the binding of HN homologous peptides with surface SLAM receptor. Moreover, the intact beta sheet configuration in water and lower cytotoxicity [cytotoxic concentration 50 (CC50) > 1000 µg/ml] of these peptides signifies its in vivo use. Among HN homologous peptides, the binding efficacy and antiviral properties of pep A was relatively high in comparison to pep B and Pep ppr peptides. The prerequisite concentration of HN homologous peptides (pep A = 12.5 µg/ml; pep B = 25 µg/ml; pep ppr = 25 µg/ml) to exemplify its antiviral effect was much lower than its CC50 level. Hence, this study signifies the therapeutic potential of synthetic HN homologous peptides.
Collapse
Affiliation(s)
- Aditya Agrawal
- Division of Animal Biochemistry, IVRI, Izatnagar, Bareilly, U.P., 243122, India.
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Science and Animal husbandry, NDVSU, Rewa, Jabalpur, 486001, India.
| | - Rajat Varshney
- Department of Veterinary Microbiology, BHU, Mirzapur, U.P., 231001, India
- Division of Bacteriology and Mycology, IVRI, Izatnagar, Bareilly, U.P., 243122, India
| | - Anil Gattani
- Division of Animal Biochemistry, IVRI, Izatnagar, Bareilly, U.P., 243122, India
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Science and Animal husbandry, NDVSU, Jabalpur, 486001, India
| | - Mahvash Hira Khan
- Division of Animal Biochemistry, IVRI, Izatnagar, Bareilly, U.P., 243122, India
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Science and Animal husbandry, NDVSU, Jabalpur, 486001, India
| | - Rohini Gupta
- Department of Veterinary Medicine, College of Veterinary Science and Animal husbandry, NDVSU, Jabalpur, 486001, India
| | - Khushal Singh Solanki
- Division of Veterinary Biotechnology, IVRI, Izatnagar, Bareilly, U.P., 243122, India
| | - Shailesh Kumar Patel
- Department of Veterinary Pathology, College of Veterinary Science and Animal husbandry, NDVSU, Rewa, Jabalpur, 486001, India
| | - R P Singh
- Division of Bacteriology and Mycology, IVRI, Izatnagar, Bareilly, U.P., 243122, India
| | - Praveen Singh
- Division of Animal Biochemistry, IVRI, Izatnagar, Bareilly, U.P., 243122, India
| |
Collapse
|
15
|
Muritala I, Bemji MN, Ozoje MO, Ajayi OL, Oluwayinka EB, Sonibare AO, James IJ, Ibeagha-Awemu EM. Comparative study of HA and HNB staining RT-LAMP assays for peste des petits ruminants virus detection in West African Dwarf goats. Trop Anim Health Prod 2023; 55:356. [PMID: 37821730 DOI: 10.1007/s11250-023-03747-5] [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: 12/05/2022] [Accepted: 09/12/2023] [Indexed: 10/13/2023]
Abstract
Peste des petits ruminants (PPR) cause severe economic losses to many countries of the world where the disease is endemic. It has been targeted for global eradication by 2030 following the successful eradication of rinderpest in 2011. The proposed eradication program would benefit from efficient and relatively reliable diagnostic tools for early PPR virus (PPRV) detection. A total of 33 eight to 12 months old West African Dwarf (WAD) goats were used. Nineteen goats infected by commingling with two PPR virus-positive animals formed the infected group (PPRV-infected goats) while 14 non-infected goats formed the control group (CTG). The suitability of hydroxyl naphthol blue (HNB) staining of reverse transcription loop-mediated isothermal amplification (RT-LAMP) and haemagglutination (HA) assays was compared for their sensitivity to detect the PPRV in PPRV-infected goats and non-infected CTG. PPR disease severity in WAD goats at different days post infection (dpi) was evaluated by clinical scoring and haemagglutination titre (HAT). HNB staining RT-LAMP reaction and HA showed sensitivities of 100% and 73.68%, respectively, for PPRV detection. Expression of PPR clinical signs began from 3 dpi, attained peak at 5 dpi, thereafter showed irregular patterns till 24 dpi. Evaluation of HAT in PPRV-infected goats at 12 dpi ranged from 2 to 64 haemagglutination units (HAU), while CTG goats had 0 HAU. In conclusion, HA could be a good tool for rapid diagnosis of PPRV in a developing country setting. However, HNB staining RT-LAMP assay demonstrated high sensitivity for accurate diagnoses of PPRV and as an important diagnostic tool when precise phenotyping is desired.
Collapse
Affiliation(s)
- Ismaila Muritala
- Department of Animal Breeding and Genetics, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Martha N Bemji
- Department of Animal Breeding and Genetics, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria.
| | - Michael O Ozoje
- Department of Animal Breeding and Genetics, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Olusola L Ajayi
- Department of Pathology, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Eniope B Oluwayinka
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Adekayode O Sonibare
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Ikechukwu J James
- Department of Animal Physiology, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Eveline M Ibeagha-Awemu
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Quebec, Canada
| |
Collapse
|
16
|
Amanova Z, Turyskeldy S, Kondybaeva Z, Sametova Z, Usembai A, Kerimbayev A, Bulatov Y. Assessment of Peste des Petits Ruminants Antibodies in Vaccinated Pregnant Ewes of Kazakh Breed Fine-Fleeced and Determination of the Decreasing Trend of Maternal Immunity in Their Lambs. Viruses 2023; 15:2054. [PMID: 37896831 PMCID: PMC10611327 DOI: 10.3390/v15102054] [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: 08/31/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
In this article, we first assessed peste des petits ruminants (PPR) antibodies in vaccinated pregnant ewes of Kazakh breed fine-fleeced immunized with the PPR vaccine and the duration of maternal immunity in their lambs. Ewes in the last trimester of pregnancy and gestation were immunized with a vaccine from the Nigeria 75/1 strain of the PPR virus (PPRV) produced by the Research Institute of Biological Safety Problems (RIBSP), Kazakhstan. Serum samples from lambs born from vaccinated and unvaccinated ewes were collected a week after birth and at intervals of 7 days for 18 weeks after birth. Serum samples collected from lambs were tested for PPR antibodies using competitive ELISA and virus neutralization test (VNT). Maternal antibodies (MAs) in lambs born from vaccinated ewes were detected for up to 18 weeks, with a tendency to decrease starting at week 14, and by the end of the experiment receded below the protective level (<1:8). In the blood serum of a 14-week-old lamb with MAs (1:8), post vaccination with a field dose (103 TCID50) of the vaccine against PPR, the titers of protective antibodies against PPRV increased to 1:16 on day 14 post vaccination, and the lamb was protected from infection with the field PPRV. A lamb of the same age with MAs in the 1:8 titer was 100% protected from infection with the field PPRV. Therefore, it is recommended that lambs of the Kazakh fine-wool breed be immunized from the age of 14 weeks or older to avoid a period of susceptibility.
Collapse
Affiliation(s)
- Zhanat Amanova
- Research Institute for Biological Safety Problems, Gvardeiskiy 080409, Kazakhstan; (S.T.); (Z.K.); (Z.S.); (A.U.); (A.K.); (Y.B.)
| | | | | | | | | | | | | |
Collapse
|
17
|
Jain J, Chaudhary Y, Gaur SK, Tembhurne P, Sekar SC, Dhanavelu M, Sehrawat S, Kaul R. Peste des petits ruminants virus non-structural V and C proteins interact with the NF-κB p65 subunit and modulate pro-inflammatory cytokine gene induction. J Gen Virol 2023; 104. [PMID: 37831061 DOI: 10.1099/jgv.0.001907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is known to induce transient immunosuppression in infected small ruminants by modulating several cellular pathways involved in the antiviral immune response. Our study shows that the PPRV-coded non-structural proteins C and V can interact with the cellular NF-κB p65 subunit. The PPRV-C protein interacts with the transactivation domain (TAD) while PPRV-V interacts with the Rel homology domain (RHD) of the NF-κB p65 subunit. Both viral proteins can suppress the NF-κB transcriptional activity and NF-κB-mediated transcription of cellular genes. PPRV-V protein expression can significantly inhibit the nuclear translocation of NF-κB p65 upon TNF-α stimulation, whereas PPRV-C does not affect it. The NF-κB-mediated pro-inflammatory cytokine gene expression is significantly downregulated in cells expressing PPRV-C or PPRV-V protein. Our study provides evidence suggesting a role of PPRV non-structural proteins V and C in the modulation of NF-κB signalling through interaction with the NF-κB p65 subunit.
Collapse
Affiliation(s)
- Juhi Jain
- Department of Microbiology, University of Delhi, South Campus, New Delhi, India
| | - Yash Chaudhary
- Department of Microbiology, University of Delhi, South Campus, New Delhi, India
| | - Sharad Kumar Gaur
- Department of Microbiology, University of Delhi, South Campus, New Delhi, India
| | | | | | | | - Sharvan Sehrawat
- Indian Institute of Science Education and Research, Mohali, India
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi, South Campus, New Delhi, India
| |
Collapse
|
18
|
Chaudhary Y, Jain J, Gaur SK, Tembhurne P, Chandrasekar S, Dhanavelu M, Sehrawat S, Kaul R. Nucleocapsid Protein (N) of Peste des petits ruminants Virus (PPRV) Interacts with Cellular Phosphatidylinositol-3-Kinase (PI3K) Complex-I and Induces Autophagy. Viruses 2023; 15:1805. [PMID: 37766213 PMCID: PMC10536322 DOI: 10.3390/v15091805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
Autophagy is an essential and highly conserved catabolic process in cells, which is important in the battle against intracellular pathogens. Viruses have evolved several ways to alter the host defense mechanisms. PPRV infection is known to modulate the components of a host cell's defense system, resulting in enhanced autophagy. In this study, we demonstrate that the N protein of PPRV interacts with the core components of the class III phosphatidylinositol-3-kinase (PI3K) complex-I and results in the induction of autophagy in the host cell over, thereby expressing this viral protein. Our data shows the interaction between PPRV-N protein and different core components of the autophagy pathway, i.e., VPS34, VPS15, BECN1 and ATG14L. The PPRV-N protein can specifically interact with VPS34 of the PI3K complex-I and colocalize with the proteins of PI3K complex-I in the same sub-cellular compartment, that is, in the cytoplasm. These interactions do not affect the intracellular localization of the different host proteins. The autophagy-related genes were transcriptionally modulated in PPRV-N-expressing cells. The expression of LC3B and SQSTM1/p62 was also modulated in PPRV-N-expressing cells, indicating the induction of autophagic activity. The formation of typical autophagosomes with double membranes was visualized by transmission electron microscopy in PPRV-N-expressing cells. Taken together, our findings provide evidence for the critical role of the N protein of the PPR virus in the induction of autophagy, which is likely to be mediated by PI3K complex-I of the host.
Collapse
Affiliation(s)
- Yash Chaudhary
- Department of Microbiology, University of Delhi, South Campus, New Delhi 110021, India; (Y.C.); (J.J.); (S.K.G.)
| | - Juhi Jain
- Department of Microbiology, University of Delhi, South Campus, New Delhi 110021, India; (Y.C.); (J.J.); (S.K.G.)
| | - Sharad Kumar Gaur
- Department of Microbiology, University of Delhi, South Campus, New Delhi 110021, India; (Y.C.); (J.J.); (S.K.G.)
| | - Prabhakar Tembhurne
- Department of Microbiology, Nagpur Veterinary College, Nagpur 440006, India;
| | - Shanmugam Chandrasekar
- Division of Virology, Indian Veterinary Research Institute, Mukteshwar, Nainital 263138, India; (S.C.); (M.D.)
| | - Muthuchelvan Dhanavelu
- Division of Virology, Indian Veterinary Research Institute, Mukteshwar, Nainital 263138, India; (S.C.); (M.D.)
| | - Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali 140306, India;
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi, South Campus, New Delhi 110021, India; (Y.C.); (J.J.); (S.K.G.)
| |
Collapse
|
19
|
Libbey JE, Fujinami RS. Morbillivirus: A highly adaptable viral genus. Heliyon 2023; 9:e18095. [PMID: 37483821 PMCID: PMC10362132 DOI: 10.1016/j.heliyon.2023.e18095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/26/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Over the course of human history, numerous diseases have been caused by the transmission of viruses from an animal reservoir into the human population. The viruses of the genus Morbillivirus are human and animal pathogens that emerged from a primordial ancestor a millennia ago and have been transmitting to new hosts, adapting, and evolving ever since. Through interaction with susceptible individuals, as yet undiscovered morbilliviruses or existing morbilliviruses in animal hosts could cause future zoonotic diseases in humans.
Collapse
|
20
|
Bessell PR, Salmon G, Schnier C, Tjasink K, Al-Riyami L, Peters A. A high level estimation of the net economic benefits to small-scale livestock producers arising from animal health product distribution initiatives. Front Vet Sci 2023; 10:1171989. [PMID: 37346278 PMCID: PMC10279859 DOI: 10.3389/fvets.2023.1171989] [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: 02/22/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Introduction A fundamental challenge for charities that facilitate distribution of animal health products to small-scale livestock producers (SSPs) in low and middle income countries (LMICs) is identifying the products and market mechanisms that provide the greatest positive impact for SSPs and estimating their associated impact. This paper describes a pragmatic approach to modeling the impact of market-led product distribution initiatives based on estimating the net economic benefit of administration of animal health products. Methods The model estimates the economic impact of diseases at the individual animal level for poultry, small ruminants, and cattle. The economic impact of mortality and growth inhibition associated with disease are then estimated in conjunction with the losses averted or recovered by preventing or treating the disease. Economic benefit is estimated in 2014-2017 values and also adjusted to 2023 values. The flexible model structure allows for addition of new geographies, new products, and increased granularity of modeled production systems. Results Applied to the Global Alliance for Livestock Veterinary Medicines (GALVmed) product distribution initiatives conducted in Africa and South Asia (SA) between 2014 and 2017, the model estimates an adjusted total net economic benefit of 139.9 million USD from sales of vaccines and poultry anthelminthics in these initiatives. Within SSA, the greatest net economic benefit was realized from East Coast fever and Newcastle disease vaccines, while in SA, peste des petits ruminants and Newcastle disease vaccines had the greatest net economic benefits. This translated to an adjusted $37.97 of net economic benefit on average per SSP customer, many of whom were small poultry producers. Discussion While the model currently estimates impacts from mortality and growth inhibition in livestock, there is the potential to extend it to cover impacts of further initiatives, including interventions targeted at diseases that impact production of milk, eggs, and reproduction.
Collapse
Affiliation(s)
| | - Gareth Salmon
- SEBI-L Supporting Evidence Based Interventions in Livestock, University of Edinburgh, Edinburgh, United Kingdom
| | - Christian Schnier
- SEBI-L Supporting Evidence Based Interventions in Livestock, University of Edinburgh, Edinburgh, United Kingdom
| | - Katharine Tjasink
- Global Alliance for Livestock Veterinary Medicines (GALVmed), Edinburgh, United Kingdom
| | - Lamyaa Al-Riyami
- Global Alliance for Livestock Veterinary Medicines (GALVmed), Edinburgh, United Kingdom
| | - Andrew Peters
- SEBI-L Supporting Evidence Based Interventions in Livestock, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
Milovanović M, Dietze K, Wernery U, Hoffmann B. Investigation of Potency and Safety of Live-Attenuated Peste des Petits Ruminant Virus Vaccine in Goats by Detection of Cellular and Humoral Immune Response. Viruses 2023; 15:1325. [PMID: 37376624 DOI: 10.3390/v15061325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/27/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
The peste des petits ruminant (PPR) virus is a transboundary virus found in small domestic ruminants that causes high morbidity and mortality in naive herds. PPR can be effectively controlled and eradicated by vaccinating small domestic ruminants with a live-attenuated peste des petits ruminant virus (PPRV) vaccine, which provides long-lasting immunity. We studied the potency and safety of a live-attenuated vaccine in goats by detecting their cellular and humoral immune responses. Six goats were subcutaneously vaccinated with a live-attenuated PPRV vaccine according to the manufacturer's instructions, and two goats were kept in contact. Following vaccination, the goats were monitored daily, and we recorded their body temperature and clinical score. Heparinized blood and serum were collected for a serological analysis, and swab samples and EDTA blood were collected to detect the PPRV genome. The safety of the used PPRV vaccine was confirmed by the absence of PPR-related clinical signs, a negative pen-side test, a low virus genome load as detected with RT-qPCR on the vaccinated goats, and the lack horizontal transmission between the in-contact goats. The strong humoral and cellular immune responses detected in the vaccinated goats showed that the live-attenuated PPRV vaccine has a strong potency in goats. Therefore, live-attenuated vaccines against PPR can be used to control and eradicate PRR.
Collapse
Affiliation(s)
- Milovan Milovanović
- Friedrich-Loeffler-Institut, Südufer 10, 17943 Greifswald-Insel Riems, Germany
| | - Klaas Dietze
- Friedrich-Loeffler-Institut, Südufer 10, 17943 Greifswald-Insel Riems, Germany
| | - Ulrich Wernery
- Central Veterinary Research Laboratory, Dubai P.O. Box 597, United Arab Emirates
| | - Bernd Hoffmann
- Friedrich-Loeffler-Institut, Südufer 10, 17943 Greifswald-Insel Riems, Germany
| |
Collapse
|
22
|
Siddiqui M, Globig A, Hoffmann B, Rahman MM, Islam MR, Chowdhury EH. Development of cell culture based peste des petits ruminants (PPR) virus vaccine candidate from Bangladeshi isolates. J Virol Methods 2023; 314:114690. [PMID: 36775141 DOI: 10.1016/j.jviromet.2023.114690] [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: 10/26/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
This study was conducted to develop a cell culture based PPR virus vaccine candidate using recent Bangladeshi strain of peste des petits ruminant's (PPR) virus. PPR virus was isolated from field outbreaks, confirmed by RT-PCR and used as viral inoculum for serial passaging in Vero cells for adaptation and attenuation. 60th serial passage had completed and RT-PCR and real time RT-PCR were done in every 5 passages for confirmation of PPR virus in tissue culture fluid (TCF). To assess the adaptation and attenuation cytopathology, virus titration, sequencing of both F and N genes and live animal experimentation were done. Different cellular alterations produced by PPR virus in infected Vero cells including syncytia formation, development of both intranuclear and intra cytoplasmic inclusion bodies and finally cell degradation are the indications of adaptation. The virus titre was found 2.5, 3.31, 3.55, 4.44, 4.71 and 6.5 Log10 TCID50/ml at 10th, 20th, 30th, 40th, 50th and 60th passages level respectively. In F gene sequence analysis it has been observed that few nucleotide (nt) and mino acid (aa) has been substituted as the effects of serial passaging of PPR virus in Vero cells. TCF at 60th passage level was found effective to produced protective antibody (Ab) titre in live animal experimentation. It is concluded that serially passaged and Vero cells adapted PPR virus TCF could be used as a vaccine candidate for further use to develop a potent & effective vaccine against PPR diseases.
Collapse
Affiliation(s)
- Msi Siddiqui
- Department of Anatomy & Histology, Faculty of Veterinary, Animal & Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh.
| | - Anja Globig
- Friedrich Loffler Institute, Federal Research Institute for Animal Health, Südufer 10, D-17493 Greifswald-Insel Riems, Germany.
| | - Bernd Hoffmann
- Friedrich Loffler Institute, Federal Research Institute for Animal Health, Südufer 10, D-17493 Greifswald-Insel Riems, Germany.
| | - M M Rahman
- Office of the Director General, Bangladesh Islamic Foundation, Ministry of Religious Affairs, Govt. of the Peoples Republic of Bangladesh, Dhaka, Bangladesh.
| | - M R Islam
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh.
| | - E H Chowdhury
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh.
| |
Collapse
|
23
|
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:vetsci10010056. [PMID: 36669056 PMCID: PMC9862675 DOI: 10.3390/vetsci10010056] [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: 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
|
24
|
Oyedele HA, Bodjo CS, Diallo H, Gelaw HB, Baziki JDD, Chitsungo E, Boukary CRM, Fikru H, Oladosu GA, Nwankpa N. Evaluation of monoclonal antibodies in immunofluorescence assay for rapid quality control of Peste des petits ruminants (PPR) vaccine. Small Rumin Res 2023. [DOI: 10.1016/j.smallrumres.2022.106865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
25
|
Wen B, Qi X, Lv D, Yang L, Tang P, Chang W, Han S, Yu S, Wei S, Xue Q, Wang J. Long noncoding RNA IRF1-AS is associated with peste des petits ruminants infection. Vet Res 2022; 53:89. [PMID: 36307867 PMCID: PMC9617334 DOI: 10.1186/s13567-022-01105-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/01/2022] [Indexed: 12/02/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute and highly contagious disease and has long been a significant threat to small ruminant productivity worldwide. However, the molecular mechanism underlying host-PPRV interactions remains unclear and the long noncoding RNAs (lncRNAs) regulation of PPR virus (PPRV) infection has rarely been reported so far. Here, we first demonstrated that PPRV infection can induce an obvious innate immune response in caprine endometrial epithelial cells (EECs) at 48 h post-infection (hpi) with an MOI of 3. Subsequently, we determined that PPRV infection is associated with 191 significantly differentially expressed (SDE) lncRNAs, namely, 137 upregulated and 54 downregulated lncRNAs, in caprine EECs compared with mock control cells at 48 hpi by using deep sequencing technology. Importantly, bioinformatics preliminarily analyses revealed that these DE lncRNAs were closely related to the immune response. Furthermore, we identified a system of lncRNAs related to the immune response and focused on the role of lncRNA 10636385 (IRF1-AS) in regulating the innate immune response. Interestingly, we found that IRF1-AS was a potent positive regulator of IFN-β and ISG production, which can significantly inhibit PPRV replication in host cells. In addition, our data revealed that IRF1-AS was positively correlated with its potential target gene, IRF1, which enhanced the activation of IRF3 and the expression of ISGs and interacted with IRF3. This study suggests that IRF1-AS could be a new host factor target for developing antiviral therapies against PPRV infection.
Collapse
Affiliation(s)
- Bo Wen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Daiyue Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.,China Institute of Veterinary Drug Control, Beijing, 100000, China
| | - Lulu Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Pan Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Wenchi Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shuizhong Han
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shengmeng Yu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shaopeng Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, 100000, China.
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
26
|
Kumar R, Chander Y, Khandelwal N, Verma A, Rawat KD, Shringi BN, Pal Y, Tripathi BN, Barua S, Kumar N. ROCK1/MLC2 inhibition induces decay of viral mRNA in BPXV infected cells. Sci Rep 2022; 12:17811. [PMID: 36280692 PMCID: PMC9592580 DOI: 10.1038/s41598-022-21610-9] [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: 03/02/2022] [Accepted: 09/29/2022] [Indexed: 01/19/2023] Open
Abstract
Rho-associated coiled-coil containing protein kinase 1 (ROCK1) intracellular cell signaling pathway regulates cell morphology, polarity, and cytoskeletal remodeling. We observed the activation of ROCK1/myosin light chain (MLC2) signaling pathway in buffalopox virus (BPXV) infected Vero cells. ROCK1 depletion by siRNA and specific small molecule chemical inhibitors (Thiazovivin and Y27632) resulted in a reduced BPXV replication, as evidenced by reductions in viral mRNA/protein synthesis, genome copy numbers and progeny virus particles. Further, we demonstrated that ROCK1 inhibition promotes deadenylation of viral mRNA (mRNA decay), mediated via inhibiting interaction with PABP [(poly(A)-binding protein] and enhancing the expression of CCR4-NOT (a multi-protein complex that plays an important role in deadenylation of mRNA). In addition, ROCK1/MLC2 mediated cell contraction, and perinuclear accumulation of p-MLC2 was shown to positively correlate with viral mRNA/protein synthesis. Finally, it was demonstrated that the long-term sequential passage (P = 50) of BPXV in the presence of Thiazovivin does not select for any drug-resistant virus variants. In conclusion, ROCK1/MLC2 cell signaling pathway facilitates BPXV replication by preventing viral mRNA decay and that the inhibitors targeting this pathway may have novel therapeutic effects against buffalopox.
Collapse
Affiliation(s)
- Ram Kumar
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India ,grid.464655.00000 0004 1768 5915Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India ,grid.418105.90000 0001 0643 7375Present Address: Animal Science Division, Indian Council of Agricultural Research, Krishi Bhawan, New Delhi, India
| | - Yogesh Chander
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India ,grid.418105.90000 0001 0643 7375Present Address: Animal Science Division, Indian Council of Agricultural Research, Krishi Bhawan, New Delhi, India ,grid.411892.70000 0004 0500 4297Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana India
| | - Nitin Khandelwal
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Assim Verma
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Krishan Dutt Rawat
- grid.411892.70000 0004 0500 4297Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana India
| | - Brij N. Shringi
- grid.464655.00000 0004 1768 5915Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Yash Pal
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Bhupendra N. Tripathi
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India ,grid.418105.90000 0001 0643 7375Present Address: Animal Science Division, Indian Council of Agricultural Research, Krishi Bhawan, New Delhi, India
| | - Sanjay Barua
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Naveen Kumar
- grid.462601.70000 0004 1768 7902Present Address: National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| |
Collapse
|
27
|
Lelisa K, Chibssa TR, Desissa F, Emiyu K, Muluneh A, Lobago DS, Gebreweld DS, Debebe K, Mohammed AA. Evaluation of diagnostic performance of H-based blocking ELISA for specific detection of peste des petits ruminants in domestic sheep, goats, cattle and camels. BMC Microbiol 2022; 22:254. [PMID: 36266634 PMCID: PMC9585824 DOI: 10.1186/s12866-022-02669-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Peste des petits ruminants virus (PPRV) causes a highly devastating disease of sheep and goats, peste des petits ruminants (PPR), which is targeted for global control and eradication by 2030. The serological diagnostic tool kits for accurate diagnosis of PPR have inherent strengths and weaknesses that require parallel validation and optimization across animal species. Thus, the objective of this study was to evaluate diagnostic performance of haemagglutinin based PPR blocking ELISA (HPPR- b-ELISA), that was developed by Africa Union Pan African Veterinary Vaccine Center for specific detection of anti- PPRV antibodies. METHODS In preliminarily investigation, diagnostic performance of the HPPR-b-ELISA®, commercial PPR competition ELISA (c-ELISA) and virus neutralization test (VNT) were compared for the detection of anti-PPRV antibodies in goats, sheep, cattle and camels. RESULTS The sensitivity and specificity of HPPR- b-ELISA® were 79.55 and 99.74%, respectively, compared to c-ELISA. The HPPR- b-ELISA® was in perfect agreement (κ = 0.86) with the c-ELISA in all sera collected from goats, sheep and cattle. There was almost perfect agreement between the species of goats (κ = 0.82) and sheep (κ = 0.98), while the agreement was substantial in cattle (κ = 0.78) and no agreement was observed in camels (κ = 0.00). Similarly, the sensitivity and specificity of the HPPR b-ELISA were 80 and 96.36%, respectively compared to VNT with almost perfect agreement in goats (κ = 0.83) and sheep (κ = 0.89), moderate in cattle (κ = 0.50) and none in camels (κ = 0.00). CONCLUSION Our study revealed that HPPR- b-ELISA is a suitable and valid method that can alternatively be used for screening and monitoring of PPR in sheep, goats and cattle except for camels.
Collapse
Affiliation(s)
- Kumela Lelisa
- Animal Health Institute, P.O Box 04, Sebeta, Oromia, Ethiopia.
| | | | - Fanta Desissa
- College of Veterinary Medicine and Agriculture, Department of Microbiology, Immunology and Veterinary Public health, Addis Ababa University, P.O. Box, 34, Addis Ababa, Ethiopia
| | - Kemal Emiyu
- Animal Health Institute, P.O Box 04, Sebeta, Oromia, Ethiopia
| | - Ayelech Muluneh
- Animal Health Institute, P.O Box 04, Sebeta, Oromia, Ethiopia
| | | | | | - Kebede Debebe
- Animal Health Institute, P.O Box 04, Sebeta, Oromia, Ethiopia
| | | |
Collapse
|
28
|
Abstract
Peste des petits ruminants virus (PPRV) infection leads to autophagy, and the molecular mechanisms behind this phenomenon are unclear. Here, we demonstrate that PPRV infection results in morphological changes of the endoplasmic reticulum (ER) and activation of activating transcription factor 6 (ATF6) of the ER stress unfolded protein response (UPR). Knockdown of ATF6 blocked the autophagy process, suggesting ATF6 is necessary for PPRV-mediated autophagy induction. Further study showed that PPRV infection upregulates expression of the ER-anchored adaptor protein stimulator of interferon genes (STING), which is well-known for its pivotal roles in restricting DNA viruses. Knockdown of STING suppressed ATF6 activation and autophagy induction, implying that STING functions upstream of ATF6 to induce autophagy. Moreover, the STING-mediated autophagy response originated from the cellular pattern recognition receptor melanoma differentiation-associated gene 5 (MDA5). The absence of MDA5 abolished the upregulation of STING and the activation of autophagy. The deficiency of autophagy-related genes (ATG) repressed the autophagy process and PPRV replication, while it had no effect on MDA5 or STING expression. Overall, our work revealed that MDA5 works upstream of STING to activate ATF6 to induce autophagy. IMPORTANCEPPRV infection induces cellular autophagy; however, the intracellular responses and signaling mechanisms that occur upon PPRV infection are obscure, and whether innate immune responses are linked with autophagy to regulate viral replication is largely unknown. Here, we uncovered that the innate immune sensor MDA5 initiated the signaling cascade by upregulating STING, which is best known for its role in anti-DNA virus infection by inducing interferon expression. We first provide evidence that STING regulates PPRV replication by activating the ATF6 pathway of unfolded protein responses (UPRs) to induce autophagy. Our results revealed that in addition to mediating responses to foreign DNA, STING can cross talk with MDA5 to regulate the cellular stress response and autophagy induced by RNA viruses; thus, STING works as an adaptor protein for cellular stress responses and innate immune responses. Modulation of STING represents a promising approach to control both DNA and RNA viruses.
Collapse
|
29
|
Sultana S, Pervin M, Sultana N, Siddique MP, Islam MR, Khan MAHNA. Identification of peste des petits ruminants virus along with co-infecting diseases of goats in Bangladesh. J Adv Vet Anim Res 2022; 9:463-470. [PMID: 36382033 PMCID: PMC9597929 DOI: 10.5455/javar.2022.i615] [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: 04/05/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Peste des petits ruminants (PPR) virus is the main infectious cause of goat mortality in Bangladesh, and co-infection may make diseases more severe. This study aimed to detect PPR and co-infecting diseases in goats. MATERIALS AND METHODS One hundred goats suspected to be infected with the PPR virus were collected from various areas of Mymensingh district, Bangladesh. A systemic post-mortem examination was carried out on PPR-suspected goats. Lungs, spleen, and lymph nodes (pre-scapular) were used for ribonucleic acid extraction, whereas lungs and mesenteric lymph nodes were used for deoxyribonucleic acid extraction. Seven-pair primer sets were used for molecular detection of pathogens specific for PPR, goat pox, contagious ecthyma (Orf), foot and mouth disease (FMD) virus, Klebsiella sp., and Mycobacterium sp. Reverse transcriptase-polymerase chain reaction (RT-PCR) or polymerase chain reaction (PCR) were used to find the exact cause. RESULTS Out of 100 PPR-suspected goats examined, 55 goats were confirmed as PPR-detected by RT-PCR. Among the 55 PPR-positive goats, 2 were co-infected with goat pox, 2 with tuberculosis, 10 with Klebsiella sp. infection, and 6 with FMD as detected by PCR and RT-PCR. Moreover, 12 goats were co-infected with PPRV and fascioliasis. CONCLUSION About 58% of PPR virus-infected goats were co-infected with other organisms. There is a need to design technology to detect the state of co-infectivity at its early onset and future preventive and therapeutic strategies for co-infecting diseases. This is the first study in Bangladesh to describe co-infecting diseases of goats along with PPR.
Collapse
Affiliation(s)
- Sajeda Sultana
- Department of Pathology, Faculty of Animal Science and Veterinary Medicine, Sher e Bangla Agricultural University, Dhaka, Bangladesh
| | - Munmun Pervin
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Nazneen Sultana
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mahbubul Pratik Siddique
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Rafiqul Islam
- Animal Health Division, Bangladesh Agricultural Research Council, Dhaka, Bangladesh
| | | |
Collapse
|
30
|
A Morbillivirus Infection Shifts DC Maturation Toward a Tolerogenic Phenotype to Suppress T Cell Activation. J Virol 2022; 96:e0124022. [PMID: 36094317 PMCID: PMC9517701 DOI: 10.1128/jvi.01240-22] [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: 11/20/2022] Open
Abstract
Viruses have evolved numerous strategies to impair immunity so that they can replicate more efficiently. Among those, the immunosuppressive effects of morbillivirus infection can be particularly problematic, as they allow secondary infections to take hold in the host, worsening disease prognosis. In the present work, we hypothesized that the highly contagious morbillivirus peste des petits ruminants virus (PPRV) could target monocytes and dendritic cells (DC) to contribute to the immunosuppressive effects produced by the infection. Monocytes isolated from healthy sheep, a natural host of the disease, were able be infected by PPRV and this impaired the differentiation and phagocytic ability of immature monocyte-derived DC (MoDC). We also assessed PPRV capacity to infect differentiated MoDC. Ovine MoDC could be productively infected by PPRV, and this drastically reduced MoDC capacity to activate allogeneic T cell responses. Transcriptomic analysis of infected MoDC indicated that several tolerogenic DC signature genes were upregulated upon PPRV infection. Furthermore, PPRV-infected MoDC could impair the proliferative response of autologous CD4+ and CD8+ T cell to the mitogen concanavalin A (ConA), which indicated that DC targeting by the virus could promote immunosuppression. These results shed new light on the mechanisms employed by morbillivirus to suppress the host immune responses. IMPORTANCE Morbilliviruses pose a threat to global health given their high infectivity. The morbillivirus peste des petits ruminants virus (PPRV) severely affects small-ruminant-productivity and leads to important economic losses in communities that rely on these animals for subsistence. PPRV produces in the infected host a period of severe immunosuppression that opportunistic pathogens exploit, which worsens the course of the infection. The mechanisms of PPRV immunosuppression are not fully understood. In the present work, we demonstrate that PPRV can infect professional antigen-presenting cells called dendritic cells (DC) and disrupt their capacity to elicit an immune response. PPRV infection promoted a DC activation profile that favored the induction of tolerance instead of the activation of an antiviral immune response. These results shed new light on the mechanisms employed by morbilliviruses to suppress the immune responses.
Collapse
|
31
|
Chen Y, Wang T, Yang Y, Fang Y, Zhao B, Zeng W, Lv D, Zhang L, Zhang Y, Xue Q, Chen X, Wang J, Qi X. Extracellular vesicles derived from PPRV-infected cells enhance signaling lymphocyte activation molecular (SLAM) receptor expression and facilitate virus infection. PLoS Pathog 2022; 18:e1010759. [PMID: 36084159 PMCID: PMC9491601 DOI: 10.1371/journal.ppat.1010759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 09/21/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. PPRV is lymphotropic in nature and SLAM was identified as the primary receptor for PPRV and other Morbilliviruses. Many viruses have been demonstrated to engage extracellular vesicles (EVs) to facilitate their replication and pathogenesis. Here, we provide evidence that PPRV infection significantly induced the secretion levels of EVs from goat PBMC, and that PPRV-H protein carried in EVs can enhance SLAM receptor expression in the recipient cells via suppressing miR-218, a negative miRNA directly targeting SLAM gene. Importantly, EVs-mediated increased SLAM expression enhances PPRV infectivity as well as the expression of various cytokines related to SLAM signaling pathway in the recipient cells. Moreover, our data reveal that PPRV associate EVs rapidly entry into the recipient cells mainly through macropinocytosis pathway and cooperated with caveolin- and clathrin-mediated endocytosis. Taken together, our findings identify a new strategy by PPRV to enhance virus infection and escape innate immunity by engaging EVs pathway. Peste des petitsruminants virus (PPRV) infection induces a transient but severe immunosuppression in the host, which threatens both small livestock and endangered susceptible wildlife populations in many countries. Despite extensive research, the mechanism underlying pathogenesis of PPRV infection remains elusive. Our data provide the first direct evidence that the EVs derived from PPRV-infected cells are involved in PPRV replication. In this study, the EVs derived from PPRV-infected goat PBMCs can enhance SLAM expression in the recipient cells, and more importantly, EVs-mediated increased SLAM expression enhances PPRV replication as well as the expression of various cytokines related to SLAM signaling pathway in the recipient cells. Taken together, our research has provided new insight into understanding the effect of EVs on PPRV replication and pathogenesis, and revealed a potential therapeutic target for antiviral intervention.
Collapse
Affiliation(s)
- Yan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ting Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yang Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Fang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Bao Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Shaanxi Animal Disease Control Center, Xi’an, China
| | - Wei Zeng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Daiyue Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Leyan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, China
| | - Xiwen Chen
- Animal Disease Prevention and Control & Healthy Breeding Engineering Technology Research Center, Mianyang Normal University, Mianyang, Sichuan, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (JW); (XQ)
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (JW); (XQ)
| |
Collapse
|
32
|
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: 3.0] [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
|
33
|
Jemberu WT, Knight-Jones TJD, Gebru A, Mekonnen SA, Yirga A, Sibhatu D, Rushton J. Economic impact of a peste des petits ruminants outbreak and vaccination cost in northwest Ethiopia. Transbound Emerg Dis 2022; 69:e2084-e2092. [PMID: 35353947 PMCID: PMC9790723 DOI: 10.1111/tbed.14544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 12/30/2022]
Abstract
Peste des petits ruminants (PPR) is an important endemic disease of small ruminants in Ethiopia. While vaccination is widely used in the country to control the disease, quantitative estimates of the actual economic losses due to outbreaks and costs of vaccination are scarce. This study assessed the economic impact and costs of PPR vaccination in Metema district, northwest Ethiopia. The economic impact of the disease was estimated from an outbreak investigation including interviews with 233 smallholder farmers in PPR-affected kebeles (subdistricts). The cost of PPR vaccination was obtained from vaccination programs in six kebeles of the district and from secondary data in the district veterinary office. In the investigated PPR outbreak, animal-level PPR morbidity and mortality rates were 51% and 22%, respectively, in sheep and 51% and 25%, respectively, in goats. The flock level morbidity rate was 83% for sheep flocks and 87% for goat flocks. The mean flock level loss was Ethiopian Birr (ETB) 7835 (USD 329 in 2018 average exchange rate) (95% CI: 5954-9718) for affected sheep flocks and ETB 7136 (USD 300) (95% CI: 5869-8404) for affected goat flocks. The losses in all study flocks during the outbreak were ETB 319 (USD 13.4) per sheep and ETB 306 (USD 12.9) per goat. Mortality accounted for more than 70% of the total losses in both sheep and goat flocks. Vaccination costs for PPR were estimated at ETB 3 per correctly vaccinated animal. Based on the estimated animal-level direct economic losses and vaccination cost, it can be conjectured that vaccination will pay if a district PPR outbreak occurs more than once every 13 years. This does not account for additional benefits from vaccine-derived herd immunity reducing disease burden in the wider population. In conclusion, PPR caused high morbidity and mortality in the affected flocks and resulted in high economic losses, equivalent to 14% of annual household income, dramatically affecting the livelihoods of affected flock owners. The vaccination practised in the district is likely to have a positive economic return, with strengthened vaccination programmes bringing reduced economic impact and improved livelihoods.
Collapse
Affiliation(s)
- Wudu T Jemberu
- Department of Veterinary Epidemiology and Public Health, University of Gondar, Gondar, Ethiopia
- International Livestock Research Institute, Addis Ababa, Ethiopia
| | | | - Alemseged Gebru
- Department of Veterinary Epidemiology and Public Health, University of Gondar, Gondar, Ethiopia
| | - Sefinew A Mekonnen
- Department of Veterinary Epidemiology and Public Health, University of Gondar, Gondar, Ethiopia
| | - Andnet Yirga
- Department of Veterinary Science, Bahir Dar University, Bahir Dar, Ethiopia
| | - Demeke Sibhatu
- National Animal Health Diagnostic and Investigation Center, Sebeta, Ethiopia
| | - Jonathan Rushton
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
34
|
Moudgil P, Kumar R, Jangir BL, Gupta R, Vaishali, Jindal N. Epidemiology, risk factors and molecular characterization of small ruminant morbillivirus in Haryana, India. Res Vet Sci 2022; 151:164-174. [PMID: 36041310 DOI: 10.1016/j.rvsc.2022.08.013] [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: 02/07/2022] [Revised: 07/02/2022] [Accepted: 08/11/2022] [Indexed: 10/15/2022]
Abstract
Peste des petitis ruminants is an economically important transboundary and notifiable viral disease of sheep and goats. In this study, 14 PPR suspected outbreaks among sheep and goats were investigated in four districts of Haryana, India, during July 2020 to October, 2021. The causative agent of the disease; small ruminant morbillivirus was detected by Reverse Transcription Polymerase Chain Reaction targeting full gene sequences of fusion protein gene and confirmed by sequencing. The overall morbidity and cumulative mortality in these outbreaks were 37.56% and 12.09%, respectively. Risk factor analysis identified significant difference in mortality based on age with higher mortality in young ones; 21% as compared to adults; 7.55%. Analysis of the vaccination status revealed significant difference in morbidity and mortality with higher morbidity and mortality in un-vaccinated animals as compared to vaccinated ones. The sequencing and phylogenetic analysis of representative samples revealed that the strains of the present study fall in lineage IV (96.6-99.1%) along with other Indian isolates but made a separate cluster (sub-lineage). The comparison of deduced amino acid (aa) sequence analysis of fusion protein of circulating field strains with reference vaccine strain and other lineage IV strains revealed four N-linked glycosylation sites instead of three. The findings of the present study revealed changes in fusion protein of some of the circulating field strains of SRMV in Haryana, India. Further detailed studies are warranted to delineate the molecular details of these circulating field strains and to evaluate the effectiveness of currently used vaccine against these mutated strains.
Collapse
Affiliation(s)
- Pallavi Moudgil
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India
| | - Ramesh Kumar
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India
| | - Babu Lal Jangir
- Department of Veterinary Pathology, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India
| | - Renu Gupta
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India.
| | - Vaishali
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India
| | - Naresh Jindal
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India
| |
Collapse
|
35
|
Chander Y, Kumar R, Verma A, Khandelwal N, Nagori H, Singh N, Sharma S, Pal Y, Puvar A, Pandit R, Shukla N, Chavada P, Tripathi BN, Barua S, Kumar N. Resistance evolution against host-directed antiviral agents: Buffalopox virus switches to use p38-ϒ under long-term selective pressure of an inhibitor targeting p38-α. Mol Biol Evol 2022; 39:6668988. [PMID: 35975687 PMCID: PMC9435063 DOI: 10.1093/molbev/msac177] [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] [Indexed: 11/25/2022] Open
Abstract
Host-dependency factors have increasingly been targeted to minimize antiviral drug resistance. In this study, we have demonstrated that inhibition of p38 mitogen-activated protein kinase (a cellular protein) suppresses buffalopox virus (BPXV) protein synthesis by targeting p38-MNK1-eIF4E signaling pathway. In order to provide insights into the evolution of drug resistance, we selected resistant mutants by long-term sequential passages (P; n = 60) in the presence of p38 inhibitor (SB239063). The P60-SB239063 virus exhibited significant resistance to SB239063 as compared to the P60-Control virus. To provide mechanistic insights on the acquisition of resistance by BPXV-P60-SB239063, we generated p38-α and p38-ϒ (isoforms of p38) knockout Vero cells by CRISPR/Cas9-mediated genome editing. It was demonstrated that unlike the wild type (WT) virus which is dependent on p38-α isoform, the resistant virus (BPXV-P60-SB239063) switches over to use p38-ϒ so as to efficiently replicate in the target cells. This is a rare evidence wherein a virus was shown to bypass the dependency on a critical cellular factor under selective pressure of a drug.
Collapse
Affiliation(s)
- Yogesh Chander
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.,Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Ram Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Assim Verma
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.,Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Nitin Khandelwal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Himanshu Nagori
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Namita Singh
- Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Shalini Sharma
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hiar, Haryana, India
| | - Yash Pal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Apurvasinh Puvar
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Rameshchandra Pandit
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Nitin Shukla
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Priyank Chavada
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Bhupendra N Tripathi
- Gujarat Biotechnology Research Centre, Department of Science & Technology, Government of Gujarat, India
| | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| |
Collapse
|
36
|
Effects of Boswellia species on viral infections with particular attention to SARS-CoV-2. Inflammopharmacology 2022; 30:1541-1553. [PMID: 35882701 PMCID: PMC9321285 DOI: 10.1007/s10787-022-01037-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/07/2022] [Indexed: 12/12/2022]
Abstract
The emergence of pathogenic viruses is a worldwide frequent cause of diseases and, therefore, the design of treatments for viral infections stands as a significant research topic. Despite many efforts, the production of vaccines is faced with many obstacles and the high rate of viral resistance caused a severe reduction in the efficacy of antiviral drugs. However, the attempt of developing novel natural drugs, as well as the exertion of medicinal plants, may be an applicable solution for the treatment of viral diseases. Boswellia species exhibited a wide range of pharmacological activities in various conditions such as bronchial asthma, rheumatism, and Crohn’s illness. Additionally, pharmacological studies reported the observance of practical antiviral activities from different parts of this substance, especially the oleo-gum-resin. Therefore, this work provided an overview on the antiviral properties of Boswellia species and their potential therapeutic effects in the field of COVID-19 pandemic.
Collapse
|
37
|
Expansion in host dynamics of peste des petits ruminants: Potential attribute of outbreaks in disease-endemic settings. Acta Trop 2022; 234:106609. [PMID: 35850237 DOI: 10.1016/j.actatropica.2022.106609] [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: 03/21/2022] [Revised: 06/22/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022]
Abstract
Since the first case report in 1942, the peste-des-petits-ruminants virus (PPRV) has been causing infection in a wide range of susceptible hosts, particularly in disease-endemic regions. In the last 40 years, various reports highlighted the evidence of disease and viral genome in around 46 animal species from nine diverse families, including Bovidae, Cervidae, Camelidae, Suidae, Canidae, Felidae, Muridae, and Elephantidae. This evidence of clinical and/ or subclinical infection and the presence of the virus in an extended range of susceptible hosts emphasizes the cross-species transmission that remains a significant obstacle to effective control, particularly in disease-endemic regions. Therefore, a better understanding of virus transmission, host susceptibility, and epidemiological investigation of the disease is crucial to achieving the goals of efficient disease control and eradication programs initiated by OIE and FAO in various diseases-endemic regions. Nevertheless, the propensity of PPRV to inter- and intra-transmission may be a possible constraint in disease control strategies in terms of the new outbreak with the involvement of unusual or novel hosts. Considering this aspect, we tried to summarize the scattered data on PPR in available information about the susceptibility of a wide range of wildlife species, large ruminants, camels, and unusual hosts.
Collapse
|
38
|
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: 0] [Impact Index Per Article: 0] [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
|
39
|
Tang J, Tang A, Du H, Jia N, Zhu J, Li C, Meng C, Liu G. Peste des Petits Ruminants Virus Exhibits Cell-Dependent Interferon Active Response. Front Cell Infect Microbiol 2022; 12:874936. [PMID: 35711660 PMCID: PMC9195304 DOI: 10.3389/fcimb.2022.874936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/04/2022] [Indexed: 12/02/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute and highly pathogenic infectious disease caused by peste des petits ruminants virus (PPRV), which can infect goats and sheep and poses a major threat to the small ruminants industry. The innate immune response plays an important role as a line of defense against the virus. The effect of PPRV on the active innate immune response has been described in several studies, with different conclusions. We infected three goat-derived cell lines with PPRV and tested their innate immune response. PPRV proliferated in caprine endometrial epithelial cells (EECs), caprine skin fibroblasts cells (GSFs), and goat fibroblast cells (GFs), and all cells expressed interferon (IFN) by poly (I: C) stimulation. PPRV infection stimulated expression of type I and type III IFN on EECs, and expression of the latter was significantly stronger, but IFN was not stimulated in fibroblasts (GSFs and GFs). Our results suggested that the effect of PPRV on IFN was cell-type specific. Nine IFN-stimulated genes (ISGs) were detected in EECs, but only ISG15 and RSAD2 were significantly upregulated. The effects of PPRV on IFN and IFN-induced ISGs were cell-type specific, which advances our understanding of the innate immune response induced by PPRV and creates new possibilities for the control of PPRV infection.
Collapse
|
40
|
PPRV-Induced Autophagy Facilitates Infectious Virus Transmission by the Exosomal Pathway. J Virol 2022; 96:e0024422. [PMID: 35319226 DOI: 10.1128/jvi.00244-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. We showed previously that PPRV induced sustained autophagy for their replication in host cells. Many studies have shown that exosomes released from virus-infected cells contain a variety of viral and host cellular factors that are able to modulate the recipient's cellular response and result in productive infection of the recipient host. Here, we show that PPRV infection results in packaging of the viral genomic RNA and partial viral proteins into exosomes of Vero cells and upregulates exosome secretion. We provide evidence showing that the exosomal viral cargo can be transferred to and establish productive infection in a new target cell. Importantly, our study reveals that PPRV-induced autophagy enhances exosome secretion and exosome-mediated virus transmission. Additionally, our data show that TSG101 may be involved in the sorting of the infectious PPRV RNA into exosomes to facilitate the release of PPRV through the exosomal pathway. Taken together, our results suggest a novel mechanism involving autophagy and exosome-mediated PPRV intercellular transmission. IMPORTANCE Autophagy plays an important role in PPRV pathogenesis. The role of exosomes in viral infections is beginning to be appreciated. The present study examined the role of autophagy in secretion of infectious PPRV from Vero cells. Our data provided the first direct evidence that ATG7-mediated autophagy enhances exosome secretion and exosome-mediated PPRV transmission. TSG101 may be involved in the sorting of the infectious PPRV RNA genomes into exosomes to facilitate the release of PPRV through the exosomal pathway. Inhibition of PPRV-induced autophagy or TSG101 expression could be used as a strategy to block exosome-mediated virus transmission.
Collapse
|
41
|
Lysholm S, Lindahl JF, Munyeme M, Misinzo G, Mathew C, Alvåsen K, Dautu G, Linde S, Mitternacht L, Olovsson E, Wilén E, Berg M, Wensman JJ. Crossing the Line: Seroprevalence and Risk Factors for Transboundary Animal Diseases Along the Tanzania-Zambia Border. Front Vet Sci 2022; 9:809128. [PMID: 35359681 PMCID: PMC8962627 DOI: 10.3389/fvets.2022.809128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/07/2022] [Indexed: 11/16/2022] Open
Abstract
Transboundary pathogens pose a threat to livelihood security in countries such as Zambia and Tanzania. This study aimed to investigate the seroprevalence of peste des petits ruminants virus (PPRV), foot and mouth disease virus (FMDV), sheep and goat pox virus (SGPV), Rift Valley fever virus (RVFV) and Brucella spp. in sheep and goats along the Tanzania-Zambia border. Another aim was to assess the association between certain predictor variables and seroprevalence, focusing on trade and proximity to an international border, to a town and to the Tanzania-Zambia highway. During September-October 2018, 486 serum samples from small ruminants in Zambia and 491 in Tanzania were collected and analyzed using enzyme-linked immunosorbent assays (ELISA). A questionnaire focused on management strategies was administered to each household. The animal-level seroprevalence in Zambia was 0.21% [95% confidence interval (CI) (0.01–1.14) for PPRV, 1.03% (95% CI 0.33–2.39) for FMDV, 0% (95% CI 0–0.76) for SGPV, 2.26% (95% CI 1.14–4.01) for RVFV and 1.65% (95% CI 0.71–3.22) for Brucella spp.]. In Tanzania, animal-level seroprevalence was 2.85% (95% CI 1.57–4.74) for PPRV, 16.9% (95% CI 13.7–20.5) for FMDV, 0.20% (95% CI 0.01–1.13) for SGPV, 3.26% (95% CI 1.87–5.24) for RVFV and 20.0% (95% CI 14.5–26.5) for Brucella spp. For PPRV (OR 6.83, 95% CI 1.37–34.0, p = 0.019) and FMDV (OR 5.68, 95% CI 1.58–20.3, p = 0.008), herds situated more than 30 km from an international border were more likely to be seropositive, while being located 10–30 km (OR 4.43, 95% CI 1.22–16.1 p = 0.024) from a border was identified as a risk factor for Brucella spp. For FMDV (OR 79.2, 95% CI 4.52–1388.9, p = 0.003), being situated within 30 km from a town was associated with seropositivity. Furthermore, contact with wild ruminants (OR 18.2, 95% CI 1.36–244), and the presence of sheep in the household (OR 5.20, 95% CI 1.00–26.9, p = 0.049), was associated with seropositivity for PPRV, and FMDV. No significant associations between trade or distance to the Tan-Zam highway and seroprevalence were found. We recommend that the impact of trade and proximity to borders, towns and roads should be further evaluated in larger studies, ideally incorporating aspects such as temporal trade fluctuations.
Collapse
Affiliation(s)
- Sara Lysholm
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
- *Correspondence: Sara Lysholm
| | - Johanna F. Lindahl
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Biosciences, International Livestock Research Institute, Nairobi, Kenya
| | - Musso Munyeme
- Department of Disease Control, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Gerald Misinzo
- SACIDS Foundation for One Health, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Coletha Mathew
- Department of Veterinary Anatomy and Pathology, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Karin Alvåsen
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - George Dautu
- Department of Veterinary Services Ministry of Fisheries and Livestock, Central Veterinary Research Institute, Lusaka, Zambia
| | - Siri Linde
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Lydia Mitternacht
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Emelie Olovsson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Elsa Wilén
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Mikael Berg
- Department of Biomedical Science and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jonas J. Wensman
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Department of Epidemiology and Disease Control, National Veterinary Institute, Uppsala, Sweden
| |
Collapse
|
42
|
Predicting the possibility of African horse sickness (AHS) introduction into China using spatial risk analysis and habitat connectivity of Culicoides. Sci Rep 2022; 12:3910. [PMID: 35273211 PMCID: PMC8913660 DOI: 10.1038/s41598-022-07512-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
African horse sickness (AHS) is a devastating equine infectious disease. On 17 March 2020, it first appeared in Thailand and threatened all the South-East Asia equine industry security. Therefore, it is imperative to carry out risk warnings of the AHS in China. The maximum entropy algorithm was used to model AHS and Culicoides separately by using climate and non-climate variables. The least cost path (LCP) method was used to analyze the habitat connectivity of Culicoides with the reclassified land cover and altitude as cost factors. The models showed the mean area under the curve as 0.918 and 0.964 for AHS and Culicoides. The prediction result map shows that there is a high risk area in the southern part of China while the habitats of the Culicoides are connected to each other. Therefore, the risk of introducing AHS into China is high and control of the border area should be strengthened immediately.
Collapse
|
43
|
Niu B, Liang R, Zhang S, Sun X, Li F, Qiu S, Zhang H, Bao S, Zhong J, Li X, Chen Q. Spatiotemporal characteristics analysis and potential distribution prediction of peste des petits ruminants (PPR) in China from 2007-2018. Transbound Emerg Dis 2021; 69:2747-2763. [PMID: 34936210 DOI: 10.1111/tbed.14426] [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: 04/26/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022]
Abstract
Peste des petits ruminants (PPR) is a highly infectious disease that mainly infects small ruminants. To date, PPR has been confirmed in more than 70 countries. In China, PPR has occurred in more than 20 provinces and cities. In this study, based on geographic information system (GIS), spatial analysis was used to examine the occurrence of PPR in China from 2007 to 2018. The results showed that PPR first occurred in Tibet and gradually spread to other provinces. The outbreaks of PPR were concentrated in 2014, 2015 and 2018. Combining climate factors with the maximum entropy (MaxEnt), the results also suggested that the potential risk areas of PPR outbreaks in China were mainly Jiangsu, Yunnan and Anhui in Southeast China. Finally, a phylogenetic tree was used to analyse the evolutionary relationship between the peste des petits ruminants virus (PPRV) in China and the global ones, and it was found that the one in China had a close genetic relationship with the one in Mongolia, India and Bangladesh. Understanding and forecasting the distribution of PPR in China will help policymakers develop targeted monitoring plans. Likewise, analysing the global PPRV epidemic trends will play an important role in the elimination and prevention of PPR.
Collapse
Affiliation(s)
- Bing Niu
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Ruirui Liang
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Shuwen Zhang
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Xiaodong Sun
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Fuchen Li
- College of Art and Science, Vanderbilt University, Nashville, Tennessee, USA
| | - Songyin Qiu
- Chinese Academy of Inspection and Quarantine, Beijing, P.R. China
| | - Hui Zhang
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Songhao Bao
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Junjie Zhong
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Xinxiang Li
- College of Sciences, Shanghai University, Shanghai, P.R. China
| | - Qin Chen
- School of Life Sciences, Shanghai University, Shanghai, P. R. China
| |
Collapse
|
44
|
Retrospective Characterization of Initial Peste des petits ruminants Outbreaks (2008-2012) in the Democratic Republic of the Congo. Viruses 2021; 13:v13122373. [PMID: 34960642 PMCID: PMC8708707 DOI: 10.3390/v13122373] [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: 10/01/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute, contagious viral disease of small ruminants, goats and sheep. The Democratic Republic of the Congo (DRC) was a PPR-free country until 2007, although in 2006, scare alerts were received from the east and the southwest of the country, reporting repeated mortalities, specifically in goats. In 2008, PPR outbreaks were seen in several villages in the west, leading to structured veterinary field operations. Blood, swabs and pathological specimens consisting of tissues from lungs, spleens, lymph nodes, kidneys, livers and hearts were ethically collected from clinically infected and/or dead animals, as appropriate, in 35 districts. Epidemiological information relating to major risk factors and socio-economic impact was progressively collected, revealing the deaths of 744,527 goats, which converted to a trade value of USD 35,674,600. Samples from infected and dead animals were routinely analyzed by the Central Veterinary Laboratory at Kinshasa for diagnosis, and after official declaration of PPR outbreaks by the FAO in July 2012, selected tissue samples were sent to The Pirbright Institute, United Kingdom, for genotyping. As a result of surveys undertaken between 2008 and 2012, PPR virus (PPRV)-specific antibodies were detected in 25 locations out of 33 tested (75.7%); PPRV nucleic acid was detected in 25 locations out of 35 (71.4%); and a typical clinical picture of PPR was observed in 23 locations out of 35 (65.7%). Analysis of the partial and full genome sequences of PPR viruses (PPRVs) obtained from lymphoid tissues of dead goats collected in Tshela in the DRC in 2012 confirmed the circulation of lineage IV PPRV, showing the highest homology (99.6-100%) with the viruses circulating in the neighboring countries of Gabon, in the Aboumi outbreak in 2011, and Nigeria (99.3% homology) in 2013, although recent outbreaks in 2016 and 2018 in the western part of the DRC that borders with East Africa demonstrated circulation of lineage II and lineage III PPRV.
Collapse
|
45
|
Choudhury SM, Ma X, Dang W, Li Y, Zheng H. Recent Development of Ruminant Vaccine Against Viral Diseases. Front Vet Sci 2021; 8:697194. [PMID: 34805327 PMCID: PMC8595237 DOI: 10.3389/fvets.2021.697194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023] Open
Abstract
Pathogens of viral origin produce a large variety of infectious diseases in livestock. It is essential to establish the best practices in animal care and an efficient way to stop and prevent infectious diseases that impact animal husbandry. So far, the greatest way to combat the disease is to adopt a vaccine policy. In the fight against infectious diseases, vaccines are very popular. Vaccination's fundamental concept is to utilize particular antigens, either endogenous or exogenous to induce immunity against the antigens or cells. In light of how past emerging and reemerging infectious diseases and pandemics were handled, examining the vaccination methods and technological platforms utilized for the animals may provide some useful insights. New vaccine manufacturing methods have evolved because of developments in technology and medicine and our broad knowledge of immunology, molecular biology, microbiology, and biochemistry, among other basic science disciplines. Genetic engineering, proteomics, and other advanced technologies have aided in implementing novel vaccine theories, resulting in the discovery of new ruminant vaccines and the improvement of existing ones. Subunit vaccines, recombinant vaccines, DNA vaccines, and vectored vaccines are increasingly gaining scientific and public attention as the next generation of vaccines and are being seen as viable replacements to conventional vaccines. The current review looks at the effects and implications of recent ruminant vaccine advances in terms of evolving microbiology, immunology, and molecular biology.
Collapse
Affiliation(s)
- Sk Mohiuddin Choudhury
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - XuSheng Ma
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wen Dang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - YuanYuan Li
- Gansu Agricultural University, Lanzhou, China
| | - HaiXue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
46
|
Li L, Wu J, Cao X, He J, Liu X, Shang Y. Analysis and Sequence Alignment of Peste des Petits Ruminants Virus ChinaSX2020. Vet Sci 2021; 8:vetsci8110285. [PMID: 34822658 PMCID: PMC8623451 DOI: 10.3390/vetsci8110285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
The peste des petits ruminants virus (PPRV) mainly infects goats and sheep and causes a highly contagious disease, PPR. Recently, a PPRV strain named ChinaSX2020 was isolated and confirmed following an indirect immunofluorescence assay and PCR using PPRV-specific antibody and primers, respectively. A sequencing of the ChinaSX2020 strain showed a genome length of 15,954 nucleotides. A phylogenetic tree analysis showed that the ChinaSX2020 genome was classified into lineage IV of the PRRV genotypes. The genome of the ChinaSX2020 strain was found to be closely related to PPRVs isolated in China between 2013 and 2014. These findings revealed that not a variety of PRRVs but similar PPRVs were continuously spreading and causing sporadic outbreaks in China.
Collapse
|
47
|
Eloiflin RJ, Auray G, Python S, Rodrigues V, Seveno M, Urbach S, El Koulali K, Holzmuller P, Totte P, Libeau G, Bataille A, Summerfield A. Identification of Differential Responses of Goat PBMCs to PPRV Virulence Using a Multi-Omics Approach. Front Immunol 2021; 12:745315. [PMID: 34671358 PMCID: PMC8521192 DOI: 10.3389/fimmu.2021.745315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/14/2021] [Indexed: 12/02/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease of small ruminants, mainly sheep and goats. Host susceptibility varies considerably depending on the PPR virus (PPRV) strain, the host species and breed. The effect of strains with different levels of virulence on the modulation of the immune system has not been thoroughly compared in an experimental setting so far. In this study, we used a multi-omics approach to investigate the host cellular factors involved in different infection phenotypes. Peripheral blood mononuclear cells (PBMCs) from Saanen goats were activated with a T-cell mitogen and infected with PPRV strains of different virulence: Morocco 2008 (high virulence), Ivory Coast 1989 (low virulence) and Nigeria 75/1 (live attenuated vaccine strain). Our results showed that the highly virulent strain replicated better than the other two in PBMCs and rapidly induced cell death and a stronger inhibition of lymphocyte proliferation. However, all the strains affected lymphocyte proliferation and induced upregulation of key antiviral genes and proteins, meaning a classical antiviral response is orchestrated regardless of the virulence of the PPRV strain. On the other hand, the highly virulent strain induced stronger inflammatory responses and activated more genes related to lymphocyte migration and recruitment, and inflammatory processes. Both transcriptomic and proteomic approaches were successful in detecting viral and antiviral effectors under all conditions. The present work identified key immunological factors related to PPRV virulence in vitro.
Collapse
Affiliation(s)
- Roger-Junior Eloiflin
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Gaël Auray
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sylvie Python
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Valérie Rodrigues
- ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France.,CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Petit-Bourg, France
| | - Martial Seveno
- BCM (BioCampus Montpellier), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Serge Urbach
- IGF (Institut de Génomique Fonctionnelle), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Khadija El Koulali
- BCM (BioCampus Montpellier), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Philippe Holzmuller
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Philippe Totte
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Genevieve Libeau
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Arnaud Bataille
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Artur Summerfield
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
48
|
Wani SA, Sahu AR, Khan RIN, Praharaj MR, Saxena S, Rajak KK, Muthuchelvan D, Sahoo A, Mishra B, Singh RK, Mishra BP, Gandham RK. Proteome Modulation in Peripheral Blood Mononuclear Cells of Peste des Petits Ruminants Vaccinated Goats and Sheep. Front Vet Sci 2021; 8:670968. [PMID: 34631844 PMCID: PMC8493254 DOI: 10.3389/fvets.2021.670968] [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: 02/22/2021] [Accepted: 05/31/2021] [Indexed: 12/03/2022] Open
Abstract
In the present study, healthy goats and sheep (n = 5) that were confirmed negative for peste des petits ruminants virus (PPRV) antibodies by monoclonal antibody-based competitive ELISA and by serum neutralization test and for PPRV antigen by s-ELISA were vaccinated with Sungri/96. A quantitative study was carried out to compare the proteome of peripheral blood mononuclear cells (PBMCs) of vaccinated goat and sheep [5 days post-vaccination (dpv) and 14 dpv] vs. unvaccinated (0 day) to divulge the alteration in protein expression following vaccination. A total of 232 and 915 proteins were differentially expressed at 5 and 14 dpv, respectively, in goats. Similarly, 167 and 207 proteins were differentially expressed at 5 and 14 dpv, respectively, in sheep. Network generated by Ingenuity Pathway Analysis was “infectious diseases, antimicrobial response, and inflammatory response,” which includes the highest number of focus molecules. The bio functions, cell-mediated immune response, and humoral immune response were highly enriched in goats at 5 dpv and at 14 dpv. At the molecular level, the immune response produced by the PPRV vaccine virus in goats is effectively coordinated and stronger than that in sheep, though the vaccine provides protection from virulent virus challenge in both. The altered expression of certain PBMC proteins especially ISG15 and IRF7 induces marked changes in cellular signaling pathways to coordinate host immune responses.
Collapse
Affiliation(s)
- Sajad Ahmad Wani
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India.,College of Pharmacy, Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, United States
| | - Amit Ranjan Sahu
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Manas Ranjan Praharaj
- Systems Biology Lab, Department of Biotechnology -National Institute of Animal Biotechnology, Hyderabad, India
| | - Shikha Saxena
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Kaushal Kishor Rajak
- Division of Biological Products, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Dhanavelu Muthuchelvan
- Division of Virology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Mukteswar, India
| | - Aditya Sahoo
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Bina Mishra
- Division of Biological Products, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - R K Singh
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Bishnu Prasad Mishra
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Ravi Kumar Gandham
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India.,Systems Biology Lab, Department of Biotechnology -National Institute of Animal Biotechnology, Hyderabad, India
| |
Collapse
|
49
|
Zhang W, Deng H, Liu Y, Chen S, Liu Y, Zhao Y. Antiviral Effectivity of Favipiravir Against Peste Des Petits Ruminants Virus Is Mediated by the JAK/STAT and PI3K/AKT Pathways. Front Vet Sci 2021; 8:722840. [PMID: 34552976 PMCID: PMC8450531 DOI: 10.3389/fvets.2021.722840] [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: 06/09/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022] Open
Abstract
Peste des petits ruminants virus (PPRV), belonging to the genus Morbillivirus in the family Paramyxoviridae, causes severe infectious disease in small ruminants and has been rapidly spreading in many parts of Africa, the Middle East, and Asia. Although vaccination is considered to be an effective means of controlling PPR, the heat-sensitive nature of the vaccines against PPRV greatly limits their application in areas with a hot climate. In the present study, we investigated the anti-PPRV effects of favipiravir and sought to identify the underlying mechanisms in vitro using the Vero cell line. MTT assays, Western blotting, indirect immunofluorescence assays, virus plaque formation assays, and qRT-PCR were used to assess the effects of favipiravir on the life cycle of PPRV and the expression of RNA-dependent RNA polymerase (RdRp). Additionally, the expression levels of JAK1, STAT1, phosphorylated (p)-STAT1, PI3K, AKT, and p-AKT, as well as those of signaling molecules acting downstream of the JAK/STAT and PI3K/AKT signaling pathways, were determined by Western blotting and qRT-PCR. The results indicated that, in PPRV-infected, favipiravir-treated Vero cells, the attachment, invasion, replication, and release of PPRV were significantly inhibited, as was the expression of RdRp, when compared with that in untreated PPRV-infected cells. Furthermore, in favipiravir-treated cells, the expression of JAK1 and STAT1 was downregulated, whereas that of p-STAT1 was significantly upregulated. Similarly, the expression levels of PKR, IRF9, ISG54, and MxA proteins that are associated with innate antiviral activity in host cells were also markedly increased. Moreover, with favipiravir treatment, the expression of PI3K and p-AKT and the p-AKT/AKT ratio were significantly decreased, whereas the expression of AKT was noticeably upregulated. The expression of GSK3, NF-κB p65, p-NF-κB p65, and BAD was also increased with favipiravir treatment, while the expression of CREB, p-CREB, p-GSK3, and Bcl-2 was slightly decreased. In addition, all the p-GSK3/GSK3, p-CREB/CREB, p-NF-κB/NF-κB, and p-BAD/BAD ratios were significantly reduced in favipiravir-treated cells. These results implied that the antiviral effectivity of favipiravir against PPRV is mediated by the JAK/STAT and PI3K/AKT pathways and that favipiravir has potential for use as an effective antiviral agent against PPRV.
Collapse
Affiliation(s)
- Weifeng Zhang
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang, China
| | - Hualong Deng
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang, China
| | - Yanfen Liu
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang, China
| | - Shaohong Chen
- Department of Bioengineering, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - You Liu
- Department of Bioengineering, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Yuntao Zhao
- Department of Bioengineering, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| |
Collapse
|
50
|
Guo Z, Zhang Z, Prajapati M, Li Y. Lymphopenia Caused by Virus Infections and the Mechanisms Beyond. Viruses 2021; 13:v13091876. [PMID: 34578457 PMCID: PMC8473169 DOI: 10.3390/v13091876] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections can give rise to a systemic decrease in the total number of lymphocytes in the blood, referred to as lymphopenia. Lymphopenia may affect the host adaptive immune responses and impact the clinical course of acute viral infections. Detailed knowledge on how viruses induce lymphopenia would provide valuable information into the pathogenesis of viral infections and potential therapeutic targeting. In this review, the current progress of viruses-induced lymphopenia is summarized and the potential mechanisms and factors involved are discussed.
Collapse
Affiliation(s)
- Zijing Guo
- State Key Laboratory on Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China;
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
| | - Zhidong Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
| | - Meera Prajapati
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
- National Animal Health Research Centre, Nepal Agricultural Research Council, Lalitpur 44700, Nepal
| | - Yanmin Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
- Correspondence: ; Tel.: +28-85528276
| |
Collapse
|