1
|
Wei X, Lu K, Chang Z, Guo H, Li Q, Yuan B, Liu C, Yang Z, Liu H. Genetic analyses and functional validation of ruminant SLAMs reveal potential hosts for PPRV. Vet Res 2025; 56:57. [PMID: 40103005 PMCID: PMC11916873 DOI: 10.1186/s13567-025-01489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/05/2024] [Indexed: 03/20/2025] Open
Abstract
Peste des petits ruminants (PPR), caused by the peste des petits ruminants virus (PPRV), is a highly contagious disease affecting ruminants. While goats and sheep are well-known hosts, PPRV has also spread to wild ruminants, and it remains unclear which ruminant species can be infected. SLAM (Signaling lymphocytic activation molecule) acts as the primary receptor for PPRV, playing a crucial role in the viral infection process. Identifying which ruminant SLAMs can mediate PPRV infection is essential for understanding the potential hosts of PPRV, which is vital for effective eradication efforts. In this study, we first extracted 77 ruminant species' SLAM sequences from ruminant genome database. Based on these sequences, we predicted the structures of ruminant SLAMs. The analysis revealed that SLAM conformation is similar across ruminant species, and the potential PPRV H protein binding domain residues were conserved among SLAMs of these 77 species. Phylogenetic analysis of SLAM grouped ruminants into six families. We then selected representative SLAMs from each ruminant family to assess their role in PPRV infection. Our findings demonstrated that ruminant SLAMs efficiently mediated PPRV infection, with enhanced viral amplification observed in cells expressing SLAM from java mouse deer (Tragulidae) and goat (Bovidae), compared to cells expressing SLAM from white tailed deer (Cervidae) and giraffe (Giraffidae). These results underscore the need to consider a broader range of potential host populations beyond goat and sheep in efforts to prevent and eradicate PPRV.
Collapse
Affiliation(s)
- Xi Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhengwu Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hanwei Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Qinfeng Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Binxuan Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chen Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China.
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China.
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| |
Collapse
|
2
|
Zou H, Niu Z, Cheng P, Wu C, Li W, Luo G, Huang S. Structure, Attachment and Transmembrane Internalisation of Peste Des Petits Ruminants Virus. Vet Med Sci 2025; 11:e70182. [PMID: 39739994 DOI: 10.1002/vms3.70182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/19/2024] [Accepted: 12/08/2024] [Indexed: 01/02/2025] Open
Abstract
Peste des petits ruminants virus (PPRV), a single-stranded negative-sense RNA virus with an envelope, belongs to the Morbillivirus in the Paramyxoviridae family and is prevalent worldwide. PPRV infection causes fever, stomatitis, diarrhoea, pneumonia, abortion and other symptoms in small ruminants, with a high mortality rate that poses a significant threat to the sustainability and productivity of the small ruminant livestock sector. The PPRV virus particles have a diameter of approximately 400-500 nm and are composed of six structural proteins: nucleocapsid protein (N), phosphoprotein (P), envelope matrix protein (M), fusion protein (F), haemagglutinin protein (H) and large protein (L). Each protein has a distinct role in the virus's life cycle. Although the life cycle activities of PPRV have been widely reported, they are still limited. Research has demonstrated that PPRV has distinct adhesion factors on various cell surfaces, such as the epithelial cell adhesion factor nectin-4 or the lymphocyte adhesion factor SLAM. After attaching to the cell, the F and H proteins on the PPRV membrane interact with each other, resulting in a conformational change in the F protein. This change allows the F protein to enter the cell through direct fusion with the host cell membrane. The virus enters the host cell via the outer vesicle endocytosis strategy and replicates and proliferates through the role of caveolin, actin, dynein and cholesterol on the host cell membrane. This review summarises the viral structure, attachment mechanism and transmembrane internalisation mechanism of PPRV. The aim of this review is to provide theoretical support for the development of PPRV inhibitors and the prevention and control of PPR.
Collapse
Affiliation(s)
- Hong Zou
- Chongqing Three Gouges Vocational College, College of Animal Science & Technology, Wanzhou, China
| | - Zheng Niu
- College of Veterinary Medicine, Northwest A & F University, Yangling, China
| | - Peng Cheng
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Chunxia Wu
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Wenjie Li
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Gan Luo
- Wanzhou Center for Animal Husbandry Industry Development of Chongqing, Wanzhou, China
| | - Shilei Huang
- Chongqing Three Gouges Vocational College, College of Animal Science & Technology, Wanzhou, China
| |
Collapse
|
3
|
Gaur SK, Chaudhary Y, Jain J, Singh R, Kaul R. Structural and functional characterization of peste des petits ruminants virus coded hemagglutinin protein using various in-silico approaches. Front Microbiol 2024; 15:1427606. [PMID: 38966393 PMCID: PMC11222573 DOI: 10.3389/fmicb.2024.1427606] [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: 05/04/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024] Open
Abstract
Peste des petits ruminants (PPR), a disease of socioeconomic importance has been a serious threat to small ruminants. The causative agent of this disease is PPR virus (PPRV) which belongs to the genus Morbillivirus. Hemagglutinin (H) is a PPRV coded transmembrane protein embedded in the viral envelope and plays a vital role in mediating the entry of virion particle into the cell. The infected host mounts an effective humoral response against H protein which is important for host to overcome the infection. In the present study, we have investigated structural, physiological and functional properties of hemagglutinin protein using various computational tools. The sequence analysis and structure prediction analysis show that hemagglutinin protein comprises of beta sheets as the predominant secondary structure, and may lack neuraminidase activity. PPRV-H consists of several important domains and motifs that form an essential scaffold which impart various critical roles to the protein. Comparative modeling predicted the protein to exist as a homo-tetramer that binds to its cognate cellular receptors. Certain amino acid substitutions identified by multiple sequence alignment were found to alter the predicted structure of the protein. PPRV-H through its predicted interaction with TLR-2 molecule may drive the expression of CD150 which could further propagate the virus into the host. Together, our study provides new insights into PPRV-H protein structure and its predicted functions.
Collapse
Affiliation(s)
| | | | | | | | - Rajeev Kaul
- Department of Microbiology, University of Delhi South Campus, New Delhi, India
| |
Collapse
|
4
|
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
|
5
|
Tully M, Batten C, Ashby M, Mahapatra M, Parekh K, Parida S, Njeumi F, Willett B, Bataille A, Libeau G, Kwiatek O, Caron A, Berguido FJ, Lamien CE, Cattoli G, Misinzo G, Keyyu J, Mdetele D, Gakuya F, Bodjo SC, Taha FA, Elbashier HM, Khalafalla AI, Osman AY, Kock R. The evaluation of five serological assays in determining seroconversion to peste des petits ruminants virus in typical and atypical hosts. Sci Rep 2023; 13:14787. [PMID: 37684280 PMCID: PMC10491793 DOI: 10.1038/s41598-023-41630-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Peste des petits ruminants (PPR) is an infectious viral disease, primarily of small ruminants such as sheep and goats, but is also known to infect a wide range of wild and domestic Artiodactyls including African buffalo, gazelle, saiga and camels. The livestock-wildlife interface, where free-ranging animals can interact with captive flocks, is the subject of scrutiny as its role in the maintenance and spread of PPR virus (PPRV) is poorly understood. As seroconversion to PPRV indicates previous infection and/or vaccination, the availability of validated serological tools for use in both typical (sheep and goat) and atypical species is essential to support future disease surveillance and control strategies. The virus neutralisation test (VNT) and enzyme-linked immunosorbent assay (ELISA) have been validated using sera from typical host species. Still, the performance of these assays in detecting antibodies from atypical species remains unclear. We examined a large panel of sera (n = 793) from a range of species from multiple countries (sourced 2015-2022) using three tests: VNT, ID VET N-ELISA and AU-PANVAC H-ELISA. A sub-panel (n = 30) was also distributed to two laboratories and tested using the luciferase immunoprecipitation system (LIPS) and a pseudotyped virus neutralisation assay (PVNA). We demonstrate a 75.0-88.0% agreement of positive results for detecting PPRV antibodies in sera from typical species between the VNT and commercial ELISAs, however this decreased to 44.4-62.3% in sera from atypical species, with an inter-species variation. The LIPS and PVNA strongly correlate with the VNT and ELISAs for typical species but vary when testing sera from atypical species.
Collapse
Affiliation(s)
| | | | - Martin Ashby
- The Pirbright Institute, Pirbright, United Kingdom
| | | | | | - Satya Parida
- The Pirbright Institute, Pirbright, United Kingdom
- Food and Agriculture Organization (FAO), United Nations, Rome, Italy
| | - Felix Njeumi
- Food and Agriculture Organization (FAO), United Nations, Rome, Italy
| | - Brian Willett
- MRC-University of Glasgow Centre for Virus Research (UoG), Glasgow, United Kingdom
| | - Arnaud Bataille
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Genevieve Libeau
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Olivier Kwiatek
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Alexandre Caron
- ASTRE, University of Montpellier, CIRAD, INRA, MUSE, Montpellier, France
| | - Francisco J Berguido
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, 2444, Seibersdorf, Austria
| | - Charles E Lamien
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, 2444, Seibersdorf, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, 2444, Seibersdorf, Austria
| | - Gerald Misinzo
- SACIDS Foundation for One Health, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Julius Keyyu
- Tanzania Wildlife Research Institute (TAWIRI), Arusha, Tanzania
| | | | - Francis Gakuya
- Wildlife Research & Training Institute (WRTI), Karagita, Kenya
| | - Sanne Charles Bodjo
- Pan African Veterinary Vaccine Centre for African Union (AU-PANVAC), Debre Zeit, Ethiopia
| | | | | | - Abdelmalik Ibrahim Khalafalla
- Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi, United Arab Emirates
- Faculty of Veterinary Medicine, University of Khartoum, Khartoum, Sudan
| | - Abdinasir Y Osman
- National Institute of Health (NIH), Ministry of Health, Mogadishu, Somalia
- Royal Veterinary College (RVC), London, United Kingdom
| | - Richard Kock
- Royal Veterinary College (RVC), London, United Kingdom
| |
Collapse
|
6
|
Li L, Li S, Han S, Li P, Du G, Wu J, Cao X, Shang Y. Inhibition of caspase-1-dependent apoptosis suppresses peste des petits ruminants virus replication. J Vet Sci 2023; 24:e55. [PMID: 37638708 PMCID: PMC10556287 DOI: 10.4142/jvs.22288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/10/2023] [Accepted: 05/26/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Peste des petits ruminants (PPR), caused by the PPR virus (PPRV), is an acute and fatal contagious disease that mainly infects goats, sheep, and other artiodactyls. Peripheral blood mononuclear cells (PBMCs) are considered the primary innate immune cells. OBJECTIVES PBMCs derived from goats were infected with PPRV and analyzed to detect the relationship between PPRV replication and apoptosis or the inflammatory response. METHODS Quantitative real-time polymerase chain reaction was used to identify PPRV replication and cytokines expression. Flow cytometry was conducted to detect apoptosis and the differentiation of CD4+ and CD8+ T cells after PPRV infection. RESULTS PPRV stimulated the differentiation of CD4+ and CD8+ T cells. In addition, PPRV induced apoptosis in goat PBMCs. Furthermore, apoptosis and the inflammatory response induced by PPRV could be suppressed by Z-VAD-FMK and Z-YVAD-FMK, respectively. Moreover, the virus titer of PPRV was attenuated by inhibiting caspase-1-dependent apoptosis and inflammation. CONCLUSIONS This study showed that apoptosis and the inflammatory response play an essential role in PPR viral replication in vitro, providing a new mechanism related to the cell host response.
Collapse
Affiliation(s)
- Lingxia Li
- College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China
- College of Animal Science and Veterinary Science, Qinghai University; Qinghai Key Laboratory of Pathogen Diagnosis and Green Prevention and Control Technology for Animal Diseases, Xining 810016, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Shengqing Li
- College of Animal Science and Veterinary Science, Qinghai University; Qinghai Key Laboratory of Pathogen Diagnosis and Green Prevention and Control Technology for Animal Diseases, Xining 810016, China
| | - Shengyi Han
- College of Animal Science and Veterinary Science, Qinghai University; Qinghai Key Laboratory of Pathogen Diagnosis and Green Prevention and Control Technology for Animal Diseases, Xining 810016, China
| | - Pengfei Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Guoyu Du
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Jinyan Wu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Xiaoan Cao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Youjun Shang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China.
| |
Collapse
|
7
|
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
|
8
|
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: 5] [Impact Index Per Article: 1.7] [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
|
9
|
Multiple Receptors Involved in Invasion and Neuropathogenicity of Canine Distemper Virus: A Review. Viruses 2022; 14:v14071520. [PMID: 35891500 PMCID: PMC9317347 DOI: 10.3390/v14071520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 12/04/2022] Open
Abstract
The canine distemper virus (CDV) is a morbillivirus that infects a broad range of terrestrial carnivores, predominantly canines, and is associated with high mortality. Similar to another morbillivirus, measles virus, which infects humans and nonhuman primates, CDV transmission from an infected host to a naïve host depends on two cellular receptors, namely, the signaling lymphocyte activation molecule (SLAM or CD150) and the adherens junction protein nectin-4 (also known as PVRL4). CDV can also invade the central nervous system by anterograde spread through olfactory nerves or in infected lymphocytes through the circulation, thus causing chronic progressive or relapsing demyelination of the brain. However, the absence of the two receptors in the white matter, primary cultured astrocytes, and neurons in the brain was recently demonstrated. Furthermore, a SLAM/nectin-4-blind recombinant CDV exhibits full cell-to-cell transmission in primary astrocytes. This strongly suggests the existence of a third CDV receptor expressed in neural cells, possibly glial cells. In this review, we summarize the recent progress in the study of CDV receptors, highlighting the unidentified glial receptor and its contribution to pathogenicity in the host nervous system. The reviewed studies focus on CDV neuropathogenesis, and neural receptors may provide promising directions for the treatment of neurological diseases caused by CDV. We also present an overview of other neurotropic viruses to promote further research and identification of CDV neural receptors.
Collapse
|
10
|
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: 0.7] [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
|
11
|
Oral S2-Ag85 DNA Vaccine Activated Intestinal Cell dsDNA and RNA Sensors to Promote the Presentation of Intestinal Antigen. J Immunol Res 2022; 2022:7200379. [PMID: 35465352 PMCID: PMC9020918 DOI: 10.1155/2022/7200379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
Objective To explore the molecular mechanism by which oral S2-Ag85DNA vaccines present intestinal antigens. The oral S2-Ag85 vaccine has been shown to protect the human body and effectively improve the titration of the vaccine by acting on intestinal mucosa cells and enhancing their immunogenicity. Method Mice were immunized with the recombinant S2-Ag85 vaccine, and antibody secretion was then detected in the intestinal tissue. The molecular mechanisms of in vitro detection sensor molecules RIG-1, Pol III, and related conductor transductor molecules DAI, STING, AIM2, IRF3, and IRF7 were determined by separating intestinal IEC, DC, and IELC cells. Results The S2-Ag85A vaccine was effective in activating dsDNA and RNA transduction pathways in intestinal cells and improving intestinal antigen presentation in mice.
Collapse
|
12
|
Nooruzzaman M, Akter MN, Begum JA, Begum S, Parvin R, Giasuddin M, Islam MR, Lamien CE, Cattoli G, Dundon WG, Chowdhury EH. Molecular insights into peste des petits ruminants virus identified in Bangladesh between 2008 and 2020. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 96:105163. [PMID: 34848354 DOI: 10.1016/j.meegid.2021.105163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 06/13/2023]
Abstract
An in-depth knowledge of the molecular evolution of the peste des petits ruminants virus (PPRV) is critical for the success of the current global eradication program. For this reason, a molecular evolutionary analysis of PPRVs circulating in Bangladesh over a decade (2008-2020) was performed. The complete genome sequencing of three PPRV isolates from 2008 (BD2), 2015 (BD12) and 2017 (BD17) as well as full length nucleocapsid (N), matrix (M) and fusion (F) gene sequencing of seven more samples from 2015 to 2020 was performed. Phylogenetic analysis classified all ten PPRVs from Bangladesh as members of lineage IV and showed that they were closely related to PPRV strains detected in China and Tibet during 2007-2008, and India during 2014-2018. Time scale Bayesian Maximum Clade Credibility (MCC) phylogenetic analysis of the three complete genomes revealed a mean Time to Most Recent Common Ancestor (TMRCA) of 2000. Comparative deduced amino acid residue analysis at various functional motifs of PPRVs related to virus structure and function, virulence and host adaptation, receptor binding sites and polymerase activity revealed conserved residues among the PPRVs from Bangladesh. In total sixteen epitopes were predicted from four immunogenic proteins i.e. N, M, F and haemagglutinin (H). Interestingly, the predicted epitopes from the N and M proteins shared conserved epitopes with two vaccine strains currently being used, indicating that the strains from Bangladesh could be potentially used as alternative local vaccines.
Collapse
Affiliation(s)
- Mohammed Nooruzzaman
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Mst Nazia Akter
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Jahan Ara Begum
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Shahana Begum
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; Department of Physiology, Faculty of Veterinary, Animal & Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Rokshana Parvin
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Md Giasuddin
- Animal Health Division, Bangladesh Livestock Research Institute, Savar, Dhaka, Bangladesh
| | - Mohammad Rafiqul Islam
- Department of Pathology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Charles E Lamien
- Animal Production and Health Laboratory, Joint FAO/IAEA Division, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO/IAEA Division, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - William G Dundon
- Animal Production and Health Laboratory, Joint FAO/IAEA Division, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | | |
Collapse
|
13
|
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
|
14
|
Eloiflin RJ, Auray G, Python S, Rodrigues V, Seveno M, Urbach S, El Koulali K, Holzmuller P, Totte P, Libeau G, Bataille A, Summerfield A. Identification of Differential Responses of Goat PBMCs to PPRV Virulence Using a Multi-Omics Approach. Front Immunol 2021; 12:745315. [PMID: 34671358 PMCID: PMC8521192 DOI: 10.3389/fimmu.2021.745315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/14/2021] [Indexed: 12/02/2022] Open
Abstract
Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease of small ruminants, mainly sheep and goats. Host susceptibility varies considerably depending on the PPR virus (PPRV) strain, the host species and breed. The effect of strains with different levels of virulence on the modulation of the immune system has not been thoroughly compared in an experimental setting so far. In this study, we used a multi-omics approach to investigate the host cellular factors involved in different infection phenotypes. Peripheral blood mononuclear cells (PBMCs) from Saanen goats were activated with a T-cell mitogen and infected with PPRV strains of different virulence: Morocco 2008 (high virulence), Ivory Coast 1989 (low virulence) and Nigeria 75/1 (live attenuated vaccine strain). Our results showed that the highly virulent strain replicated better than the other two in PBMCs and rapidly induced cell death and a stronger inhibition of lymphocyte proliferation. However, all the strains affected lymphocyte proliferation and induced upregulation of key antiviral genes and proteins, meaning a classical antiviral response is orchestrated regardless of the virulence of the PPRV strain. On the other hand, the highly virulent strain induced stronger inflammatory responses and activated more genes related to lymphocyte migration and recruitment, and inflammatory processes. Both transcriptomic and proteomic approaches were successful in detecting viral and antiviral effectors under all conditions. The present work identified key immunological factors related to PPRV virulence in vitro.
Collapse
Affiliation(s)
- Roger-Junior Eloiflin
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Gaël Auray
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sylvie Python
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Valérie Rodrigues
- ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France.,CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Petit-Bourg, France
| | - Martial Seveno
- BCM (BioCampus Montpellier), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Serge Urbach
- IGF (Institut de Génomique Fonctionnelle), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Khadija El Koulali
- BCM (BioCampus Montpellier), Univ. Montpellier, CNRS (Centre national de la recherche scientifique), INSERM, Montpellier, France
| | - Philippe Holzmuller
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Philippe Totte
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Genevieve Libeau
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Arnaud Bataille
- CIRAD (Agricultural Research Centre for International Development), UMR (Unité Mixte de Recherche), ASTRE (Animal, Health, Territories, Risks and Ecosystems), Montpellier, France.,ASTRE (Animal, Health, Territories, Risks and Ecosystems), University of Montpellier, CIRAD (Agricultural Research Centre for International Development), INRAE (Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement), Montpellier, France
| | - Artur Summerfield
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
15
|
Gautam S, Joshi C, Sharma AK, Singh KP, Gurav A, Sankar M, Ramakrishnan MA, Chaudhary D, Chauhan RS, Dhama K, Dhanavelu M. Virus distribution and early pathogenesis of highly pathogenic peste-des-petits-ruminants virus in experimentally infected goats. Microb Pathog 2021; 161:105232. [PMID: 34627939 DOI: 10.1016/j.micpath.2021.105232] [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: 05/30/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Despite causing one of the most dreaded diseases of small ruminants, relatively little is known about the pathogenic events, antigen distribution and the cells responsible for the uptake and transmission of peste-des-petits-ruminants virus (PPRV) during primitive stages of infection. OBJECTIVES We aimed at deciphering the sequential tissue tropism, pathological events and putative role of M2c macrophages during incubatory, prodromal and invasive stages of PPRV infection. METHODOLOGY A total of 10 goats were sequentially sacrificed at 1, 2, 3, 4, and 5 days post-infection (dpi, n = 2 per time-point) following intranasal inoculation with a highly virulent strain of PPRV (lineage IV PPRV/Izatnagar/94). Histological evaluation to assess PPRV mediated pathologies, RT-qPCR and immunohistochemistry (IHC) to decipher sequential virus distribution, and dual immunolabelling to determine the role of M2c macrophage in early PPRV uptake and transmission was performed. RESULTS PPRV/Izatnagar/94 caused major pathologies in the lung tissues. Unprecedentedly, PPRV nucleic acid and antigens were detected in various tissues as early as one dpi. RT-qPCR revealed PPRV in the nasal cavity, trachea, bronchi, tongue and lymph nodes draining these tissues from 1 dpi. IHC affirms cells residing in the lamina propria and submucosa of the respiratory tract and tongue and peribronchiolar areas of lungs as the primary target of PPRV. Following initial replication in the respiratory tract, PPRV is transmitted to the regional lymph nodes where primary viral amplification occurs. After viraemia and secondary replication in generalized lymphoid tissues, PPRV infects and replicates in the epithelial cells. Further, we localized CD163+ M2c macrophages in the goat tissues, but dual IHC elucidated that M2c macrophages do not facilitate uptake and transmission of PPRV during the early stages of infection. CONCLUSION Our study substantiates the disease establishment process and pathogenesis of PPRV/Izatnagar/94 during the incubatory and prodromal stages of infection. Further, we have also observed M2c macrophage distribution in the goat tissues and demonstrated that they do not pick and transmit PPRV.
Collapse
Affiliation(s)
- Siddharth Gautam
- ICAR - Indian Veterinary Research Institute, Mukteshwar, Nainital, U.K., 263138, India.
| | - Chitra Joshi
- Department of Animal Husbandry, Almora, U.K., 263601, India
| | - Anil K Sharma
- ICAR - Indian Veterinary Research Institute, Izatnagar, U.P., 243122, India
| | - Karam P Singh
- ICAR - Indian Veterinary Research Institute, Izatnagar, U.P., 243122, India
| | - Amol Gurav
- ICAR - Indian Veterinary Research Institute, Mukteshwar, Nainital, U.K., 263138, India
| | - Muthu Sankar
- ICAR - Indian Veterinary Research Institute, Mukteshwar, Nainital, U.K., 263138, India
| | | | - Dheeraj Chaudhary
- ICAR - Indian Veterinary Research Institute, Mukteshwar, Nainital, U.K., 263138, India
| | - Ramswaroop S Chauhan
- College of Veterinary and Animal Sciences, GBPUAT, U.S. Nagar, U.K., 263145, India
| | - Kuldeep Dhama
- ICAR - Indian Veterinary Research Institute, Izatnagar, U.P., 243122, India
| | | |
Collapse
|
16
|
Zinc finger antiviral protein (ZAP) inhibits small ruminant morbillivirus replication in vitro. Vet Microbiol 2021; 260:109163. [PMID: 34311269 DOI: 10.1016/j.vetmic.2021.109163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/18/2021] [Indexed: 11/23/2022]
Abstract
Small ruminant morbillivirus (SRMV) is a highly contagious and economically important viral disease of small domestic and wild ruminants. Difficulty with its stable proliferation in ovis aries-derived cells has led to a relative lag in the study of its natural immunity and pathogenesis. Here we report the antiviral properties of ZAP against SRMV, a single-stranded negative-stranded RNA virus of the genus Morbillivirus. ZAP expression was significantly induced in sheep endometrial epithelial cells following SRMV infection. ZAP inhibited SRMV replication in cells after infection, while its overexpression in Vero-SLAM cells significantly increased their resistance to SRMV replication. The ZAP protein co-localized with SRMV RNA in the cytoplasm and ZAP-responsive elements were mapped to the 5' untranslated region of SRMV nucleocapsid, phosphoprotein, matrix, and fusion. In summary, ZAP confers resistance to SRMV infection by directly targeting viral RNA and inhibiting viral replication. Our findings further extend the ranges of viral targets of ZAP and help elucidate the mechanism of SRMV replication.
Collapse
|
17
|
Begum S, Nooruzzaman M, Islam MR, Chowdhury EH. A Sequential Study on the Pathology of Peste Des Petits Ruminants and Tissue Distribution of the Virus Following Experimental Infection of Black Bengal Goats. Front Vet Sci 2021; 8:635671. [PMID: 33681333 PMCID: PMC7933573 DOI: 10.3389/fvets.2021.635671] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
We studied the sequential pathology of peste des petits ruminants (PPR) in Black Bengal goats and analyzed virus distribution in tissues and virus shedding following experimental infection with a Bangladeshi isolate of lineage IV PPR virus (PPRV). The early clinical signs like fever, depression, and ocular and nasal discharges first appeared at 4-7 days post-infection (dpi). Three out of eight inoculated goats died at 13, 15, and 18 dpi, and the rest were killed at different time points from 5 to 18 dpi. Initially, the virus multiplied mostly in the lymphoid organs of the pharyngeal region and caused extensive lymphoid destruction and hemorrhages. This was followed by viremia, massive virus replication in the lungs, and pneumonia along with the appearance of the clinical signs. Subsequently, the virus spread to other organs causing necrotic and hemorrhagic lesions, as well as the virus localized in the upper respiratory, oral and intestinal mucosa resulting in catarrhal, erosive, and ulcerative lesions. On hematological and biochemical investigation progressive leukopenia and hypoproteinemia, a gradual increase of serum metabolites and enzymes associated with liver and kidney damage, and electrolyte imbalance were observed. Seroconversion started at 7 dpi and all the surviving animals had serum antibodies at 14 dpi. Virus shedding was observed in nasal and ocular secretions at 4 dpi and in feces and urine at 14 dpi, which gradually increased and continued till the end of the experiment (18 dpi) despite seroconversion. Therefore, the virus shedding of naturally infected seroconverted goats should be monitored for effective control strategies.
Collapse
Affiliation(s)
- Shahana Begum
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mohammed Nooruzzaman
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mohammad Rafiqul Islam
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Emdadul Haque Chowdhury
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| |
Collapse
|
18
|
Zhang W, Jia K, Jia P, Xiang Y, Lu X, Liu W, Yi M. Marine medaka heat shock protein 90ab1 is a receptor for red-spotted grouper nervous necrosis virus and promotes virus internalization through clathrin-mediated endocytosis. PLoS Pathog 2020; 16:e1008668. [PMID: 32639977 PMCID: PMC7371229 DOI: 10.1371/journal.ppat.1008668] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/20/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022] Open
Abstract
Nervous necrosis virus (NNV) can infect many species of fish and causes serious acute or persistent infection. However, its pathogenic mechanism is still far from clear. Specific cellular surface receptors are crucial determinants of the species tropism of a virus and its pathogenesis. Here, the heat shock protein 90ab1 of marine model fish species marine medaka (MmHSP90ab1) was identified as a novel receptor of red-spotted grouper NNV (RGNNV). MmHSP90ab1 interacted directly with RGNNV capsid protein (CP). Specifically, MmHSP90ab1 bound to the linker region (LR) of CP through its NM domain. Inhibition of MmHSP90ab1 by HSP90-specific inhibitors or MmHSP90ab1 siRNA caused significant inhibition of viral binding and entry, whereas its overexpression led to the opposite effect. The binding of RGNNV to cultured marine medaka hMMES1 cells was inhibited by blocking cell surface-localized MmHSP90ab1 with anti-HSP90β antibodies or pretreating virus with recombinant MmHSP90ab1 or MmHSP90ab1-NM protein, indicating MmHSP90ab1 was an attachment receptor for RGNNV. Furthermore, we found that MmHSP90ab1 formed a complex with CP and marine medaka heat shock cognate 70, a known NNV receptor. Exogenous expression of MmHSP90ab1 independently facilitated the internalization of RGNNV into RGNNV impenetrable cells (HEK293T), which was blocked by chlorpromazine, an inhibitor of clathrin-dependent endocytosis. Further study revealed that MmHSP90ab1 interacted with the marine medaka clathrin heavy chain. Collectively, these data suggest that MmHSP90ab1 is a functional part of the RGNNV receptor complex and involved in the internalization of RGNNV via the clathrin endocytosis pathway.
Collapse
Affiliation(s)
- Wanwan Zhang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Kuntong Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
- * E-mail: (KJ); (MY)
| | - Peng Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Yangxi Xiang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Xiaobing Lu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Wei Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Meisheng Yi
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
- * E-mail: (KJ); (MY)
| |
Collapse
|
19
|
Dou Y, Liang Z, Prajapati M, Zhang R, Li Y, Zhang Z. Expanding Diversity of Susceptible Hosts in Peste Des Petits Ruminants Virus Infection and Its Potential Mechanism Beyond. Front Vet Sci 2020; 7:66. [PMID: 32181263 PMCID: PMC7059747 DOI: 10.3389/fvets.2020.00066] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/27/2020] [Indexed: 01/12/2023] Open
Abstract
Peste des petits ruminants (PPR) is a severe respiratory and digestive tract disease of domestic small ruminants caused by PPR virus (PPRV) of the genus Morbillivirus. Although the primary hosts of PPRV are goats and sheep, the host range of PPRV has been continuously expanding and reported to infect various animal hosts over the last decades, which could bring a potential challenge to effectively control and eradicate PPR globally. In this review, we focused on current knowledge about host expansion and interspecies infection of PPRV and discussed the potential mechanisms involved.
Collapse
Affiliation(s)
- Yongxi Dou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China.,CAAS-ILRI Joint Laboratory for Ruminant Disease Control, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Zhongxiang Liang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Meera Prajapati
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China.,CAAS-ILRI Joint Laboratory for Ruminant Disease Control, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China.,Animal Health Research Division, Nepal Agricultural Research Council, Lalitpur, Nepal
| | - Rui Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Yanmin Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Zhidong Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China.,CAAS-ILRI Joint Laboratory for Ruminant Disease Control, Lanzhou Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Lanzhou, China.,College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| |
Collapse
|
20
|
Li L, Wu J, Liu D, Du G, Liu Y, Shang Y, Liu X. Transcriptional Profiles of Murine Bone Marrow-Derived Dendritic Cells in Response to Peste des Petits Ruminants Virus. Vet Sci 2019; 6:vetsci6040095. [PMID: 31795377 PMCID: PMC6958494 DOI: 10.3390/vetsci6040095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 11/16/2022] Open
Abstract
Background: Peste des petits ruminants virus (PPRV) is the causative agent of PPR, which can cause an acute, highly contagious and fatal disease of sheep and goats, resulting in significant economic losses for commercial animal husbandry due to its high mortality and morbidity. As professional antigen-presenting cells, dendritic cells (DCs) play a unique role in innate immunity. This study aimed to gain a deeper understanding of the transcriptional response of bone marrow-derived dendritic cells (BMDCs) stimulated with PPRV. Results: Transcriptional profiling was performed using RNA sequencing. Herein, we reported that compared to untreatedBMDCs, 4492 differentially expressed genes (DEGs) were identified following PPRV stimulation, out of these DEGs 2311 were upregulated and 2181 were downregulated, respectively. A total of three gene ontology (GO) term clusters of biological process, cell component and molecular function were significantly enriched in 963 GO terms in the PPRV-stimulated BMDCs. These GO clusters were related to inflammatory response, cell division and vacuole, anchoring junction, positive regulation of cellular component and nucleoside binding. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of DEGs were enriched in a chemokine signaling pathway, protein processing in endoplasmic reticulum, cell cycle and mTOR signaling pathway. Additionally, identified DEGs of BMDCs were further validated by qRT-PCR and the results were in accordance with the change of the genes. This study suggested the effects of PPRV stimulation on the maturation and function of BMDCs. Conclusion: We found that the dramatic BMDCs transcriptome changes triggered were predominantly related to an inflammatory response and chemokine signaling pathway.
Collapse
|