1
|
Branda F, Pavia G, Ciccozzi A, Quirino A, Marascio N, Matera G, Romano C, Locci C, Azzena I, Pascale N, Sanna D, Casu M, Ceccarelli G, Ciccozzi M, Scarpa F. Zoonotic Paramyxoviruses: Evolution, Ecology, and Public Health Strategies in a Changing World. Viruses 2024; 16:1688. [PMID: 39599803 PMCID: PMC11599060 DOI: 10.3390/v16111688] [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: 09/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
The family Paramyxoviridae includes a number of negative RNA viruses known for their wide host range and significant zoonotic potential. In recent years, there has been a surge in the identification of emerging zoonotic paramyxoviruses, particularly those hosted by bat species, which serve as key reservoirs. Among these, the genera Henipavirus and Pararubulavirus are of particular concern. Henipaviruses, including the highly pathogenic Hendra and Nipah viruses, have caused severe outbreaks with high mortality rates in both humans and animals. In contrast, zoonotic pararubulaviruses such as the Menangle virus typically induce mild symptoms or remain asymptomatic in human hosts. This review summarizes current knowledge on the evolution, ecology, and epidemiology of emerging zoonotic paramyxoviruses, focusing on recently discovered viruses and their potential to cause future epidemics. We explore the molecular mechanisms underlying host-switching events, viral replication strategies, and immune evasion tactics that facilitate interspecies transmission. In addition, we discuss ecological factors influencing virus emergence, including changes in bat populations and habitats and the role of wildlife-human interfaces. We also examine the public health impact of these emerging viruses, underlining the importance of enhanced surveillance, developing improved diagnostic tools, and implementing proactive strategies to prevent potential outbreaks. By providing a comprehensive overview of recent advances and gaps in knowledge, this review aims to inform future research directions and public health policies related to zoonotic paramyxoviruses.
Collapse
Affiliation(s)
- Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy (M.C.)
| | - Grazia Pavia
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Alessandra Ciccozzi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
| | - Angela Quirino
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Nadia Marascio
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Giovanni Matera
- Unit of Clinical Microbiology, Department of Health Sciences, “Magna Græcia” University of Catanzaro—“Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (G.P.); (A.Q.); (N.M.); (G.M.)
| | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy (M.C.)
| | - Chiara Locci
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
| | - Ilenia Azzena
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
| | - Noemi Pascale
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
- Department of Chemical Physical Mathematical and Natural Sciences, University of Sassari, 07100 Sassari, Italy
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
| | - Marco Casu
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (I.A.); (N.P.); (M.C.)
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University Hospital Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy;
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy (M.C.)
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.C.); (C.L.); (D.S.)
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Fukuhara H, Yumoto K, Sako M, Kajikawa M, Ose T, Kawamura M, Yoda M, Chen S, Ito Y, Takeda S, Mwaba M, Wang J, Hashiguchi T, Kamishikiryo J, Maita N, Kitatsuji C, Takeda M, Kuroki K, Maenaka K. Glycan-shielded homodimer structure and dynamical features of the canine distemper virus hemagglutinin relevant for viral entry and efficient vaccination. eLife 2024; 12:RP88929. [PMID: 39046448 PMCID: PMC11268888 DOI: 10.7554/elife.88929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
Abstract
Canine distemper virus (CDV) belongs to morbillivirus, including measles virus (MeV) and rinderpest virus, which causes serious immunological and neurological disorders in carnivores, including dogs and rhesus monkeys, as recently reported, but their vaccines are highly effective. The attachment glycoprotein hemagglutinin (CDV-H) at the CDV surface utilizes signaling lymphocyte activation molecule (SLAM) and Nectin-4 (also called poliovirus-receptor-like-4; PVRL4) as entry receptors. Although fusion models have been proposed, the molecular mechanism of morbillivirus fusion entry is poorly understood. Here, we determined the crystal structure of the globular head domain of CDV-H vaccine strain at 3.2 Å resolution, revealing that CDV-H exhibits a highly tilted homodimeric form with a six-bladed β-propeller fold. While the predicted Nectin-4-binding site is well conserved with that of MeV-H, that of SLAM is similar but partially different, which is expected to contribute to host specificity. Five N-linked sugars covered a broad area of the CDV-H surface to expose receptor-binding sites only, supporting the effective production of neutralizing antibodies. These features are common to MeV-H, although the glycosylation sites are completely different. Furthermore, real-time observation using high-speed atomic force microscopy revealed highly mobile features of the CDV-H dimeric head via the connector region. These results suggest that sugar-shielded tilted homodimeric structure and dynamic conformational changes are common characteristics of morbilliviruses and ensure effective fusion entry and vaccination.
Collapse
Affiliation(s)
- Hideo Fukuhara
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
- Division of Pathogen Structure, Research Center for Zoonosis Control, Hokkaido UniversitySapporoJapan
| | - Kohei Yumoto
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Miyuki Sako
- Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Mizuho Kajikawa
- Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Toyoyuki Ose
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Mihiro Kawamura
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Mei Yoda
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Surui Chen
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Yuri Ito
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Shin Takeda
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Mwila Mwaba
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Jiaqi Wang
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Takao Hashiguchi
- Department of Virology, Faculty of Medicine, Kyushu UniversityFukuokaJapan
| | - Jun Kamishikiryo
- Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Nobuo Maita
- Institute for Enzyme Research, University of TokushimaTokushimaJapan
| | - Chihiro Kitatsuji
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Makoto Takeda
- Department of Microbiology, Graduate School of Medicine and Faculty of Medicine, The University of TokyoTokyoJapan
| | - Kimiko Kuroki
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
- Division of Pathogen Structure, Research Center for Zoonosis Control, Hokkaido UniversitySapporoJapan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido UniversitySapporoJapan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido UniversitySapporoJapan
- Core Research for Evolutional Science and Technology, Japan Science and Technology AgencySaitamaJapan
| |
Collapse
|
5
|
Pinchon E, Henry S, Leon F, Fournier-Wirth C, Foulongne V, Cantaloube JF. Rapid Detection of Measles Virus Using Reverse Transcriptase/Recombinase Polymerase Amplification Coupled with CRISPR/Cas12a and a Lateral Flow Detection: A Proof-of-Concept Study. Diagnostics (Basel) 2024; 14:517. [PMID: 38472989 DOI: 10.3390/diagnostics14050517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The measles virus is highly contagious, and efforts to simplify its diagnosis are essential. A reverse transcriptase/recombinase polymerase amplification assay coupled with CRISPR/Cas12a and an immunochromatographic lateral flow detection (RT-RPA-CRISPR-LFD) was developed for the simple visual detection of measles virus. The assay was performed in less than 1 h at an optimal temperature of 42 °C. The detection limit of the assay was 31 copies of an RNA standard in the reaction tube. The diagnostic performances were evaluated on a panel of 27 measles virus RT-PCR-positive samples alongside 29 measles virus negative saliva samples. The sensitivity and specificity were 96% (95% CI, 81-99%) and 100% (95% CI, 88-100%), respectively, corresponding to an accuracy of 98% (95% CI, 94-100%; p < 0.0001). This method will open new perspectives in the development of the point-of-care testing diagnosis of measles.
Collapse
Affiliation(s)
- Elena Pinchon
- Pathogénèse et Contrôle des Infections Chroniques et Emergentes, Etablissement Français du Sang, Université de Montpellier, Inserm, 34184 Montpellier, France
| | - Steven Henry
- Pathogénèse et Contrôle des Infections Chroniques et Emergentes, Etablissement Français du Sang, Université de Montpellier, Inserm, 34184 Montpellier, France
| | - Fanny Leon
- Pathogénèse et Contrôle des Infections Chroniques et Emergentes, Etablissement Français du Sang, Université de Montpellier, Inserm, 34184 Montpellier, France
| | - Chantal Fournier-Wirth
- Pathogénèse et Contrôle des Infections Chroniques et Emergentes, Etablissement Français du Sang, Université de Montpellier, Inserm, 34184 Montpellier, France
| | - Vincent Foulongne
- Pathogénèse et Contrôle des Infections Chroniques et Emergentes, Etablissement Français du Sang, Université de Montpellier, Inserm, 34184 Montpellier, France
| | - Jean-François Cantaloube
- Pathogénèse et Contrôle des Infections Chroniques et Emergentes, Etablissement Français du Sang, Université de Montpellier, Inserm, 34184 Montpellier, France
| |
Collapse
|
6
|
Zinzula L, Scholz J, Nagy I, Di Guardo G, Orsini M. Biophysical characterization of the cetacean morbillivirus haemagglutinin glycoprotein. Virus Res 2023; 336:199231. [PMID: 37769814 PMCID: PMC10550842 DOI: 10.1016/j.virusres.2023.199231] [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: 08/21/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023]
Abstract
Cetacean morbillivirus (CeMV) is an enveloped, non-segmented, negative-stranded RNA virus that infects marine mammals, spreading across species and causing lethal disease outbreaks worldwide. Among the eight proteins encoded by the CeMV genome, the haemagglutinin (H) glycoprotein is responsible for the virus attachment to host cell receptors. CeMV H represents an attractive target for antiviral and diagnostic research, yet the elucidation of the molecular mechanisms underlying its role in infection and inter-species transmission was hampered thus far due to the unavailability of recombinant versions of the protein. Here we present the cloning, expression and purification of a recombinant CeMV H ectodomain (rH-ecto), providing an initial characterization of its biophysical and structural properties. Sodium dodecyl sulphate - polyacrylamide gel electrophoresis (PAGE) combined to Western blot analysis and periodic acid Schiff assay showed that CeMV rH-ecto is purifiable at homogeneity from insect cells as a secreted, soluble and glycosylated protein. Miniaturized differential scanning fluorimetry, Blue Native PAGE and size exclusion chromatography coupled to multiangle light scattering revealed that CeMV rH-ecto is globularly folded, thermally stable and exists in solution in the oligomeric states of dimer and multiple of dimers. Furthermore, negative stain electron microscopy single particle analysis allowed us to delineate a low-resolution molecular architecture of the CeMV rH-ecto dimer, which recapitulates native assemblies from other morbilliviral H proteins, such as those from measles virus and canine distemper virus. This set of experiments by orthogonal techniques validates the CeMV rH-ecto as an experimental model for future biochemical studies on its structure and functions.
Collapse
Affiliation(s)
- Luca Zinzula
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany; Centro di Educazione Ambientale e alla Sostenibilità (CEAS) Laguna di Nora, Pula, Italy.
| | - Judith Scholz
- Core Facility, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - István Nagy
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany; Center of Research and Development, Eszterházy Károly Catholic University, Eger, Hungary
| | - Giovanni Di Guardo
- Retired Professor of General Pathology and Veterinary Pathophysiology, Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Massimiliano Orsini
- Laboratory of Microbial Ecology and Genomics, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| |
Collapse
|
7
|
Pennisi MG, Belák S, Tasker S, Addie DD, Boucraut-Baralon C, Egberink H, Frymus T, Hartmann K, Hofmann-Lehmann R, Lloret A, Marsilio F, Thiry E, Truyen U, Möstl K, Hosie MJ. Feline Morbillivirus: Clinical Relevance of a Widespread Endemic Viral Infection of Cats. Viruses 2023; 15:2087. [PMID: 37896864 PMCID: PMC10611265 DOI: 10.3390/v15102087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Feline morbillivirus (FeMV) was first isolated in 2012 from stray cats in Hong Kong. It has been found in association with tubulointerstitial nephritis (TIN), the most common cause of feline chronic kidney disease (CKD). However, viral host spectrum and virus tropism go beyond the domestic cat and kidney tissues. The viral genetic diversity of FeMV is extensive, but it is not known if this is clinically relevant. Urine and kidney tissues have been widely tested in attempts to confirm associations between FeMV infection and renal disease, but samples from both healthy and sick cats can test positive and some cross-sectional studies have not found associations between FeMV infection and CKD. There is also evidence for acute kidney injury following infection with FeMV. The results of prevalence studies differ greatly depending on the population tested and methodologies used for detection, but worldwide distribution of FeMV has been shown. Experimental studies have confirmed previous field observations that higher viral loads are present in the urine compared to other tissues, and renal TIN lesions associated with FeMV antigen have been demonstrated, alongside virus lymphotropism and viraemia-associated lymphopenia. Longitudinal field studies have revealed persistent viral shedding in urine, although infection can be cleared spontaneously.
Collapse
Affiliation(s)
| | - Sándor Belák
- Department of Biomedical Sciences and Veterinary Public Health (BVF), Swedish University of Agricultural Sciences (SLU), P.O. Box 7036, 750 07 Uppsala, Sweden;
| | - Séverine Tasker
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU, UK;
- Linnaeus Veterinary Limited, Shirley, Solihull B90 4BN, UK
| | - Diane D. Addie
- Independent Researcher, 64000 Pyrénées Aquitaine, France;
| | | | - Herman Egberink
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, University of Utrecht, 3584 CL Utrecht, The Netherlands;
| | - Tadeusz Frymus
- Department of Small Animal Diseases with Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGWW, 02-787 Warsaw, Poland;
| | - Katrin Hartmann
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, 80539 Munich, Germany;
| | - Regina Hofmann-Lehmann
- Clinical Laboratory, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland;
| | - Albert Lloret
- Fundació Hospital Clínic Veterinari, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
| | - Fulvio Marsilio
- Faculty of Veterinary Medicine, Università degli Studi di Teramo, 64100 Teramo, Italy;
| | - Etienne Thiry
- Veterinary Virology and Animal Viral Diseases, Department of Infectious and Parasitic Diseases, FARAH Research Centre, Faculty of Veterinary Medicine, Liège University, B-4000 Liège, Belgium;
| | - Uwe Truyen
- Institute of Animal Hygiene and Veterinary Public Health, University of Leipzig, 04103 Leipzig, Germany;
| | - Karin Möstl
- Institute of Virology, Department for Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Margaret J. Hosie
- MRC-University of Glasgow Centre for Virus Research, Garscube Estate, Glasgow G61 1QH, UK;
| |
Collapse
|
8
|
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: 0.5] [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
|
9
|
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
|
10
|
Haas GD, Lee B. Paramyxoviruses from bats: changes in receptor specificity and their role in host adaptation. Curr Opin Virol 2023; 58:101292. [PMID: 36508860 PMCID: PMC9974588 DOI: 10.1016/j.coviro.2022.101292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
Global metagenomic surveys have revealed that bats host a diverse array of paramyxoviruses, including species from at least five major genera. An essential determinant of successful spillover is the entry of a virus into a new host. We evaluate the role of receptor usage in the zoonotic potential of bat-borne henipaviruses, morbilliviruses, pararubulaviruses, orthorubulaviruses, and jeilongviruses; successful spillover into humans depends upon compatibility of a respective viral attachment protein with its cognate receptor. We also emphasize the importance of postentry restrictions in preventing spillover. Metagenomics and characterization of newly identified paramyxoviruses have greatly improved our understanding of spillover determinants, allowing for better forecasts of which bat-borne viruses may pose the greatest risk for cross-species transmission into humans.
Collapse
Affiliation(s)
- Griffin D Haas
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.
| |
Collapse
|
11
|
Chen Y, Wang T, Yang Y, Fang Y, Zhao B, Zeng W, Lv D, Zhang L, Zhang Y, Xue Q, Chen X, Wang J, Qi X. Extracellular vesicles derived from PPRV-infected cells enhance signaling lymphocyte activation molecular (SLAM) receptor expression and facilitate virus infection. PLoS Pathog 2022; 18:e1010759. [PMID: 36084159 PMCID: PMC9491601 DOI: 10.1371/journal.ppat.1010759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 09/21/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. PPRV is lymphotropic in nature and SLAM was identified as the primary receptor for PPRV and other Morbilliviruses. Many viruses have been demonstrated to engage extracellular vesicles (EVs) to facilitate their replication and pathogenesis. Here, we provide evidence that PPRV infection significantly induced the secretion levels of EVs from goat PBMC, and that PPRV-H protein carried in EVs can enhance SLAM receptor expression in the recipient cells via suppressing miR-218, a negative miRNA directly targeting SLAM gene. Importantly, EVs-mediated increased SLAM expression enhances PPRV infectivity as well as the expression of various cytokines related to SLAM signaling pathway in the recipient cells. Moreover, our data reveal that PPRV associate EVs rapidly entry into the recipient cells mainly through macropinocytosis pathway and cooperated with caveolin- and clathrin-mediated endocytosis. Taken together, our findings identify a new strategy by PPRV to enhance virus infection and escape innate immunity by engaging EVs pathway. Peste des petitsruminants virus (PPRV) infection induces a transient but severe immunosuppression in the host, which threatens both small livestock and endangered susceptible wildlife populations in many countries. Despite extensive research, the mechanism underlying pathogenesis of PPRV infection remains elusive. Our data provide the first direct evidence that the EVs derived from PPRV-infected cells are involved in PPRV replication. In this study, the EVs derived from PPRV-infected goat PBMCs can enhance SLAM expression in the recipient cells, and more importantly, EVs-mediated increased SLAM expression enhances PPRV replication as well as the expression of various cytokines related to SLAM signaling pathway in the recipient cells. Taken together, our research has provided new insight into understanding the effect of EVs on PPRV replication and pathogenesis, and revealed a potential therapeutic target for antiviral intervention.
Collapse
Affiliation(s)
- Yan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ting Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yang Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Fang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Bao Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Shaanxi Animal Disease Control Center, Xi’an, China
| | - Wei Zeng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Daiyue Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Leyan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, China
| | - Xiwen Chen
- Animal Disease Prevention and Control & Healthy Breeding Engineering Technology Research Center, Mianyang Normal University, Mianyang, Sichuan, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (JW); (XQ)
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (JW); (XQ)
| |
Collapse
|
12
|
Echeverry-Bonilla DF, Buriticá-Gaviria EF, Orjuela-Acosta D, Chinchilla-Cardenas DJ, Ruiz-Saenz J. The First Report and Phylogenetic Analysis of Canine Distemper Virus in Cerdocyon thous from Colombia. Viruses 2022; 14:v14091947. [PMID: 36146754 PMCID: PMC9502595 DOI: 10.3390/v14091947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Canine distemper virus (CDV) is the etiological agent of a highly prevalent viral infectious disease of domestic and wild carnivores. This virus poses a conservation threat to endangered species worldwide due to its ability to jump between multiple species and produce a disease, which is most often fatal. Although CDV infection has been regularly diagnosed in Colombian wildlife, to date the molecular identity of circulating CDV lineages is currently unknown. Our aim was to evaluate the presence and phylogenetic characterization of CDV detected in samples from naturally infected Cerdocyon thous from Colombia. We sequenced for the first time the CDV infecting wildlife in Colombia and demonstrated the presence of South America/North America-4 Lineage with a higher relationship to sequences previously reported from domestic and wild fauna belonging to the United States of America. Our results are crucial for the understanding of the interspecies transmission of CDV in the domestic/wild interface and for the prevention and control of such an important multi-host pathogen.
Collapse
Affiliation(s)
- Diego Fernando Echeverry-Bonilla
- Hospital Veterinario, Universidad del Tolima, Calle 20 Sur # 23A-160 Barrio Miramar, Ibagué 730010, Tolima, Colombia
- Grupo de Investigación en Medicina y Cirugía de Pequeños Animales, Facultad de Medicina Veterinaria y Zootecnia, Universidad del Tolima, Calle 20 Sur # 23A-160 Barrio Miramar, Ibagué 730010, Tolima, Colombia
| | - Edwin Fernando Buriticá-Gaviria
- Hospital Veterinario, Universidad del Tolima, Calle 20 Sur # 23A-160 Barrio Miramar, Ibagué 730010, Tolima, Colombia
- Grupo de Investigación en Medicina y Cirugía de Pequeños Animales, Facultad de Medicina Veterinaria y Zootecnia, Universidad del Tolima, Calle 20 Sur # 23A-160 Barrio Miramar, Ibagué 730010, Tolima, Colombia
| | - Delio Orjuela-Acosta
- Hospital Veterinario, Universidad del Tolima, Calle 20 Sur # 23A-160 Barrio Miramar, Ibagué 730010, Tolima, Colombia
| | - Danny Jaír Chinchilla-Cardenas
- Mascolab, Laboratorio de Biología Molecular, Calle 49 Sur # 45ª-300, Oficina 1202, Centro Empresarial S48 Tower, Envigado 055422, Antioquia, Colombia
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680002, Colombia
- Correspondence:
| |
Collapse
|
13
|
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: 12] [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
|
14
|
Du X, Goffin E, Gillard L, Machiels B, Gillet L. A Single Oral Immunization with Replication-Competent Adenovirus-Vectored Vaccine Induces a Neutralizing Antibody Response in Mice against Canine Distemper Virus. Viruses 2022; 14:1847. [PMID: 36146652 PMCID: PMC9501072 DOI: 10.3390/v14091847] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 12/02/2022] Open
Abstract
Canine Distemper Virus (CDV) is a fatal and highly contagious pathogen of multiple carnivores. While injectable vaccines are very effective in protecting domestic animals, their use in the wild is unrealistic. Alternative vaccines are therefore needed. Adenovirus (AdV) vectors are popular vaccine vectors due to their capacity to elicit potent humoral and cellular immune responses against the antigens they carry. In parallel, vaccines based on live human AdV-4 and -7 have been used in U.S. army for several decades as replicative oral vaccines against respiratory infection with the same viruses. Based on these observations, the use of oral administration of replication competent AdV-vectored vaccines has emerged as a promising tool especially for wildlife vaccination. Developing this type of vaccine is not easy, however, given the high host specificity of AdVs and their very low replication in non-target species. To overcome this problem, the feasibility of this approach was tested using mouse adenovirus 1 (MAV-1) in mice as vaccine vectors. First, different vaccine vectors expressing the entire or part H or F proteins of CDV were constructed. These different strains were then used as oral vaccines in BALB/c mice and the immune response to CDV was evaluated. Only the strain expressing the full length CDV H protein generated a detectable and neutralizing immune response to CDV. Secondly, using this strain, we were able to show that although this type of vaccine is sensitive to pre-existing immunity to the vector, a second oral administration of the same vaccine is able to boost the immune response against CDV. Overall, this study demonstrates the feasibility of using replicating AdVs as oral vaccine vectors to immunize against CDV in wildlife carnivores.
Collapse
Affiliation(s)
| | | | | | | | - Laurent Gillet
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, 4000 Liège, Belgium
| |
Collapse
|
15
|
Zhang W, Weng J, Yao L, Jia P, Yi M, Jia K. Nectin4 antagonises type I interferon production by targeting TRAF3 for autophagic degradation and disrupting TRAF3-TBK1 complex formation. Int J Biol Macromol 2022; 218:654-664. [PMID: 35878672 DOI: 10.1016/j.ijbiomac.2022.07.151] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022]
Abstract
Autophagy, a conserved cellular degradative process, plays a crucial role in innate immunity during viral infections. Nervous necrosis virus (NNV), a leading cause of fish diseases with morbidity and mortality, triggers cell autophagy to promote viral replication; however, the details of how NNV utilises autophagy to facilitate its own replication remain largely unexplored. Here, we investigated the mechanism by which the sea perch Nectin4 (LjNectin4), a receptor of NNV, regulates autophagy and the innate immune system by targeting TNFR-associated factor 3 (TRAF3). Our data demonstrated that LjNectin4 directly binds to the NNV capsid protein and facilitates NNV entry, indicating that LjNectin4 functions as an NNV receptor. Moreover, LjNectin4 promoted NNV replication by inhibiting key elements of the RLR signalling pathway (MDA5, MAVS, TRAF3, TBK1, and IRF3)-induced IFN response. Mechanistically, LjNectin4 directly interacted with TRAF3 and promoted its autophagy-mediated lysosomal degradation. Domain mapping of the interaction between TRAF3 and LjNectin4 or TBK1 showed that both LjNectin4 and TBK1 interacted with the ZF2 and TRAF-C domains of TRAF3, suggesting that LjNectin4 blocked TRAF3-TBK1 complex formation. Collectively, our study revealed that NNV utilises LjNectin4 to suppress IFN production by mediating TRAF3 autophagic degradation and disrupting the TRAF3-TBK1 complex, thereby promoting NNV replication.
Collapse
Affiliation(s)
- Wanwan Zhang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510000, China; Pearl River Estuary Marine Ecosystem Research Station, Ministry of Education, Zhuhai 519000, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Juehua Weng
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510000, China; Pearl River Estuary Marine Ecosystem Research Station, Ministry of Education, Zhuhai 519000, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Lan Yao
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510000, China; Pearl River Estuary Marine Ecosystem Research Station, Ministry of Education, Zhuhai 519000, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Peng Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510000, China; Fuzhou Medical University, Jiangxi, Fuzhou 344000, China
| | - Meisheng Yi
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510000, China; Pearl River Estuary Marine Ecosystem Research Station, Ministry of Education, Zhuhai 519000, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Kuntong Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510000, China; Pearl River Estuary Marine Ecosystem Research Station, Ministry of Education, Zhuhai 519000, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China.
| |
Collapse
|
16
|
Kimpston CN, Hatke AL, Castelli B, Otto N, Tiffin HS, Machtinger ET, Brown JD, Van Why KR, Marconi RT. High Prevalence of Antibodies against Canine Parvovirus and Canine Distemper Virus among Coyotes and Foxes from Pennsylvania: Implications for the Intersection of Companion Animals and Wildlife. Microbiol Spectr 2022; 10:e0253221. [PMID: 35080421 PMCID: PMC8791182 DOI: 10.1128/spectrum.02532-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 11/20/2022] Open
Abstract
Canine distemper virus (CDV) and Canine parvovirus (CPV) can cause deadly infections in wildlife and companion animals. In this report, we screened serum from free-ranging eastern coyotes (Canis latrans; N = 268), red foxes (Vulpes vulpes; N = 63), and gray foxes (Urocyon cinereoargenteus; N = 16) from Pennsylvania, USA, for antibodies (Abs) to CDV and CPV. This comprehensive screening was achieved using a commercially available enzyme-linked immunosorbent assay (ELISA)-based colorimetric assay. Abs to CDV and CPV were detected in 25.4% and 45.5% of coyotes, 36.5% and 52.4% of red foxes, and 12.5% and 68.8% of gray foxes, respectively. Abs to both viruses were detected in 9.7% of coyotes, 19.1% of red foxes, and 12.5% of gray foxes. This study demonstrates significant wildlife exposure in a northeastern state to CDV and CPV. As wildlife species continue to urbanize, the probability of spillover between domestic animals and wildlife will increase. Ongoing surveillance of wildlife for CDV and CPV exposure is warranted. IMPORTANCECanine distemper virus (CDV) and Canine parvovirus (CPV) are significant health threats to domestic dogs (Canis familiaris) and wildlife. CDV and CPV have been identified in diverse vertebrates, including endangered wildlife species. Susceptibility to these viral pathogens varies significantly among geographic regions and between host species. High morbidity and mortality have been reported with infection by either virus in susceptible species, including dogs. As humans and companion animals encroach on wildlife habitat, and as wildlife becomes increasingly urbanized, the potential for transmission between species increases. This study assessed CPV and CDV Ab prevalence in wild canids (eastern coyotes, red foxes, and gray foxes) harvested in Pennsylvania between 2015 and 2020. High Ab prevalence was demonstrated for both viruses in each species. Ongoing monitoring of CPV and CDV in wildlife and increased efforts to vaccinate dogs and prevent spillover events are essential.
Collapse
Affiliation(s)
- Caellaigh N. Kimpston
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States
| | - Amanda L. Hatke
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States
| | - Benjamin Castelli
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States
| | - Nathan Otto
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States
| | - Hannah S. Tiffin
- Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States
| | - Erika T. Machtinger
- Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States
| | - Justin D. Brown
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States
| | - Kyle R. Van Why
- USDA-APHIS Wildlife Services, Harrisburg, Pennsylvania, United States
| | - Richard T. Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States
| |
Collapse
|
17
|
Wang B, Li S, Qiao Y, Fu Y, Nie J, Jiang S, Yao X, Pan Y, Zhao L, Wu C, Shi Y, Yin Y, Shan Y. Self-assembling ferritin nanoparticles coupled with linear sequences from canine distemper virus haemagglutinin protein elicit robust immune responses. J Nanobiotechnology 2022; 20:32. [PMID: 35012571 PMCID: PMC8744384 DOI: 10.1186/s12951-021-01229-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/24/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Canine distemper virus (CDV), which is highly infectious, has caused outbreaks of varying scales in domestic and wild animals worldwide, so the development of a high-efficiency vaccine has broad application prospects. Currently, the commercial vaccine of CDV is an attenuated vaccine, which has the disadvantages of a complex preparation process, high cost and safety risk. It is necessary to develop a safe and effective CDV vaccine that is easy to produce on a large scale. In this study, sequences of CDV haemagglutinin (HA) from the Yanaka strain were aligned, and three potential linear sequences, termed YaH3, YaH4, and YaH5, were collected. To increase the immunogenicity of the epitopes, ferritin was employed as a self-assembling nanoparticle element. The ferritin-coupled forms were termed YaH3F, YaH4F, and YaH5F, respectively. A full-length HA sequence coupled with ferritin was also constructed as a DNA vaccine to compare the immunogenicity of nanoparticles in prokaryotic expression. RESULT The self-assembly morphology of the proteins from prokaryotic expression was verified by transmission electron microscopy. All the proteins self-assembled into nanoparticles. The expression of the DNA vaccine YaHF in HEK-293T cells was also confirmed in vitro. After subcutaneous injection of epitope nanoparticles or intramuscular injection of DNA YaHF, all vaccines induced strong serum titres, and long-term potency of antibodies in serum could be detected after 84 days. Strong anti-CDV neutralizing activities were observed in both the YaH4F group and YaHF group. According to antibody typing and cytokine detection, YaH4F can induce both Th1 and Th2 immune responses. The results of flow cytometry detection indicated that compared with the control group, all the immunogens elicited an increase in CD3. Simultaneously, the serum antibodies induced by YaH4F and YaHF could significantly enhance the ADCC effect compared with the control group, indicating that the antibodies in the serum effectively recognized the antigens on the cell surface and induced NK cells to kill infected cells directly. CONCLUSIONS YaH4F self-assembling nanoparticle obtained by prokaryotic expression has no less of an immune effect than YaHF, and H4 has great potential to become a key target for the easy and rapid preparation of epitope vaccines.
Collapse
Affiliation(s)
- Bo Wang
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China
| | - Shuang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yongbo Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yu Fu
- Changchun Xinuo BioTechnology Co., Ltd, Changchun, 130015, Jilin, China
| | - Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Shun Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yi Pan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Linye Zhao
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China
| | - Congmei Wu
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yuhe Yin
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China.
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China. .,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China.
| |
Collapse
|
18
|
Bitencourt MM, Bezerra AMR. Infection agents of Didelphidae (Didelphimorphia) of Brazil: an underestimated matter in zoonoses research. MAMMALIA 2021. [DOI: 10.1515/mammalia-2021-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Zoonoses are diseases or infections naturally transmissible from vertebrate animals to humans, and can be bacterial, viral or parasitic. The growth of urbanization, industrialization and the advance of agriculture and livestock facilitate the spread of infectious and parasitic agents from wild animals to the human population and to their domestic animals. Among the various reservoirs of zoonotic agents, we find that didelphid species, due to their high capacity for adaptation in urban environments, as an important study case. We reviewed the literature data on the pathogens, including with zoonotic potential of marsupial species occurring in Brazil, accounted for infections by agents that we categorized into Bacteria, Viruses, Protozoa, and Helminths. Aiming identifies possible knowledge gaps, we also surveyed the origin of studied samples and the institutions leading the researches on host didelphids. Among the hosts, the genus Didelphis in the cycles of these agents stands out. Moreover, we found that the majority of reported cases are in the Southeastern Brazil, mean the data from other Brazilian localities and didelphid species could be underestimated. Most studies took place in graduate programs of public research institutions, emphasizing the importance of the funding public research for the Brazilian scientific development.
Collapse
Affiliation(s)
- Matheus M. Bitencourt
- Programa de Pós-Graduação em Zoologia, Instituto de Ciências Biológicas, Universidade Federal do Pará , Rua Augusto Corrêa, 01, CEP 66075-110 , Belém , PA , Brazil
| | - Alexandra M. R. Bezerra
- Mastozoologia/COZOO, Museu Paraense Emilio Goeldi, Campus de Pesquisa , Av. Perimetral 1901, CEP 66077-830 , Belém , PA , Brazil
| |
Collapse
|
19
|
Zinzula L, Mazzariol S, Di Guardo G. Molecular signatures in cetacean morbillivirus and host species proteomes: Unveiling the evolutionary dynamics of an enigmatic pathogen? Microbiol Immunol 2021; 66:52-58. [PMID: 34779039 DOI: 10.1111/1348-0421.12949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 02/01/2023]
Abstract
Cetacean morbillivirus (CeMV) infects marine mammals often causing a fatal respiratory and neurological disease. Recently, CeMV has expanded its geographic and host species range, with cases being reported worldwide among dolphins, whales, seals, and other aquatic mammalian species, and therefore has emerged as the most threatening nonanthropogenic factor affecting marine mammal's health and conservation. Extensive research efforts have aimed to understand CeMV epidemiology and ecology, however, the molecular mechanisms underlying its transmission and pathogenesis are still poorly understood. In particular, the field suffers from a knowledge gap on the structural and functional properties of CeMV proteins and their host interactors. Nevertheless, the body of scientific literature produced in recent years has inaugurated new investigational trends, driving future directions in CeMV molecular research. In this mini-review, the most recent literature has been summarized in the context of such research trends, and categorized into four priority research topics, such as (1) the interaction between CeMV glycoprotein and its host cell receptors across several species; (2) the CeMV molecular determinants responsible for different disease phenotype; (3) the host molecular determinants responsible for differential susceptibility to CeMV infection; (4) the CeMV molecular determinants responsible for difference virulence among circulating CeMV strains. Arguably, these are the most urgent topics that need to be investigated and that most promisingly will help to shed light on the details of CeMV evolutionary dynamics in the immediate future.
Collapse
Affiliation(s)
- Luca Zinzula
- Department of Molecular Structural Biology, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Sandro Mazzariol
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro (Padova), Italy
| | | |
Collapse
|
20
|
Canine Morbillivirus from Colombian Lineage Exhibits In Silico and In Vitro Potential to Infect Human Cells. Pathogens 2021; 10:pathogens10091199. [PMID: 34578231 PMCID: PMC8471232 DOI: 10.3390/pathogens10091199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
Canine morbillivirus (CDV) is a viral agent that infects domestic dogs and a vast array of wildlife species. It belongs to the Paramyxoviridae family, genus Morbillivirus, which is shared with the Measles virus (MeV). Both viruses employ orthologous cellular receptors, SLAM in mononuclear cells and Nectin-4 in epithelial cells, to enter the cells. Although CDV and MeV hemagglutinin (H) have similar functions in viral pathogenesis and cell tropism, the potential interaction of CDV-H protein with human cellular receptors is still uncertain. Considering that CDV is classified as a multi-host pathogen, the potential risk of CDV transmission to humans has not been fully discarded. In this study, we aimed to evaluate both in silico and in vitro, whether there is a cross-species transmission potential from CDV to humans. To accomplish this, the CDV-H protein belonging to the Colombian lineage was modelled. After model validations, molecular docking and molecular dynamics simulations were carried out between Colombian CDV-H protein and canine and human cellular receptors to determine different aspects of the protein-protein interactions. Moreover, cell lines expressing orthologous cellular receptors, with both reference and wild-type CDV strains, were conducted to determine the CDV cross-species transmission potential from an in vitro model. This in silico and in vitro approach suggests the possibility that CDV interacts with ortholog human SLAM (hSLAM) and human Nectin-4 receptors to infect human cell lines, which could imply a potential cross-species transmission of CDV from dogs to humans.
Collapse
|
21
|
Piewbang C, Wardhani SW, Dankaona W, Yostawonkul J, Boonrungsiman S, Surachetpong W, Kasantikul T, Techangamsuwan S. Feline morbillivirus-1 in dogs with respiratory diseases. Transbound Emerg Dis 2021; 69:e175-e184. [PMID: 34355534 DOI: 10.1111/tbed.14278] [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: 05/27/2021] [Revised: 06/29/2021] [Accepted: 08/04/2021] [Indexed: 01/04/2023]
Abstract
Feline morbillivirus-1 (FeMV-1) is a viral pathogen associated with kidney disease in domestic cats and wild felids. We initially identified the FeMV-1 from the lung of a necropsied dog with severe pulmonary disease by the reverse transcription polymerase chain reaction (RT-PCR). Thereafter, we investigated FeMV-1 in nasal and oral swab samples from 73 healthy and 113 dogs with respiratory illnesses. We found polymerase chain reaction (PCR)-positive FeMV-1 from only 14/113 (12.39%) dogs with respiratory disease (p = .001). Of these 14 dogs, six were co-infected with other canine respiratory viruses (6/14; 42.86%). Two independent immunohistochemistry procedures, using antibodies against matrix and phosphoprotein of FeMV-1, confirmed the presence of FeMV-1 in lung tissues of two necropsied dogs (out of a total of 22 dogs, 9.09%) that died from respiratory disease. This finding corresponded to transmission electron microscopy findings that paramyxoviral particles exist in lung epithelia. FeMV-1 antigen localization was also evident in the kidney, lymphoid and brain tissues of two deceased dogs. FeMV-1 was successfully isolated from a necropsied dog and from two living dogs, all with respiratory illnesses, which supports FeMV infection in dogs. The detection of FeMV-1 in dog tissues expands the known tropism of this virus to a non-felid host. Our findings indicate that FeMV-1, alone or in co-infection with other viral pathogens, might contribute to respiratory illness and death in dogs.
Collapse
Affiliation(s)
- Chutchai Piewbang
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Animal Virome and Diagnostic Development Research Group, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sabrina Wahyu Wardhani
- Animal Virome and Diagnostic Development Research Group, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,The International Graduate Program of Veterinary Science and Technology (VST), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Wichan Dankaona
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Animal Virome and Diagnostic Development Research Group, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Jakarwan Yostawonkul
- The International Graduate Program of Veterinary Science and Technology (VST), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Suwimon Boonrungsiman
- National Nanotechnology Center, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Win Surachetpong
- Department of Veterinary Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Tanit Kasantikul
- Clemson Veterinary Diagnostic Center, Clemson University, Columbia, South Carolina, USA
| | - Somporn Techangamsuwan
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Animal Virome and Diagnostic Development Research Group, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
22
|
Computational Analysis Reveals a Critical Point Mutation in the N-Terminal Region of the Signaling Lymphocytic Activation Molecule Responsible for the Cross-Species Infection with Canine Distemper Virus. Molecules 2021; 26:molecules26051262. [PMID: 33652764 PMCID: PMC7956568 DOI: 10.3390/molecules26051262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Infection of hosts by morbilliviruses is facilitated by the interaction between viral hemagglutinin (H-protein) and the signaling lymphocytic activation molecule (SLAM). Recently, the functional importance of the n-terminal region of human SLAM as a measles virus receptor was demonstrated. However, the functional roles of this region in the infection process by other morbilliviruses and host range determination remain unknown, partly because this region is highly flexible, which has hampered accurate structure determination of this region by X-ray crystallography. In this study, we analyzed the interaction between the H-protein from canine distemper virus (CDV-H) and SLAMs by a computational chemistry approach. Molecular dynamics simulations and fragment molecular orbital analysis demonstrated that the unique His28 in the N-terminal region of SLAM from Macaca is a key determinant that enables the formation of a stable interaction with CDV-H, providing a basis for CDV infection in Macaca. The computational chemistry approach presented should enable the determination of molecular interactions involving regions of proteins that are difficult to predict from crystal structures because of their high flexibility.
Collapse
|
23
|
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
|
24
|
Di Francesco CE, Smoglica C, Angelucci S. Infectious Diseases and Wildlife Conservation Medicine: The Case of the Canine Distemper in European Wolf Population. Animals (Basel) 2020; 10:ani10122426. [PMID: 33352915 PMCID: PMC7766025 DOI: 10.3390/ani10122426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/27/2022] Open
Abstract
Canine distemper is a contagious infectious disease, caused by canine distemper virus (CDV) belonging to Morbillivirus genus, Paramyxoviridae family, representing a serious threat for domestic and wild carnivores [...]
Collapse
Affiliation(s)
- Cristina E. Di Francesco
- Faculty of Veterinary Medicine, University of Teramo, Loc. Piano D’Accio, 64110 Teramo, Italy; (C.S.); (S.A.)
- Correspondence:
| | - Camilla Smoglica
- Faculty of Veterinary Medicine, University of Teramo, Loc. Piano D’Accio, 64110 Teramo, Italy; (C.S.); (S.A.)
| | - Simone Angelucci
- Faculty of Veterinary Medicine, University of Teramo, Loc. Piano D’Accio, 64110 Teramo, Italy; (C.S.); (S.A.)
- Majella National Park, Caramanico Terme, 65023 Pescara, Italy
| |
Collapse
|
25
|
|
26
|
Seki F, Yamamoto Y, Fukuhara H, Ohishi K, Maruyama T, Maenaka K, Tokiwa H, Takeda M. Measles Virus Hemagglutinin Protein Establishes a Specific Interaction With the Extreme N-Terminal Region of Human Signaling Lymphocytic Activation Molecule to Enhance Infection. Front Microbiol 2020; 11:1830. [PMID: 32922371 PMCID: PMC7457132 DOI: 10.3389/fmicb.2020.01830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/13/2020] [Indexed: 11/26/2022] Open
Abstract
Measles virus (MV) is a human pathogen that is classified in the genus Morbillivirus in the family Paramyxoviridae together with several non-human animal morbilliviruses. They cause severe systemic infections by using signaling lymphocytic activation molecule (SLAM) and poliovirus receptor-like 4 expressed on immune and epithelial cells, respectively, as receptors. The viral hemagglutinin (H) protein is responsible for the receptor-binding. Previously determined structures of MV-H and SLAM complexes revealed a major binding interface between the SLAM V domain and MV-H with four binding components (sites 1–4) in the interface. We studied the MV-H and human SLAM (hSLAM) complex structure in further detail by in silico analyses and determined missing regions or residues in the previously determined complex structures. These analyses showed that, in addition to sites 1–4, MV-H establishes a unique interaction with the extreme N-terminal region (ExNTR) of hSLAM. The first principles calculation-based fragment molecular orbital computation method revealed that methionine at position 29 (hSLAM-Met29) is the key residue for the interaction. hSLAM-Met29 was predicted to establish a CH-π interaction with phenylalanine at position 549 of MV-H (MVH-Phe549). A cell-cell fusion assay showed that the hSLAM-Met29 and MVH-Phe549 interaction is important for hSLAM-dependent MV membrane fusion. Furthermore, Jurkat cell lines expressing hSLAM with or without Met29 and recombinant MV possessing the H protein with or without Phe549 showed that the hSLAM-Met29 and MVH-Phe549 interaction enhanced hSLAM-dependent MV infection by ~10-fold. We speculate that in the evolutionary history of morbilliviruses, this interaction may have contributed to MV adaptation to humans because this interaction is unique for MV and only MV uses hSLAM efficiently among morbilliviruses.
Collapse
Affiliation(s)
- Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Tokyo, Japan
| | - Hideo Fukuhara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Kazue Ohishi
- Faculty of Engineering, Tokyo Polytechnic University, Atsugi, Japan
| | | | - Katsumi Maenaka
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hiroaki Tokiwa
- Department of Chemistry, Rikkyo University, Tokyo, Japan
| | - Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|