1
|
Rode KD, Van Hemert C, Wilson RR, Woodruff SP, Pabilonia K, Ballweber L, Kwok O, Dubey JP. Increased pathogen exposure of a marine apex predator over three decades. PLoS One 2024; 19:e0310973. [PMID: 39441768 PMCID: PMC11498681 DOI: 10.1371/journal.pone.0310973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
Environmental changes associated with global warming create new opportunities for pathogen and parasite transmission in Arctic wildlife. As an apex predator ranging over large, remote areas, changes in pathogens and parasites in polar bears are a useful indicator of changing transmission dynamics in Arctic ecosystems. We examined prevalence and risk factors associated with exposure to parasites and viral and bacterial pathogens in Chukchi Sea polar bears. Serum antibodies to six pathogens were detected and prevalence increased between 1987-1994 and 2008-2017 for five: Toxoplasma gondii, Neospora caninum, Francisella tularensis, Brucella abortus/suis, and canine distemper virus. Although bears have increased summer land use, this behavior was not associated with increased exposure. Higher prevalence of F. tularensis, Coxiella burnetii, and B. abortus/suis antibodies in females compared to males, however, could be associated with terrestrial denning. Exposure was related to diet for several pathogens indicating increased exposure in the food web. Elevated white blood cell counts suggest a possible immune response to some pathogens. Given that polar bears face multiple stressors in association with climate change and are a subsistence food, further work is warranted to screen for signs of disease.
Collapse
Affiliation(s)
- Karyn D. Rode
- U.S. Geological Survey, Alaska Science Center, Anchorage, Alaska, United States of America
| | - Caroline Van Hemert
- U.S. Geological Survey, Alaska Science Center, Anchorage, Alaska, United States of America
| | - Ryan R. Wilson
- U.S. Fish and Wildlife Service, Marine Mammals Management, Anchorage, Alaska, United States of America
| | - Susannah P. Woodruff
- U.S. Fish and Wildlife Service, Marine Mammals Management, Anchorage, Alaska, United States of America
| | - Kristy Pabilonia
- Colorado State University Veterinary Diagnostic Laboratory, Fort Collins, Colorado, United States of America
| | - Lora Ballweber
- Colorado State University Veterinary Diagnostic Laboratory, Fort Collins, Colorado, United States of America
| | - Oliver Kwok
- US Department of Agriculture, Agricultural Research Service, Beltsville, Maryland, United States of America
| | - Jitender P. Dubey
- US Department of Agriculture, Agricultural Research Service, Beltsville, Maryland, United States of America
| |
Collapse
|
2
|
Rendon-Marin S, Rincón-Tabares DS, Tabares-Guevara JH, Arbeláez N, Forero-Duarte JE, Díaz FJ, Robledo SM, Hernandez JC, Ruiz-Saenz J. Evaluation of the Safety and Immunogenicity of a Multiple Epitope Polypeptide from Canine Distemper Virus (CDV) in Mice. Vaccines (Basel) 2024; 12:1140. [PMID: 39460307 PMCID: PMC11511104 DOI: 10.3390/vaccines12101140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Morbillivirus canis is the etiological agent of a highly contagious disease that affects diverse domestic and wild animals. Vaccination is considered the most suitable strategy for controlling CDV dissemination, transmission, and distemper disease. However, the emergence of new CDV strains has led to the need to update the current vaccine strategies employed to prevent CDV infection in domestic and wild animals. Currently, there is a lack of effective alternatives for wild animals. Diverse computational tools, especially peptide-based therapies, enable the development of new universal vaccines. OBJECTIVE The aim of this study was to evaluate the safety and humoral and cellular immune response of a new generation of vaccines based on CDV peptides as single-peptide mixtures or multiepitope CDV polypeptides in mice. METHODS Twenty-four BALB/c mice were subjected to a three-dose regimen for 28 days. Seroconversion was evaluated via ELISA, and cellular immune responses were evaluated via flow cytometry through activation-induced markers (AIMs). RESULTS Compared with the placebo, the peptide mixture and multiepitope CDV polypeptide were safe, and seroconversion was statistically significant in the multiepitope CDV polypeptide and commercial vaccine (CV) groups. The numbers of antigen-specific CD4+CD134+ and IFN-γ+ T cells, CD8+ T cells and TNF-α- and IL-6-producing cells were greater in the mice immunized with the multiepitope CDV polypeptide than in the control mice. CONCLUSION This combined approach represents a potential step forward in developing new immunization candidates or enhancing current commercial vaccines to control CDV disease in domestic dogs and wild animals.
Collapse
Affiliation(s)
- Santiago Rendon-Marin
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680001, Colombia;
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050001, Colombia;
| | - Daniel-Santiago Rincón-Tabares
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Jorge H. Tabares-Guevara
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Natalia Arbeláez
- Grupo PECET, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (N.A.); (S.M.R.)
| | - Jorge E. Forero-Duarte
- Grupo de Investigación en Microbiología Ambiental, Escuela de Microbiología, Universidad de Antioquia, Medellín 050001, Colombia;
| | - Francisco J. Díaz
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Sara M. Robledo
- Grupo PECET, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (N.A.); (S.M.R.)
| | - Juan C. Hernandez
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050001, Colombia;
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (D.-S.R.-T.); (J.H.T.-G.); (F.J.D.)
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680001, Colombia;
| |
Collapse
|
3
|
Herzog CM, Aklilu F, Sibhatu D, Shegu D, Belaineh R, Mohammed AA, Kidane M, Schulz C, Willett BJ, Cleaveland S, Bailey D, Peters AR, Cattadori IM, Hudson PJ, Asgedom H, Buza J, Forza MS, Chibssa TR, Gebre S, Juleff N, Bjørnstad ON, Baron MD, Kapur V. Empirical and model-based evidence for a negligible role of cattle in peste des petits ruminants virus transmission and eradication. Commun Biol 2024; 7:937. [PMID: 39095591 PMCID: PMC11297268 DOI: 10.1038/s42003-024-06619-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Peste des petits ruminants virus (PPRV) is a multi-host pathogen with sheep and goats as main hosts. To investigate the role of cattle in the epidemiology of PPR, we simulated conditions similar to East African zero-grazing husbandry practices in a series of trials with local Zebu cattle (Bos taurus indicus) co-housed with goats (Capra aegagrus hircus). Furthermore, we developed a mathematical model to assess the impact of PPRV-transmission from cattle to goats. Of the 32 cattle intranasally infected with the locally endemic lineage IV strain PPRV/Ethiopia/Habru/2014 none transmitted PPRV to 32 co-housed goats. However, these cattle or cattle co-housed with PPRV-infected goats seroconverted. The results confirm previous studies that cattle currently play a negligible role in PPRV-transmission and small ruminant vaccination is sufficient for eradication. However, the possible emergence of PPRV strains more virulent for cattle may impact eradication. Therefore, continued monitoring of PPRV circulation and evolution is recommended.
Collapse
Affiliation(s)
- Catherine M Herzog
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA.
| | | | | | | | | | | | | | - Claudia Schulz
- Institute of Virology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Brian J Willett
- MRC-University of Glasgow Centre for Virus Research, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Sarah Cleaveland
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Andrew R Peters
- Supporting Evidence Based Interventions (SEBI), University of Edinburgh, Edinburgh, UK
| | - Isabella M Cattadori
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA
| | - Peter J Hudson
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA
| | | | - Joram Buza
- Nelson Mandela African Institute of Science and Technology, Arusha, Tanzania
| | | | | | | | - Nick Juleff
- Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Ottar N Bjørnstad
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA
| | | | - Vivek Kapur
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
4
|
Chang X, Su H, Ma S, Li Y, Tan Y, Li Y, Dong S, Lin J, Zhou B, Zhang H. Transcriptome analysis of Vero cells infected with attenuated vaccine strain CDV-QN-1. Microb Pathog 2024; 193:106786. [PMID: 38971506 DOI: 10.1016/j.micpath.2024.106786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
To better understand the interaction between attenuated vaccines and host antiviral responses, we used bioinformatics and public transcriptomics data to analyze the immune response mechanisms of host cells after canine distemper virus (CDV) infection in Vero cells and screened for potential key effector factors. In this study, CDV-QN-1 infect with Vero cells at an MOI of 0.5, and total RNA was extracted from the cells 24 h later and reverse transcribed into cDNA. Transcriptome high-throughput sequencing perform using Illumina. The results showed that 438 differentially expressed genes were screened, of which 409 were significantly up-regulated and 29 were significantly down-regulated. Eight differentially expressed genes were randomly selected for RT-qPCR validation, and the change trend was consistent with the transcriptomics data. GO and KEGG analysis of differentially expressed genes revealed that most of the differentially expressed genes in CDV-QN-1 infection in the early stage were related to immune response and antiviral activity. The enriched signaling pathways mainly included the interaction between cytokines and cytokine receptors, the NF-kappa B signaling pathway, the Toll-like receptor signaling pathway, and the NOD-like receptor signaling pathway. This study provides a foundation for further exploring the pathogenesis of CDV and the innate immune response of host cells in the early stage of infection.
Collapse
Affiliation(s)
- Xiaoyun Chang
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Hong Su
- China Animal Health and Epidemiology Center, Qingdao, Shandong, China
| | - Shuai Ma
- Qingdao Animal Disease Prevention and Control Center, Qingdao, Shandong, China
| | - Yingguang Li
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yue Tan
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yan Li
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Shaoming Dong
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Jiaxu Lin
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Baokun Zhou
- Qingdao Jimo District Animal Health Quarantine Center, Qingdao, Shandong, China.
| | - Hongliang Zhang
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China.
| |
Collapse
|
5
|
Libbey JE, Fujinami RS. Morbillivirus: A highly adaptable viral genus. Heliyon 2023; 9:e18095. [PMID: 37483821 PMCID: PMC10362132 DOI: 10.1016/j.heliyon.2023.e18095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/26/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Over the course of human history, numerous diseases have been caused by the transmission of viruses from an animal reservoir into the human population. The viruses of the genus Morbillivirus are human and animal pathogens that emerged from a primordial ancestor a millennia ago and have been transmitting to new hosts, adapting, and evolving ever since. Through interaction with susceptible individuals, as yet undiscovered morbilliviruses or existing morbilliviruses in animal hosts could cause future zoonotic diseases in humans.
Collapse
|
6
|
Ikegame S, Carmichael JC, Wells H, Furler O'Brien RL, Acklin JA, Chiu HP, Oguntuyo KY, Cox RM, Patel AR, Kowdle S, Stevens CS, Eckley M, Zhan S, Lim JK, Veit EC, Evans MJ, Hashiguchi T, Durigon E, Schountz T, Epstein JH, Plemper RK, Daszak P, Anthony SJ, Lee B. Metagenomics-enabled reverse-genetics assembly and characterization of myotis bat morbillivirus. Nat Microbiol 2023; 8:1108-1122. [PMID: 37142773 PMCID: PMC11089651 DOI: 10.1038/s41564-023-01380-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 04/06/2023] [Indexed: 05/06/2023]
Abstract
Morbilliviruses are among the most contagious viral pathogens of mammals. Although previous metagenomic surveys have identified morbillivirus sequences in bats, full-length morbilliviruses from bats are limited. Here we characterize the myotis bat morbillivirus (MBaMV) from a bat surveillance programme in Brazil, whose full genome was recently published. We demonstrate that the fusion and receptor binding protein of MBaMV utilize bat CD150 and not human CD150, as an entry receptor in a mammalian cell line. Using reverse genetics, we produced a clone of MBaMV that infected Vero cells expressing bat CD150. Electron microscopy of MBaMV-infected cells revealed budding of pleomorphic virions, a characteristic morbillivirus feature. MBaMV replication reached 103-105 plaque-forming units ml-1 in human epithelial cell lines and was dependent on nectin-4. Infection of human macrophages also occurred, albeit 2-10-fold less efficiently than measles virus. Importantly, MBaMV is restricted by cross-neutralizing human sera elicited by measles, mumps and rubella vaccination and is inhibited by orally bioavailable polymerase inhibitors in vitro. MBaMV-encoded P/V genes did not antagonize human interferon induction. Finally, we show that MBaMV does not cause disease in Jamaican fruit bats. We conclude that, while zoonotic spillover into humans may theoretically be plausible, MBaMV replication would probably be controlled by the human immune system.
Collapse
Affiliation(s)
- Satoshi Ikegame
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jillian C Carmichael
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Heather Wells
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Robert L Furler O'Brien
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Joshua A Acklin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hsin-Ping Chiu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Robert M Cox
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Aum R Patel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shreyas Kowdle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christian S Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miles Eckley
- Center for Vector-borne Infectious Diseases Department of Microbiology, Immunology and Pathology College of Veterinary Medicine Colorado State University, Fort Collins, CO, USA
| | - Shijun Zhan
- Center for Vector-borne Infectious Diseases Department of Microbiology, Immunology and Pathology College of Veterinary Medicine Colorado State University, Fort Collins, CO, USA
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ethan C Veit
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew J Evans
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Edison Durigon
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Tony Schountz
- Center for Vector-borne Infectious Diseases Department of Microbiology, Immunology and Pathology College of Veterinary Medicine Colorado State University, Fort Collins, CO, USA
| | | | - Richard K Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | | | - Simon J Anthony
- Department of Pathology, Microbiology, and Immunology, UC Davis School of Veterinary Medicine, Davis, CA, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
7
|
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
|
8
|
Pujol J, Rousseau C, Vergneau-Grosset C. SEROLOGIC RESPONSE AND ADVERSE EFFECTS OF RECOMBINANT CANINE DISTEMPER VACCINATION IN THREE AQUARIUM-HOUSED WALRUSES ( ODOBENUS ROSMARUS). J Zoo Wildl Med 2023; 54:131-136. [PMID: 36971637 DOI: 10.1638/2022-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 03/29/2023] Open
Abstract
Fatalities have been associated with phocine and canine distemper viruses in marine mammals, including pinnipeds. No data are available regarding distemper disease or vaccination in walruses. This study evaluates seroconversion and clinical adverse effects following administration of a canarypox-vectored recombinant distemper vaccination (two 1-ml doses, 3 wk apart) in three adult aquarium-housed walruses. Serum antibodies to distemper were measured using seroneutralization on blood samples collected under operant conditioning prior to and for 12 mon after vaccination or until titers were <32. All walruses seroconverted. Medium positive titers (64-128) were detected for 4 to 9.5 mon in two of three individuals. Interindividual variability was noted, with one individual displaying only low positive titers. Major swelling at the site of injection and lameness for a week following injection occurred in all three walruses. Further studies on dosing amount and interval are needed to make vaccine recommendations in this species.
Collapse
Affiliation(s)
- Julie Pujol
- Département de sciences cliniques, Faculty of Veterinary Medicine of the Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | | | - Claire Vergneau-Grosset
- Département de sciences cliniques, Faculty of Veterinary Medicine of the Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada,
| |
Collapse
|
9
|
Ferguson JM, González-González A, Kaiser JA, Winzer SM, Anast JM, Ridenhour B, Miura TA, Parent CE. Hidden variable models reveal the effects of infection from changes in host survival. PLoS Comput Biol 2023; 19:e1010910. [PMID: 36812266 PMCID: PMC9987815 DOI: 10.1371/journal.pcbi.1010910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/06/2023] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
The impacts of disease on host vital rates can be demonstrated using longitudinal studies, but these studies can be expensive and logistically challenging. We examined the utility of hidden variable models to infer the individual effects of infectious disease from population-level measurements of survival when longitudinal studies are not possible. Our approach seeks to explain temporal deviations in population-level survival after introducing a disease causative agent when disease prevalence cannot be directly measured by coupling survival and epidemiological models. We tested this approach using an experimental host system (Drosophila melanogaster) with multiple distinct pathogens to validate the ability of the hidden variable model to infer per-capita disease rates. We then applied the approach to a disease outbreak in harbor seals (Phoca vituline) that had data on observed strandings but no epidemiological data. We found that our hidden variable modeling approach could successfully detect the per-capita effects of disease from monitored survival rates in both the experimental and wild populations. Our approach may prove useful for detecting epidemics from public health data in regions where standard surveillance techniques are not available and in the study of epidemics in wildlife populations, where longitudinal studies can be especially difficult to implement.
Collapse
Affiliation(s)
- Jake M. Ferguson
- Department of Biology, University of Hawaiʻi at Mānoa, Honolulu, Hawaii, United States of America
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
| | - Andrea González-González
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
- Department of Biology, University of Florida, Gainesville, Florida, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Johnathan A. Kaiser
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Sara M. Winzer
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Justin M. Anast
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Ben Ridenhour
- Department of Mathematics, University of Idaho, Moscow, Idaho, United States of America
| | - Tanya A. Miura
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Christine E. Parent
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- Institute for Interdisciplinary Data Sciences, University of Idaho, Moscow, Idaho, United States of America
| |
Collapse
|
10
|
Canine Distemper Virus in Endangered Species: Species Jump, Clinical Variations, and Vaccination. Pathogens 2022; 12:pathogens12010057. [PMID: 36678405 PMCID: PMC9862170 DOI: 10.3390/pathogens12010057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Canine morbillivirus (Canine distemper virus, CDV) is the cause of distemper in a large number of different species, some of which are endangered. The clinical outcome associated with infection is variable and based on many factors, including the host species, the immune response of the individual animal to the infection, and variation in virus tropism and virulence. Unfortunately, the viral characteristics associated with virulence versus attenuation are not fully characterized, nor are the specific mutations that allow this virus to easily move and adapt from one species to another. Due to its wide host range, this virus is difficult to manage in ecosystems that are home to endangered species. Vaccination of the domestic dog, historically considered the reservoir species for this virus, at dog-wildlife interfaces has failed to control virus spread. CDV appears to be maintained by a metareservoir rather than a single species, requiring the need to vaccinate the wildlife species at risk. This is controversial, and there is a lack of a safe, effective vaccine for nondomestic species. This review focuses on topics that are paramount to protecting endangered species from a stochastic event, such as a CDV outbreak, that could lead to extinction.
Collapse
|
11
|
George AM, Wille M, Wang J, Anderson K, Cohen S, Moselen J, Yang Lee LY, Suen WW, Bingham J, Dalziel AE, Whitney P, Stannard H, Hurt AC, Williams DT, Deng YM, Barr IG. A novel and highly divergent Canine Distemper Virus lineage causing distemper in ferrets in Australia. Virology 2022; 576:117-126. [DOI: 10.1016/j.virol.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 11/28/2022]
|
12
|
Karki M, Rajak KK, Singh RP. Canine morbillivirus (CDV): a review on current status, emergence and the diagnostics. Virusdisease 2022; 33:309-321. [PMID: 36039286 PMCID: PMC9403230 DOI: 10.1007/s13337-022-00779-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/15/2022] [Indexed: 11/12/2022] Open
Abstract
The increasing host range of canine morbillivirus (CDV) affecting important wildlife species such as Lions, Leopard, and Red Pandas has raised the concern. Canine distemper is a pathogen of dogs affecting the respiratory, gastrointestinal, and nervous systems. Seventeen lineages of CDV are reported, and the eighteenth lineage was proposed in 2019 from India. Marked genomic differences in the genome of wild-type virus and vaccine strain are also reported.The variations at the epitope level can be differentiated using specific monoclonal antibodies in neutralization tests. Keeping in mind the current status of the emergence of CDV, genetic and molecular study of circulating strains of the specific geographical region are the essential components of the disease control strategy. New target-based diagnostics and vaccines are in need to counter the effects of the emerging virus population. Control of CDV is necessary to save the endangered, vulnerable, and many other wildlife species to maintain balance in the ecological system. This review provides an overview on emergence reported in CDV, diagnostics developed till today, and a perspective on the disease control strategy, keeping wildlife in consideration.
Collapse
|
13
|
Divergent Viruses Discovered in Swine Alter the Understanding of Evolutionary History and Genetic Diversity of the Respirovirus Genus and Related Porcine Parainfluenza Viruses. Microbiol Spectr 2022; 10:e0024222. [PMID: 35647875 PMCID: PMC9241844 DOI: 10.1128/spectrum.00242-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Paramyxoviridae is a rapidly growing family of viruses, whose potential for cross-species transmission makes it difficult to predict the harm of newly emerging viruses to humans and animals. To better understand their diversity, evolutionary history, and co-evolution with their hosts, we analyzed a collection of porcine parainfluenza virus (PPIV) genomes to reconstruct the species classification basis and evolutionary history of the Respirovirus genus. We sequenced 17 complete genomes of porcine respirovirus 1 (also known as porcine parainfluenza virus 1; PPIV-1), thereby nearly tripling the number of currently available PPIV-1 genomes. We found that PPIV-1 was widely prevalent in China with two divergent lineages, PPIV-1a and PPIV-1b. We further provided evidence that a new species, porcine parainfluenza virus 2 (PPIV-2), had recently emerged in China. Our results pointed to a need for revising the current species demarcation criteria of the Respirovirus genus. In addition, we used PPIV-1 as an example to explore recombination and diversity of the Respirovirus genus. Interestingly, we only detected heterosubtypic recombination events between PPIV-1a and PPIV-1b with no intrasubtypic recombination events. The recombination hotspots highlighted a diverse geography-dependent genome structure of paramyxovirus infecting swine in China. Furthermore, we found no evidence of co-evolution between respirovirus and its host, indicating frequent cross-species transmission. In summary, our analyses showed that swine can be infected with a broad range of respiroviruses and recombination may serve as an important evolutionary mechanism for the Respirovirus genus’ greater diversity in genome structure than previously anticipated. IMPORTANCE Livestock have emerged as critically underrecognized sources of paramyxovirus diversity, including pigs serving as the source of Nipah virus (NiV) and swine parainfluenza virus type 3, and goats and bovines harboring highly divergent viral lineages. Here, we identified a new species of Respirovirus genus named PPIV-2 in swine and proposed to revise the species demarcation criteria of the Respirovirus genus. We found heterosubtypic recombination events and high genetic diversity in PPIV-1. Further, we showed that genetic recombination may have occurred in the Respirovirus genus which may be associated with host range expansion. The continued expansion of Respirovirus genus diversity in livestock with relatively high human contact rates requires enhanced surveillance and ongoing evaluation of emerging cross-species transmission threats.
Collapse
|
14
|
Gainor K, Ghosh S. A comprehensive review of viruses in terrestrial animals from the Caribbean islands of Greater and Lesser Antilles. Transbound Emerg Dis 2022; 69:e1299-e1325. [PMID: 35578793 DOI: 10.1111/tbed.14595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 11/26/2022]
Abstract
Viruses pose a major threat to animal health worldwide, causing significant mortalities and morbidities in livestock, companion animals and wildlife, with adverse implications on human health, livelihoods, food safety and security, regional/national economies, and biodiversity. The Greater and Lesser Antilles consist of a cluster of islands between the North and South Americas and is habitat to a wide variety of animal species. This review is the first to put together decades of information on different viruses circulating in companion animals, livestock, and wildlife from the Caribbean islands of Greater and Lesser Antilles. Although animal viral diseases have been documented in the Caribbean region since the 1940s, we found that studies on different animal viruses are limited, inconsistent, and scattered. Furthermore, a significant number of the reports were based on serological assays, yielding preliminary data. The available information was assessed to identify knowledge gaps and limitations, and accordingly, recommendations were made, with the overall goal to improve animal health and production, and combat zoonoses in the region. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kerry Gainor
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, St. Kitts, West Indies
| | - Souvik Ghosh
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, St. Kitts, West Indies
| |
Collapse
|
15
|
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
|
16
|
Lee B, Ikegame S, Carmichael J, Wells H, Furler R, Acklin J, Chiu HP, Oguntuyo K, Cox R, Patel A, Kowdle S, Stevens C, Eckley M, Zhan S, Lim J, Hashiguchi T, Durigon EL, Schountz T, Epstein J, Plemper R, Daszak P, Anthony S. Zoonotic potential of a novel bat morbillivirus. RESEARCH SQUARE 2021. [PMID: 34611656 PMCID: PMC8491849 DOI: 10.21203/rs.3.rs-926789/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Bats are significant reservoir hosts for many viruses with zoonotic potential1. SARS-CoV-2, Ebola virus, and Nipah virus are examples of such viruses that have caused deadly epidemics and pandemics when spilled over from bats into human and animal populations2,3. Careful surveillance of viruses in bats is critical for identifying potential zoonotic pathogens. However, metagenomic surveys in bats often do not result in full-length viral sequences that can be used to regenerate such viruses for targeted characterization4. Here, we identify and characterize a novel morbillivirus from a vespertilionid bat species (Myotis riparius) in Brazil, which we term myotis bat morbillivirus (MBaMV). There are 7 species of morbilliviruses including measles virus (MeV), canine distemper virus (CDV) and rinderpest virus (RPV)5. All morbilliviruses cause severe disease in their natural hosts6–10, and pathogenicity is largely determined by species specific expression of canonical morbillivirus receptors, CD150/SLAMF111 and NECTIN412. MBaMV used Myotis spp CD150 much better than human and dog CD150 in fusion assays. We confirmed this using live MBaMV that was rescued by reverse genetics. Surprisingly, MBaMV replicated efficiently in primary human myeloid but not lymphoid cells. Furthermore, MBaMV replicated in human epithelial cells and used human NECTIN4 almost as well as MeV. Our results demonstrate the unusual ability of MBaMV to infect and replicate in some human cells that are critical for MeV pathogenesis and transmission. This raises the specter of zoonotic transmission of a bat morbillivirus.
Collapse
|
17
|
Oleaga Á, Vázquez CB, Royo LJ, Barral TD, Bonnaire D, Armenteros JÁ, Rabanal B, Gortázar C, Balseiro A. Canine distemper virus in wildlife in south-western Europe. Transbound Emerg Dis 2021; 69:e473-e485. [PMID: 34536064 DOI: 10.1111/tbed.14323] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
Multi-host pathogens emerging and re-emerging at the wildlife-domestic animal interface affect wildlife management and conservation. This is the case of canine distemper virus (CDV), a paramyxovirus closely related to human measles virus and rinderpest virus of cattle. With an area of 10,603 km2 , Asturias region in Atlantic Spain is a hotspot of carnivore diversity, which includes the largest Eurasian brown bear (Ursus arctos arctos) population and one of the largest wolf (Canis lupus) populations in south-western Europe. In 2020-2021, we recorded mortality due to distemper in four carnivore species including three mustelids (Eurasian badger Meles meles, European marten Martes martes and European polecat Mustela putorius) and one canid (red fox, Vulpes vulpes). Clinical signs and pathology were similar across species and consistent with the emergence of a highly pathogenic viral strain, with CDV antigen mainly located in the central nervous system, lungs, spleen and lymph nodes. A molecular study in eight wild carnivore species, also including the Iberian wolf, Eurasian brown bear, American mink (Neovison vison) and stone marten (Martes foina), revealed 19.51% (16/82) of positivity. Phylogenetic analysis demonstrated that CDV belonged to the previously described European lineage. A retrospective serosurvey (2008-2020) showed a high seroprevalence of CDV antibodies (43.4%) in 684 analyzed badgers, indicating a long-term though not stable viral circulation in this multi-host community. The possible triggers of the 2020-2021 outbreak and the implications for carnivore management and conservation are discussed.
Collapse
Affiliation(s)
- Álvaro Oleaga
- Sociedad de Servicios del Principado de Asturias S.A. (SERPA), Gijón, Spain
| | - Cristina Blanco Vázquez
- Servicio Regional de Investigación y Desarrollo Agroalimentario del Principado de Asturias (SERIDA), Villaviciosa, Spain
| | - Luis José Royo
- Servicio Regional de Investigación y Desarrollo Agroalimentario del Principado de Asturias (SERIDA), Villaviciosa, Spain
| | - Thiago Doria Barral
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Debby Bonnaire
- Ecole Supérieure d'Ingénieurs Agroalimentaires de Bretagne atlantique, Université de Bretagne Occidentale, Brest, France.,Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - José Ángel Armenteros
- Consejería de Fomento, Ordenación del Territorio y Medio Ambiente del Principado de Asturias, Oviedo, Spain
| | - Benjamín Rabanal
- Laboratorio de Técnicas Instrumentales, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Christian Gortázar
- SaBio. Instituto de Investigación en Recursos Cinegéticos-IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Ana Balseiro
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain.,Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (CSIC-Universidad de León), León, Spain
| |
Collapse
|
18
|
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
|
19
|
Li Y, Yi L, Cheng S, Wang Y, Wang J, Sun J, Zhang Q, Xu X. Inhibition of canine distemper virus replication by blocking pyrimidine nucleotide synthesis with A77 1726, the active metabolite of the anti-inflammatory drug leflunomide. J Gen Virol 2021; 102. [PMID: 33416466 DOI: 10.1099/jgv.0.001534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Canine distemper virus (CDV) is the aetiological agent that causes canine distemper (CD). Currently, no antiviral drugs have been approved for CD treatment. A77 1726 is the active metabolite of the anti-rheumatoid arthritis (RA) drug leflunomide. It inhibits the activity of Janus kinases (JAKs) and dihydroorotate dehydrogenase (DHO-DHase), a rate-limiting enzyme in de novo pyrimidine nucleotide synthesis. A77 1726 also inhibits the activity of p70 S6 kinase (S6K1), a serine/threonine kinase that phosphorylates and activates carbamoyl-phosphate synthetase (CAD), a second rate-limiting enzyme in the de novo pathway of pyrimidine nucleotide synthesis. Our present study focuses on the ability of A77 1726 to inhibit CDV replication and its underlying mechanisms. Here we report that A77 1726 decreased the levels of the N and M proteins of CDV and lowered the virus titres in the conditioned media of CDV-infected Vero cells. CDV replication was not inhibited by Ruxolitinib (Rux), a JAK-specific inhibitor, but by brequinar sodium (BQR), a DHO-DHase-specific inhibitor, and PF-4708671, an S6K1-specific inhibitor. Addition of exogenous uridine, which restores intracellular pyrimidine nucleotide levels, blocked the antiviral activity of A77 1726, BQR and PF-4708671. A77 1726 and PF-4708671 inhibited the activity of S6K1 in CDV-infected Vero cells, as evidenced by the decreased levels of CAD and S6 phosphorylation. S6K1 knockdown suppressed CDV replication and enhanced the antiviral activity of A77 1726. These observations collectively suggest that the antiviral activity of A77 1726 against CDV is mediated by targeting pyrimidine nucleotide synthesis via inhibiting DHO-DHase activity and S6K1-mediated CAD activation.
Collapse
Affiliation(s)
- Yao Li
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Li Yi
- State Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Economic Animals and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Sipeng Cheng
- State Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Economic Animals and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Yongshan Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing 210014, PR China
| | - Jiongjiong Wang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Jing Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Quan Zhang
- Institutes of Agricultural Science and Technology Development, Yangzhou University Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, PR China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China.,College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Xiulong Xu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China.,Institutes of Agricultural Science and Technology Development, Yangzhou University Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, PR China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| |
Collapse
|
20
|
Codon usage bias in the H gene of canine distemper virus. Microb Pathog 2020; 149:104511. [PMID: 32961282 DOI: 10.1016/j.micpath.2020.104511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/30/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022]
Abstract
Canine distemper virus (CDV), a non-segmented single negative-stranded RNA (ssRNA), is the etiological agent of canine distemper. Canine distemper is a highly contagious and lethal viral disease in domestic dogs and wild carnivores. Study of the evolution of CDV presents an essential key to improve the vaccine efficacy. In this study, a total of 328 full-length CDV hemagglutinin (H) gene sequences were subjected to phylogenetic, amino acid mutations, and codon usage analysis. In accordance with previous study, CDV genotypes consisted of fifteen lineages. The unique amino acid substitution sites in each CDV lineages have been identified for the first time, including America-1 (Q330H), America-2 (I585S), Asia-1 (A359V), Asia-2 (H61R), Asia-3 (P108Q), Asia-4 (K213T), India-1/Asia-5(S497P), Arctic (S20L), Africa-1(N489S), Colombian (V41I), EWL (I44V), Europe (D560E), Europe-1/South America-1(K161Q), South America-2 (R580Q), and East African (S214A). Codon usage analysis indicated that H gene exhibited low codon usage bias and further neutrality plot analysis demonstrated that natural selection played a dominated role in driving CPV evolution. The effective number of codons (ENC) plots show that all the different sequences are below the standard curve, indicating that mutational pressure is not the only factor affecting CUB but other forces, including natural selection. The neutrality analysis showed that the slope of the regression line was 0.1501, indicating natural selection dominates directional mutation pressure in driving the codon usage pattern. In addition, nucleotide composition, relative synonymous codon usage value, dinucleotide content, and geographical distribution have been proven to influence the codon usage bias of the CDV H gene. The novel findings enhanced the understanding of CDV evolution.
Collapse
|
21
|
Bailey D. Morbilliviruses: Entry, Exit and Everything In-Between. Viruses 2019; 11:v11111036. [PMID: 31703308 PMCID: PMC6893775 DOI: 10.3390/v11111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 11/16/2022] Open
Affiliation(s)
- Dalan Bailey
- The Pirbright Institute, Ash Rd., Guildford GU24 0NF, UK
| |
Collapse
|