1
|
Song L, Shan H, Huang J. Development of HEK293T-produced recombinant receptor-Fc proteins as potential candidates against canine distemper virus. Front Vet Sci 2023; 10:1180673. [PMID: 37215466 PMCID: PMC10196245 DOI: 10.3389/fvets.2023.1180673] [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: 03/06/2023] [Accepted: 04/12/2023] [Indexed: 05/24/2023] Open
Abstract
Canine distemper (CD) is a highly contagious viral disease worldwide. Although live attenuated vaccine is available as a preventive measure against the disease, cases of vaccination failure highlight the importance of potential alternative agent against canine distemper virus (CDV). CDV infects cells mainly by binding signaling lymphocyte activation molecule (SLAM) and Nectin-4 receptor. Here, to develop a new and safe antiviral biological agent for CD, we constructed and expressed CDV receptor proteins fused with Fc region of canine IgG-B, namely, SLAM-Fc, Nectin-Fc and SLAM-Nectin-Fc in HEK293T cells, and antiviral activity of these receptor-Fc proteins was subsequently evaluated. The results showed that the receptor-Fc proteins efficiently bound to receptor binding domain (RBD) of CDV-H, meanwhile, these receptor-Fc proteins competitively inhibited the binding of His-tagged receptor proteins (SLAM-His or Nectin-His) to CDV-H-RBD-Flag protein. Importantly, receptor-Fc proteins exhibited potent anti-CDV activity in vitro. Treatment with receptor-Fc proteins at the pre-entry stage dramatically suppressed CDV infectivity in Vero cells stably expressing canine SLAM. The minimum effective concentration (MEC) of SLAM-Fc, Nectin-Fc and SLAM-Nectin-Fc was 0.2 μg/mL, 0.2 μg/mL, 0.02 μg/mL. The 50% inhibition concentration (IC50) of three proteins was 0.58 μg/mL, 0.32 μg/mL and 0.18 μg/mL, respectively. Moreover, treatment with receptor-Fc proteins post viral infection can also inhibit CDV reproduction, the MEC of SLAM-Fc, Nectin-Fc and SLAM-Nectin-Fc was same as pre-treatment, and the IC50 of receptor-Fc proteins was 1.10 μg/mL, 0.99 μg/mL and 0.32 μg/mL, respectively. The results suggested that the receptor-Fc proteins were more effective for pre-entry treatment than post-infection treatment, furthermore, SLAM-Nectin-Fc was more effective than SLAM-Fc and Nectin-Fc. These findings revealed the receptor-Fc proteins were promising candidates as inhibitor against CDV.
Collapse
Affiliation(s)
- Lingling Song
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, Qingdao, China
- Qingdao Research Center for Veterinary Biological Engineering and Technology, Qingdao, China
| | - Hu Shan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, Qingdao, China
- Qingdao Research Center for Veterinary Biological Engineering and Technology, Qingdao, China
| | - Juan Huang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, Qingdao, China
- Qingdao Research Center for Veterinary Biological Engineering and Technology, Qingdao, China
| |
Collapse
|
2
|
Seki F, Takeda M. Novel and classical morbilliviruses: Current knowledge of three divergent morbillivirus groups. Microbiol Immunol 2022; 66:552-563. [PMID: 36151905 DOI: 10.1111/1348-0421.13030] [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: 06/01/2022] [Revised: 08/30/2022] [Accepted: 09/23/2022] [Indexed: 12/24/2022]
Abstract
Currently, seven species of morbillivirus have been classified. Six of these species (Measles morbillivirus, Rinderpest morbillivirus, Small ruminant morbillivirus, Canine morbillivirus, Phocine morbillivirus, and Cetacean morbillivirus) are highly infectious and cause serious systemic diseases in humans, livestock, domestic dogs, and wild animals. These species commonly use the host proteins signaling lymphocytic activation molecule (SLAM) and nectin-4 as receptors, and this usage contributes to their virulence. The seventh species (Feline morbillivirus: FeMV) is phylogenetically divergent from the six SLAM-using species. FeMV differs from the SLAM-using morbillivirus group in pathogenicity and infectivity, and is speculated to use non-SLAM receptors. Recently, novel species of morbilliviruses have been discovered in bats, rodents, and domestic pigs. Because the ability to use SLAM and nectin-4 is closely related to the infectivity and pathogenicity of morbilliviruses, investigation of the potential usage of these receptors is useful for estimating infectivity and pathogenicity. The SLAM-binding sites in the receptor-binding protein show high similarity among the SLAM-using morbilliviruses. This feature may help to estimate whether novel morbillivirus species can use SLAM as a receptor. A novel morbillivirus species isolated from wild mice diverged from the classified morbilliviruses in the phylogenetic tree, forming a third group separate from the SLAM-using morbillivirus group and FeMV. This suggests that the novel rodent morbillivirus may exhibit a different risk from the SLAM-using morbillivirus group, and analyses of its viral pathogenicity and infectivity toward humans are warranted.
Collapse
Affiliation(s)
- Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| |
Collapse
|
3
|
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: 2.0] [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
|
4
|
Garcia PAT, Cartoceti A, Affolter VK, Jackson K, Keel MK, Agnew D, Cooley T, Melotti J, Fitzgerald SD, Pesavento PA. Distribution of canine distemper virus and nectin-4 in raccoon ( Procyon lotor) skin. Vet Pathol 2022; 59:782-786. [PMID: 35689359 DOI: 10.1177/03009858221102598] [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: 11/16/2022]
Abstract
Raccoons (Procyon lotor) are abundant in urban/wildland interfaces and are key sources of canine distemper virus (CDV) outbreaks in domestic, zoo, and free-ranging wildlife species. CDV is pantropic, which provides multiple potential routes of transmission (urine, respiratory secretions, feces), but the specific role of skin as a target of infection, as a diagnostic sample, or as a potential source of environmental persistence and transmission is unknown. We have characterized the distribution of CDV and its known receptor, nectin-4, in skin samples of 36 raccoons. Even with skin samples that were grossly and histologically normal, immunohistochemistry of skin was useful in the diagnosis of CDV infection, which was found in both epithelium and endothelium. Nectin-4 was codistributed with cellular targets of viral infection. Skin secretions, shed keratinocytes, and hair of CDV infected raccoons are all potential environmental fomites.
Collapse
|
5
|
Abstract
Canine distemper virus (CDV) is a highly contagious pathogen and is known to enter the host via the respiratory tract and disseminate to various organs. Current hypotheses speculate that CDV uses the homologous cellular receptors of measles virus (MeV), SLAM and nectin-4, to initiate the infection process. For validation, here, we established the well-differentiated air-liquid interface (ALI) culture model from primary canine tracheal airway epithelial cells. By applying the green fluorescent protein (GFP)-expressing CDV vaccine strain and recombinant wild-type viruses, we show that cell-free virus infects the airway epithelium mainly via the paracellular route and only after prior disruption of tight junctions by pretreatment with EGTA; this infection was related to nectin-4 but not to SLAM. Remarkably, when CDV-preinfected DH82 cells were cocultured on the basolateral side of canine ALI cultures grown on filter supports with a 1.0-μm pore size, cell-associated CDV could be transmitted via cell-to-cell contact from immunocytes to airway epithelial cultures. Finally, we observed that canine ALI cultures formed syncytia and started to release cell-free infectious viral particles from the apical surface following treatment with an inhibitor of the JAK/STAT signaling pathway (ruxolitinib). Our findings show that CDV can overcome the epithelial barrier through different strategies, including infection via immunocyte-mediated transmission and direct infection via the paracellular route when tight junctions are disrupted. Our established model can be adapted to other animals for studying the transmission routes and the pathogenicity of other morbilliviruses. IMPORTANCE Canine distemper virus (CDV) is not only an important pathogen of carnivores, but it also serves as a model virus for analyzing measles virus pathogenesis. To get a better picture of the different stages of infection, we used air-liquid interface cultures to analyze the infection of well-differentiated airway epithelial cells by CDV. Applying a coculture approach with DH82 cells, we demonstrated that cell-mediated infection from the basolateral side of well-differentiated epithelial cells is more efficient than infection via cell-free virus. In fact, free virus was unable to infect intact polarized cells. When tight junctions were interrupted by treatment with EGTA, cells became susceptible to infection, with nectin-4 serving as a receptor. Another interesting feature of CDV infection is that infection of well-differentiated airway epithelial cells does not result in virus egress. Cell-free virions are released from the cells only in the presence of an inhibitor of the JAK/STAT signaling pathway. Our results provide new insights into how CDV can overcome the barrier of the airway epithelium and reveal similarities and some dissimilarities compared to measles virus.
Collapse
|
6
|
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
|
7
|
Yadav AK, Rajak KK, Kumar A, Bhatt M, Chakravarti S, Muthu S, Dubal ZB, Khulape S, Yousuf RW, Rai V, Kumar B, Muthuchelvan D, Gupta PK, Singh RP, Singh R. Replication competence of canine distemper virus in cell lines expressing signaling lymphocyte activation molecule (SLAM) of goat, sheep and dog origin. Microb Pathog 2021; 156:104940. [PMID: 33962006 DOI: 10.1016/j.micpath.2021.104940] [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: 12/04/2020] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 11/30/2022]
Abstract
Cellular receptors play an important role in entry and cell to cell spread of morbillivirus infections. The cells expressing SLAM and Nectin-4 have been used for successful and efficient isolation of canine distemper virus (CDV) in high titre. There are several methods for generation of cells expressing receptor molecules. Here, we have used a comparatively cheaper and easily available method, pcDNA 3.1 (+) for engineering Vero cells to express SLAM gene of goat, sheep and dog origin (Vero/Goat/SLAM (VGS), Vero/Sheep/SLAM (VSS) and Vero/Dog/SLAM (VDS), respectively). The generated cell lines were then compared to test their efficacy to support CDV replication. CDV could be grown in high titre in the cells expressing SLAM and a difference of log two could be recorded in virus titre between VDS and native Vero cells. Also, CDV could be grown in a higher titre in VDS as compared to VGS and VSS. The finding of this study supports the preferential use of SLAM expressing cells over the native Vero cells by CDV. Further, the higher titre of CDV in cells expressing dog-SLAM as compared to the cells expressing SLAM of non-CDV hosts (i.e. goat and sheep) points towards the preferential use of dog SLAM by the CDV and may be a plausible reason for differential susceptibility of small ruminants and Canines to CDV.
Collapse
Affiliation(s)
- Ajay Kumar Yadav
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India; ICAR -National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Kaushal Kishor Rajak
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India.
| | - Ashok Kumar
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Mukesh Bhatt
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India; ICAR -National Organic Farming Research Institute, Tadong, Gangtok, Sikkim, 737102, India
| | - Soumendu Chakravarti
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Sankar Muthu
- Division of Parasitology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Z B Dubal
- Division of Veterinary Public Health, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Sagar Khulape
- ICAR-D-FMD, Indian Veterinary Research Institute (IVRI), Mukteswar, 263138, Nainital, Uttarakhand, India
| | - Raja Wasim Yousuf
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Vishal Rai
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Bablu Kumar
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Dhanavelu Muthuchelvan
- Division of Virology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Mukteswar, 263138, Nainital, Uttrakhand, India
| | - Praveen Kumar Gupta
- Division of Animal Biotechnology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Rabindra Prasad Singh
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Rajkumar Singh
- Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India.
| |
Collapse
|
8
|
Sylvatic Canine Morbillivirus in Captive Panthera Highlights Viral Promiscuity and the Need for Better Prevention Strategies. Pathogens 2021; 10:pathogens10050544. [PMID: 33946447 PMCID: PMC8147164 DOI: 10.3390/pathogens10050544] [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: 03/26/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022] Open
Abstract
Canine Distemper Virus (CDV) is a multi-host morbillivirus that infects virtually all Carnivora and a few non-human primates. Here we describe a CDV outbreak in an exotic felid rescue center that led to the death of eight felids in the genus Panthera. Similar to domestic dogs and in contrast to previously described CDV cases in Panthera, severe pneumonia was the primary lesion and no viral antigens or CDV-like lesions were detected in the central nervous system. Four tigers succumbed to opportunistic infections. Viral hemagglutinin (H)-gene sequence was up to 99% similar to strains circulating contemporaneously in regional wildlife. CDV lesions in raccoons and skunk were primarily encephalitis. A few affected felids had at least one previous vaccination for CDV, while most felids at the center were vaccinated during the outbreak. Panthera sharing a fence or enclosure with infected conspecifics had significantly higher chances of getting sick or dying, suggesting tiger-tiger spread was more likely than recurrent spillover. Prior vaccination was incomplete and likely not protective. This outbreak highlights the need for further understanding of CDV epidemiology for species conservation and public health.
Collapse
|
9
|
Wang Y, Chen J, Hu B, Gong C, Shi N, Liu M, Yan X, Bai X, Zhao J. Mink SLAM V-Region V74I Substitutions Contribute to the Formation of Syncytia Induced by Canine Distemper Virus. Front Vet Sci 2021; 7:570283. [PMID: 33585591 PMCID: PMC7874165 DOI: 10.3389/fvets.2020.570283] [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: 06/07/2020] [Accepted: 12/18/2020] [Indexed: 11/20/2022] Open
Abstract
The Signal lymphatic activation molecule (SLAM, also known as CD150) as the cellular receptor of canine distemper virus (CDV) plays an important role in the virus-host interaction. However, it is still unknown whether amino acid differences in the SLAM variable (V) region affect the formation of syncytia. Here, using raccoon dog SLAM (rSLAM) and mink SLAM (mSLAM), we performed SLAM-V homologous modeling, site-directed mutagenesis, and surface expression analysis, as well as a cell fusion assay, to study the interaction between SLAM and CDV. More specifically, our investigation focused on two amino acid residues (74 and 129) of SLAM, previously predicted to play a relevant role in receptor-ligand interaction. Our results indicated that only residues at position 60, 74, and 129 were different between rSLAM and mSLAM among the 29 amino acids that might interact with CDV H, and residues 74 and 129 were located in the interface region interacting with CDV H. The amino acid substitution at the positions of 74 have a significant effect on the expression of mSLAM. The SLAM-V74I mutation in mink significantly improved the cell fusion efficiency of CDV. In contrast, the SLAM-I74V mutation in the raccoon dog significantly decreased cell fusion efficiency. We conclude that residue 74 of SLAM plays an important role during the the formation of syncytia. Only when implementing CDV infection analysis, the rSLAM-Q129R can significantly decreased the mean number of syncytia, but the mSLAM-R129Q can't. Additionally, residue 60 show variability between rSLAM and mSLAM. We believe that our study makes a significant contribution to the literature because we provide molecular data, partially accounting for the differences in host membrane and virus interaction laying the foundation for further molecular work.
Collapse
Affiliation(s)
- Yawen Wang
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, China.,Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jie Chen
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, China
| | - Bo Hu
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, China
| | - Chengyan Gong
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, China
| | - Ning Shi
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, China
| | - Mengjia Liu
- Dongying Customs District, People's Republic of China, Dongying, China
| | - Xijun Yan
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, China
| | - Xue Bai
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, China
| | - Jianjun Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
10
|
Takeda M, Seki F, Yamamoto Y, Nao N, Tokiwa H. Animal morbilliviruses and their cross-species transmission potential. Curr Opin Virol 2020; 41:38-45. [PMID: 32344228 DOI: 10.1016/j.coviro.2020.03.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
Like measles virus (MV), whose primary hosts are humans, non-human animal morbilliviruses use SLAM (signaling lymphocytic activation molecule) and PVRL4 (nectin-4) expressed on immune and epithelial cells, respectively, as receptors. PVRL4's amino acid sequence is highly conserved across species, while that of SLAM varies significantly. However, non-host animal SLAMs often function as receptors for different morbilliviruses. Uniquely, human SLAM is somewhat specific for MV, but canine distemper virus, which shows the widest host range among morbilliviruses, readily gains the ability to use human SLAM. The host range for morbilliviruses is also modulated by their ability to counteract the host's innate immunity, but the risk of cross-species transmission of non-human animal morbilliviruses to humans could occur if MV is successfully eradicated.
Collapse
Affiliation(s)
- Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Nishi-Ikebukuro 3-34-1, Toshima-ku, Tokyo 171-8501, Japan
| | - Naganori Nao
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| | - Hiroaki Tokiwa
- Department of Chemistry, Rikkyo University, Nishi-Ikebukuro 3-34-1, Toshima-ku, Tokyo 171-8501, Japan
| |
Collapse
|
11
|
meng X, Zhu X, Alfred N, Zhang Z. Identification of amino acid residues involved in the interaction between peste-des-petits-ruminants virus haemagglutinin protein and cellular receptors. J Gen Virol 2020; 101:242-251. [PMID: 31859612 PMCID: PMC7416607 DOI: 10.1099/jgv.0.001368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/19/2019] [Indexed: 02/04/2023] Open
Abstract
Peste-des-petits-ruminants virus (PPRV) haemagglutinin (H) protein mediates binding to cellular receptors and then initiates virus entry. To identify the key residues of PPRV H (Hv) protein of the Nigeria 75/1 strain involved in binding to receptors, interaction of the Hv and mutated Hv (mHv) proteins with receptors (SLAM and Nectin 4) and their mutants (mSLAM1, mSLAM2, mSLAM3 and mNectin 4) was investigated using surface plasmon resonance imaging (SPRi) and coimmunoprecipitation (co-IP) assays. The results showed that the Hv protein failed to interact with mSLAM3, but interacted at a strong or medium intensity with SLAM, mSLAM2, Nectin 4 and mNectin 4, and at a low level with mSLAM1. The mHv protein was unable to interact with SLAM and its mutants, but bound to Nectin 4 and mNectin 4 with medium and weak intensity, respectively. Further analysis showed that the Hv protein could precipitate mSLAM1, mSLAM2 and mNectin 4, but not mSLAM3. The mHv protein failed to coprecipitate with SLAM and its mutants. The binding activities of mNectin 4 and Nectin 4 to mHv were less than 30.36 and 51.94 % of the wild-type levels, respectively. Based on the results obtained, amino acids at positions R389, L464, I498, R503, R533, Y541, Y543, F552 and Y553 of H protein and I61, H62, L64, K76, K78, E123, H130, I210, A211, S226 and R227 in SLAM were identified to be essential for the specificity of H-SLAM interaction, while the critical residues of H-Nectin 4 interaction require further study. These findings would improve our understanding of the invasive mechanisms of PPRV.
Collapse
Affiliation(s)
- Xuelian meng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| | - Xueliang Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| | - Niyokwishimira Alfred
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| | - Zhidong Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| |
Collapse
|
12
|
Jiang Y, Jia S, Zheng D, Li F, Wang S, Wang L, Qiao X, Cui W, Tang L, Xu Y, Xia X, Li Y. Protective Immunity against Canine Distemper Virus in Dogs Induced by Intranasal Immunization with a Recombinant Probiotic Expressing the Viral H Protein. Vaccines (Basel) 2019; 7:vaccines7040213. [PMID: 31835572 PMCID: PMC6963260 DOI: 10.3390/vaccines7040213] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/26/2022] Open
Abstract
Canine distemper virus (CDV) elicits a severe contagious disease in a broad range of hosts. CDV mortality rates are 50% in domestic dogs and 100% in ferrets. Its primary infection sites are respiratory and intestinal mucosa. This study aimed to develop an effective mucosal CDV vaccine using a non-antibiotic marked probiotic pPGΔCm-T7g10-EGFP-H/L. casei 393 strain expressing the CDV H protein. Its immunogenicity in BALB/c mice was evaluated using intranasal and oral vaccinations, whereas in dogs the intranasal route was used for vaccination. Our results indicate that this probiotic vaccine can stimulate a high level of secretory immunoglobulin A (sIgA)-based mucosal and IgG-based humoral immune responses in mice. SIgA levels in the nasal lavage and lungs were significantly higher in intranasally vaccinated mice than those in orally vaccinated mice. Both antigen-specific IgG and sIgA antibodies were effectively elicited in dogs through the intranasal route and demonstrated superior immunogenicity. The immune protection efficacy of the probiotic vaccine was evaluated by challenging the immunized dogs with virulent CDV 42 days after primary immunization. Dogs of the pPGΔCm-T7g10-EGFP-H/L. casei 393 group were completely protected against CDV. The proposed probiotic vaccine could be promising for protection against CDV infection in dogs.
Collapse
Affiliation(s)
- Yanping Jiang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Shuo Jia
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Dianzhong Zheng
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Fengsai Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Shengwen Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Li Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Xinyuan Qiao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Wen Cui
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Lijie Tang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Yigang Xu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
| | - Xianzhu Xia
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130000, China
- Correspondence: (X.X.); (Y.L.); Tel./Fax: +86-451-5519-0363 (Y.L.)
| | - Yijing Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (S.J.); (D.Z.); (F.L.); (S.W.); (L.W.); (X.Q.); (W.C.); (L.T.); (Y.X.)
- Correspondence: (X.X.); (Y.L.); Tel./Fax: +86-451-5519-0363 (Y.L.)
| |
Collapse
|
13
|
Tan G, Yi Z, Song H, Xu F, Li F, Aliyari R, Zhang H, Du P, Ding Y, Niu J, Wang X, Su L, Qin FXF, Cheng G. Type-I-IFN-Stimulated Gene TRIM5γ Inhibits HBV Replication by Promoting HBx Degradation. Cell Rep 2019; 29:3551-3563.e3. [PMID: 31825835 PMCID: PMC6996557 DOI: 10.1016/j.celrep.2019.11.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 09/08/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023] Open
Abstract
To understand the molecular mechanisms that mediate the anti-hepatitis B virus (HBV) effect of interferon (IFN) therapy, we conduct high-throughput bimolecular fluorescence complementation screening to identify potential physical interactions between the HBx protein and 145 IFN-stimulated genes (ISGs). Seven HBx-interacting ISGs have consistent and significant inhibitory effects on HBV replication, among which TRIM5γ suppresses HBV replication by promoting K48-linked ubiquitination and degradation of the HBx protein on the K95 ubiquitin site. The B-Box domain of TRIM5γ under overexpression conditions is sufficient to trigger HBx degradation and is responsible both for interacting with HBx and recruiting TRIM31, which is an ubiquitin ligase that triggers HBx ubiquitination. High expression levels of TRIM5γ in IFN-α-treated HBV patients might indicate a better therapeutic effect. Thus, our studies identify a crucial role for TRIM5γ and TRIM31 in promoting HBx degradation, which may facilitate the development of therapeutic agents for the treatment of patients with IFN-resistant HBV infection.
Collapse
Affiliation(s)
- Guangyun Tan
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130061, China.
| | - Zhaohong Yi
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Hongxiao Song
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Fengchao Xu
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Feng Li
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Roghiyh Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Hong Zhang
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin 130021, China
| | - Peishuang Du
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Yanhua Ding
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin 130021, China
| | - Junqi Niu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xiaosong Wang
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Lishan Su
- Department of Immunology, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130061, China; CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - F Xiao-Feng Qin
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu 215123, China.
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
14
|
Host Cellular Receptors for the Peste des Petits Ruminant Virus. Viruses 2019; 11:v11080729. [PMID: 31398809 PMCID: PMC6723671 DOI: 10.3390/v11080729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Peste des Petits Ruminant (PPR) is an important transboundary, OIE-listed contagious viral disease of primarily sheep and goats caused by the PPR virus (PPRV), which belongs to the genus Morbillivirus of the family Paramyxoviridae. The mortality rate is 90–100%, and the morbidity rate may reach up to 100%. PPR is considered economically important as it decreases the production and productivity of livestock. In many endemic poor countries, it has remained an obstacle to the development of sustainable agriculture. Hence, proper control measures have become a necessity to prevent its rapid spread across the world. For this, detailed information on the pathogenesis of the virus and the virus host interaction through cellular receptors needs to be understood clearly. Presently, two cellular receptors; signaling lymphocyte activation molecule (SLAM) and Nectin-4 are known for PPRV. However, extensive information on virus interactions with these receptors and their impact on host immune response is still required. Hence, a thorough understanding of PPRV receptors and the mechanism involved in the induction of immunosuppression is crucial for controlling PPR. In this review, we discuss PPRV cellular receptors, viral host interaction with cellular receptors, and immunosuppression induced by the virus with reference to other Morbilliviruses.
Collapse
|
15
|
Duque-Valencia J, Sarute N, Olarte-Castillo XA, Ruíz-Sáenz J. Evolution and Interspecies Transmission of Canine Distemper Virus-An Outlook of the Diverse Evolutionary Landscapes of a Multi-Host Virus. Viruses 2019; 11:v11070582. [PMID: 31247987 PMCID: PMC6669529 DOI: 10.3390/v11070582] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/13/2019] [Accepted: 05/18/2019] [Indexed: 12/17/2022] Open
Abstract
Canine distemper virus (CDV) is a worldwide distributed virus which belongs to the genus Morbillivirus within the Paramyxoviridae family. CDV spreads through the lymphatic, epithelial, and nervous systems of domestic dogs and wildlife, in at least six orders and over 20 families of mammals. Due to the high morbidity and mortality rates and broad host range, understanding the epidemiology of CDV is not only important for its control in domestic animals, but also for the development of reliable wildlife conservation strategies. The present review aims to give an outlook of the multiple evolutionary landscapes and factors involved in the transmission of CDV by including epidemiological data from multiple species in urban, wild and peri-urban settings, not only in domestic animal populations but at the wildlife interface. It is clear that different epidemiological scenarios can lead to the presence of CDV in wildlife even in the absence of infection in domestic populations, highlighting the role of CDV in different domestic or wild species without clinical signs of disease mainly acting as reservoirs (peridomestic and mesocarnivores) that are often found in peridomestic habits triggering CDV epidemics. Another scenario is driven by mutations, which generate genetic variation on which random drift and natural selection can act, shaping the genetic structure of CDV populations leading to some fitness compensations between hosts and driving the evolution of specialist and generalist traits in CDV populations. In this scenario, the highly variable protein hemagglutinin (H) determines the cellular and host tropism by binding to signaling lymphocytic activation molecule (SLAM) and nectin-4 receptors of the host; however, the multiple evolutionary events that may have facilitated CDV adaptation to different hosts must be evaluated by complete genome sequencing. This review is focused on the study of CDV interspecies transmission by examining molecular and epidemiological reports based on sequences of the hemagglutinin gene and the growing body of studies of the complete genome; emphasizing the importance of long-term multidisciplinary research that tracks CDV in the presence or absence of clinical signs in wild species, and helping to implement strategies to mitigate the infection. Integrated research incorporating the experience of wildlife managers, behavioral and conservation biologists, veterinarians, virologists, and immunologists (among other scientific areas) and the inclusion of several wild and domestic species is essential for understanding the intricate epidemiological dynamics of CDV in its multiple host infections.
Collapse
Affiliation(s)
- July Duque-Valencia
- Grupo de Investigación en Ciencias Animales-GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, sede Medellín 050012, Colombia
| | - Nicolás Sarute
- Sección Genética Evolutiva, Facultad de Ciencias, Universidad de la Republica, Montevideo 11200, Uruguay
- Department of Microbiology and Immunology, UIC College of Medicine, Chicago, IL 60612, USA
| | - Ximena A Olarte-Castillo
- Facultad de Ciencias Exactas, Naturales y Agropecuarias. Universidad de Santander (UDES), sede Bucaramanga 680002, Colombia
| | - Julián Ruíz-Sáenz
- Grupo de Investigación en Ciencias Animales-GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, sede Medellín 050012, Colombia.
| |
Collapse
|
16
|
Yang B, Qi X, Chen Z, Chen S, Xue Q, Jia P, Wang T, Wang J. Binding and entry of peste des petits ruminants virus into caprine endometrial epithelial cells profoundly affect early cellular gene expression. Vet Res 2018; 49:8. [PMID: 29368634 PMCID: PMC5784595 DOI: 10.1186/s13567-018-0504-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 12/18/2017] [Indexed: 12/22/2022] Open
Abstract
Peste des petits ruminants virus (PPRV), the etiological agent of peste des petits ruminants (PPR), causes an acute or subacute disease in small ruminants. Although abortion is observed in an unusually large proportion of pregnant goats during outbreaks of PPR, the pathogenic mechanism underlying remains unclear. Here, the gene expression profile of caprine endometrial epithelial cells (EECs) infected with PPRV Nigeria 75/1 was determined by DNA microarray to investigate the cellular response immediately after viral entry. The microarray analysis revealed that a total of 146 genes were significantly dysregulated by PPRV internalization within 1 h post-infection (hpi). Of these, 85 genes were upregulated and 61 genes were downregulated. Most of these genes, including NFKB1A, JUNB, and IL1A, have not previously been reported in association with PPRV infection in goats. Following viral replication (24 hpi), the expression of 307 genes were significantly upregulated and that of 261 genes were downregulated. The data for the genes differentially expressed in EECs were subjected to a time sequence profile analysis, gene network analysis and pathway analysis. The gene network analysis showed that 13 genes (EIF2AK3, IL10, TLR4, ZO3, NFKBIB, RAC1, HSP90AA1, SMAD7, ARG2, JUNB, ZFP36, APP, and IL1A) were located in the core of the network. We clearly demonstrate that PPRV infection upregulates the expression of nectin-4 after 1 hpi, which peaked at 24 hpi in EECs. In conclusion, this study demonstrates the early cellular gene expression in the caprine endometrial epithelial cells after the binding and entry of PPRV.
Collapse
Affiliation(s)
- Bo Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhijie Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shuying Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, 100000, China
| | - Peilong Jia
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ting Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
17
|
Loots AK, Mitchell E, Dalton DL, Kotzé A, Venter EH. Advances in canine distemper virus pathogenesis research: a wildlife perspective. J Gen Virol 2017; 98:311-321. [PMID: 27902345 DOI: 10.1099/jgv.0.000666] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Canine distemper virus (CDV) has emerged as a significant disease of wildlife, which is highly contagious and readily transmitted between susceptible hosts. Initially described as an infectious disease of domestic dogs, it is now recognized as a global multi-host pathogen, infecting and causing mass mortalities in a wide range of carnivore species. The last decade has seen the effect of numerous CDV outbreaks in various wildlife populations. Prevention of CDV requires a clear understanding of the potential hosts in danger of infection as well as the dynamic pathways CDV uses to gain entry to its host cells and its ability to initiate viral shedding and disease transmission. We review recent research conducted on CDV infections in wildlife, including the latest findings on the causes of host specificity and cellular receptors involved in distemper pathogenesis.
Collapse
Affiliation(s)
- Angelika K Loots
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa.,National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa
| | - Emily Mitchell
- National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa
| | - Desiré L Dalton
- National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa.,Genetics Department, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Antoinette Kotzé
- National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa.,Genetics Department, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Estelle H Venter
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| |
Collapse
|
18
|
Investigation of the morphological transition of a phospholipid bilayer membrane in an external electric field via molecular dynamics simulation. J Mol Model 2017; 23:113. [PMID: 28289956 DOI: 10.1007/s00894-017-3292-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/20/2017] [Indexed: 12/12/2022]
Abstract
Elucidating the mechanisms for morphological transitions of the phospholipid bilayer membrane during cellular activity should lead to greater understanding of these membrane transitions and allow us to optimize biotechnologies such as drug delivery systems in organisms. To investigate the mechanism for and the dynamics of morphological changes in the phospholipid membrane, we performed molecular dynamics simulation of a phospholipid membrane with and without membrane protein under the influence of electric fields with different strengths. In the absence of membrane protein, it was possible to control the transition from one lamellar membrane morphology to another by applying a strong electric field. The strong electric field initially disordered the lipid molecules in the membrane, leading to the formation of a hydrophilic pore. The lipid molecules then spontaneously fused into a new lamellar membrane morphology. In the presence of membrane protein, a morphological transition from lamellar membrane to vesicle under the influence of a strong electric field was observed. Studying the complex transition dynamics associated with these changes in membrane morphology allowed us to gain deep insight into the electrofusion and electroporation that occur in the presence or absence of membrane protein, and the results obtained here should prove useful in work aimed at controlling membrane morphology. Graphical Abstract Memebrane morphological transition under the electric field of 0.6 V/nm with the membrane protein (down) and without membrane protein (up).
Collapse
|
19
|
Canine Distemper Virus Fusion Activation: Critical Role of Residue E123 of CD150/SLAM. J Virol 2015; 90:1622-37. [PMID: 26608324 DOI: 10.1128/jvi.02405-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/18/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Measles virus (MeV) and canine distemper virus (CDV) possess tetrameric attachment proteins (H) and trimeric fusion proteins, which cooperate with either SLAM or nectin 4 receptors to trigger membrane fusion for cell entry. While the MeV H-SLAM cocrystal structure revealed the binding interface, two distinct oligomeric H assemblies were also determined. In one of the conformations, two SLAM units were sandwiched between two discrete H head domains, thus spotlighting two binding interfaces ("front" and "back"). Here, we investigated the functional relevance of both interfaces in activating the CDV membrane fusion machinery. While alanine-scanning mutagenesis identified five critical regulatory residues in the front H-binding site of SLAM, the replacement of a conserved glutamate residue (E at position 123, replaced with A [E123A]) led to the most pronounced impact on fusion promotion. Intriguingly, while determination of the interaction of H with the receptor using soluble constructs revealed reduced binding for the identified SLAM mutants, no effect was recorded when physical interaction was investigated with the full-length counterparts of both molecules. Conversely, although mutagenesis of three strategically selected residues within the back H-binding site of SLAM did not substantially affect fusion triggering, nevertheless, the mutants weakened the H-SLAM interaction recorded with the membrane-anchored protein constructs. Collectively, our findings support a mode of binding between the attachment protein and the V domain of SLAM that is common to all morbilliviruses and suggest a major role of the SLAM residue E123, located at the front H-binding site, in triggering the fusion machinery. However, our data additionally support the hypothesis that other microdomain(s) of both glycoproteins (including the back H-binding site) might be required to achieve fully productive H-SLAM interactions. IMPORTANCE A complete understanding of the measles virus and canine distemper virus (CDV) cell entry molecular framework is still lacking, thus impeding the rational design of antivirals. Both viruses share many biological features that partially rely on the use of analogous Ig-like host cell receptors, namely, SLAM and nectin 4, for entering immune and epithelial cells, respectively. Here, we provide evidence that the mode of binding between the membrane-distal V domain of SLAM and the attachment protein (H) of morbilliviruses is very likely conserved. Moreover, although structural information revealed two discrete conformational states of H, one of the structures displayed two H-SLAM binding interfaces ("front" and "back"). Our data not only spotlight the front H-binding site of SLAM as the main determinant of membrane fusion promotion but suggest that the triggering efficiency of the viral entry machinery may rely on a local conformational change within the front H-SLAM interactive site rather than the binding affinity.
Collapse
|
20
|
Zhao J, Shi N, Sun Y, Martella V, Nikolin V, Zhu C, Zhang H, Hu B, Bai X, Yan X. Pathogenesis of canine distemper virus in experimentally infected raccoon dogs, foxes, and minks. Antiviral Res 2015. [PMID: 26210812 DOI: 10.1016/j.antiviral.2015.07.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Canine distemper virus (CDV) infects a broad range of carnivores and causes a highly contagious disease with severe immunosuppression. The disease severity markedly varies in different species. To investigate the pathogenesis of CDV in raccoon dog (Nyctereutes procyonoides), fox (Vulpes vulpes) and mink (Neovison vison) species, three groups of CDV sero-negative animals were infected with CDV strain LN(10)1. This CDV strain belongs to the Asia-1 genotype, which is epidemiologically predominant in carnivores in China. CDV infection provoked marked differences in virulence in the three species that were studied. Raccoon dogs developed fever, severe conjunctivitis, and pathological lesions, with 100% (5/5) mortality and with high viral RNA loads in organs within 15 days post infection (dpi). In infected foxes, the onset of the disease was delayed, with 40% (2/5) mortality by 21 dpi. Infected minks developed only mild clinical signs and pathological lesions, and mortality was not observed. Raccoon dogs and foxes showed more severe immune suppression (lymphopenia, decreased lymphocyte proliferation, viremia and low-level virus neutralizing antibodies) than minks. We also observed a distinct pattern of cytokine mRNA transcripts at different times after infection. Decreased IFN-γ and IL-4 mRNA responses were evident in the animals with fatal disease, while up-regulation of these cytokines was observed in the animals surviving the infection. Increased TNF-α response was detected in animals with mild or severe clinical signs. Based on the results, we could distinguish three different patterns of disease after experimental CDV infection, e.g. a mild form in minks, a moderate form in foxes and a severe disease in raccoon dogs. The observed differences in susceptibility to CDV could be related to distinct host cytokine profiles. Comparative evaluation of CDV pathogenesis in various animal species is pivotal to generate models suitable for the evaluation of CDV-host interactions and of vaccine response.
Collapse
Affiliation(s)
- Jianjun Zhao
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China; State Key Laboratory for Molecular Biology of Special Economic Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China.
| | - Ning Shi
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Yangang Sun
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Vito Martella
- Department of Veterinary Medicine, University of Bari, Bari, Italy
| | - Veljko Nikolin
- Boehringer Ingelheim Veterinary Research Center GmbH & Co. KG, Hannover, Germany
| | - Chunsheng Zhu
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Hailing Zhang
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Bo Hu
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Xue Bai
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Xijun Yan
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China; State Key Laboratory for Molecular Biology of Special Economic Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China.
| |
Collapse
|
21
|
Ludlow M, McQuaid S, Milner D, de Swart RL, Duprex WP. Pathological consequences of systemic measles virus infection. J Pathol 2014; 235:253-65. [DOI: 10.1002/path.4457] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/30/2014] [Accepted: 10/03/2014] [Indexed: 01/25/2023]
Affiliation(s)
- Martin Ludlow
- Department of Microbiology; Boston University School of Medicine; MA USA
| | - Stephen McQuaid
- Tissue Pathology Laboratories; Belfast Health and Social Care Trust; Northern Ireland
| | - Dan Milner
- Department of Immunology and Infectious Diseases; Harvard School of Public Health; Boston MA USA
- Department of Pathology; Brigham and Women's Hospital; Boston MA USA
| | - Rik L de Swart
- Department of Viroscience; Erasmus MC; Rotterdam The Netherlands
| | - W Paul Duprex
- Department of Microbiology; Boston University School of Medicine; MA USA
| |
Collapse
|
22
|
Mateo M, Navaratnarajah CK, Willenbring RC, Maroun JW, Iankov I, Lopez M, Sinn PL, Cattaneo R. Different roles of the three loops forming the adhesive interface of nectin-4 in measles virus binding and cell entry, nectin-4 homodimerization, and heterodimerization with nectin-1. J Virol 2014; 88:14161-71. [PMID: 25275122 PMCID: PMC4249131 DOI: 10.1128/jvi.02379-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Many viruses utilize cell adhesion molecules of the immunoglobulin superfamily as receptors. In particular, viruses of different classes exploit nectins. The large DNA viruses, herpes simplex and pseudorabies viruses, use ubiquitous nectins 1 and 2. The negative-strand RNA virus measles virus (MeV) uses tissue-specific nectin-4, and the positive-strand RNA virus poliovirus uses nectin-like 5 (necl-5), also known as poliovirus receptor. These viruses contact the BC, C'C", and FG loops on the upper tip of their receptor's most membrane-distal domain. This location corresponds to the newly defined canonical adhesive interface of nectins, but how viruses utilize this interface has remained unclear. Here we show that the same key residues in the BC and FG loops of nectin-4 govern binding to the MeV attachment protein hemagglutinin (H) and cell entry, nectin-4 homodimerization, and heterodimerization with nectin-1. On the other hand, residues in the C'C" loop necessary for homo- and heterotypic interactions are dispensable for MeV-induced fusion and cell entry. Remarkably, the C'C" loop governs dissociation of the nectin-4 and H ectodomains. We provide formal proof that H can interfere with the formation of stable nectin-1/nectin-4 heterodimers. Finally, while developing an alternative model to study MeV spread, we observed that polarized primary pig airway epithelial sheets cannot be infected. We show that a single amino acid variant in the BC loop of pig nectin-4 fully accounts for restricted MeV entry. Thus, the three loops forming the adhesive interface of nectin-4 have different roles in supporting MeV H association and dissociation and MeV-induced fusion. IMPORTANCE Different viruses utilize nectins as receptors. Nectins are immunoglobulin superfamily glycoproteins that mediate cell-cell adhesion in vertebrate tissues. They interact through an adhesive interface located at the top of their membrane-distal domain. How viruses utilize the three loops forming this interface has remained unclear. We demonstrate that while nectin-nectin interactions require residues in all three loops, the association of nectin-4 with the measles virus hemagglutinin requires only the BC and FG loops. However, we discovered that residues in the C'C" loop modulate the dissociation of nectin-4 from the viral hemagglutinin. Analogous mechanisms may support cell entry of other viruses that utilize nectins or other cell adhesion molecules of the immunoglobulin superfamily as receptors.
Collapse
Affiliation(s)
- Mathieu Mateo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Robin C Willenbring
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA Virology and Gene Therapy track, Mayo Graduate School, Rochester, Minnesota, USA
| | - Justin W Maroun
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA Virology and Gene Therapy track, Mayo Graduate School, Rochester, Minnesota, USA
| | - Ianko Iankov
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Marc Lopez
- INSERM, UMR1068/CRCM, Institut Paoli-Calmettes and University of Aix-Marseille, Marseille, France
| | - Patrick L Sinn
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA Virology and Gene Therapy track, Mayo Graduate School, Rochester, Minnesota, USA
| |
Collapse
|
23
|
Melia MM, Earle JP, Abdullah H, Reaney K, Tangy F, Cosby SL. Use of SLAM and PVRL4 and identification of pro-HB-EGF as cell entry receptors for wild type phocine distemper virus. PLoS One 2014; 9:e106281. [PMID: 25171206 PMCID: PMC4149546 DOI: 10.1371/journal.pone.0106281] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Signalling lymphocyte activation molecule (SLAM) has been identified as an immune cell receptor for the morbilliviruses, measles (MV), canine distemper (CDV), rinderpest and peste des petits ruminants (PPRV) viruses, while CD46 is a receptor for vaccine strains of MV. More recently poliovirus like receptor 4 (PVRL4), also known as nectin 4, has been identified as a receptor for MV, CDV and PPRV on the basolateral surface of polarised epithelial cells. PVRL4 is also up-regulated by MV in human brain endothelial cells. Utilisation of PVRL4 as a receptor by phocine distemper virus (PDV) remains to be demonstrated as well as confirmation of use of SLAM. We have observed that unlike wild type (wt) MV or wtCDV, wtPDV strains replicate in African green monkey kidney Vero cells without prior adaptation, suggesting the use of a further receptor. We therefore examined candidate molecules, glycosaminoglycans (GAG) and the tetraspan proteins, integrin β and the membrane bound form of heparin binding epithelial growth factor (proHB-EGF),for receptor usage by wtPDV in Vero cells. We show that wtPDV replicates in Chinese hamster ovary (CHO) cells expressing SLAM and PVRL4. Similar wtPDV titres are produced in Vero and VeroSLAM cells but more limited fusion occurs in the latter. Infection of Vero cells was not inhibited by anti-CD46 antibody. Removal/disruption of GAG decreased fusion but not the titre of virus. Treatment with anti-integrin β antibody increased rather than decreased infection of Vero cells by wtPDV. However, infection was inhibited by antibody to HB-EGF and the virus replicated in CHO-proHB-EGF cells, indicating use of this molecule as a receptor. Common use of SLAM and PVRL4 by morbilliviruses increases the possibility of cross-species infection. Lack of a requirement for wtPDV adaptation to Vero cells raises the possibility of usage of proHB-EGF as a receptor in vivo but requires further investigation.
Collapse
Affiliation(s)
- Mary M. Melia
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - John Philip Earle
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Haniah Abdullah
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Katherine Reaney
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Frederic Tangy
- Viral Genomics and Vaccination Laboratory, Institut Pasteur, CNRS-URA3015, Paris, France
| | - Sara Louise Cosby
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
24
|
Delpeut S, Noyce RS, Richardson CD. The tumor-associated marker, PVRL4 (nectin-4), is the epithelial receptor for morbilliviruses. Viruses 2014; 6:2268-86. [PMID: 24892636 PMCID: PMC4074928 DOI: 10.3390/v6062268] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/14/2014] [Accepted: 05/15/2014] [Indexed: 01/25/2023] Open
Abstract
PVRL4 (nectin-4) was recently identified as the epithelial receptor for members of the Morbillivirus genus, including measles virus, canine distemper virus and peste des petits ruminants virus. Here, we describe the role of PVRL4 in morbillivirus pathogenesis and its promising use in cancer therapies. This discovery establishes a new paradigm for the spread of virus from lymphocytes to airway epithelial cells and its subsequent release into the environment. Measles virus vaccine strains have emerged as a promising oncolytic platform for cancer therapy in the last ten years. Given that PVRL4 is a well-known tumor-associated marker for several adenocarcinoma (lung, breast and ovary), the measles virus could potentially be used to specifically target, infect and destroy cancers expressing PVRL4.
Collapse
Affiliation(s)
- Sebastien Delpeut
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| | - Ryan S Noyce
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| | - Christopher D Richardson
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| |
Collapse
|