1
|
Vigil K, Wu H, Aw TG. A systematic review on global zoonotic virus-associated mortality events in marine mammals. One Health 2024; 19:100872. [PMID: 39206255 PMCID: PMC11357810 DOI: 10.1016/j.onehlt.2024.100872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Marine mammals play a critical role as sentinels for tracking the spread of zoonotic diseases, with viruses being the primary causative factor behind infectious disease induced mortality events. A systematic review was conducted to document marine mammal mortality events attributed to zoonotic viral infections in published literature across the globe. This rigorous search strategy yielded 2883 studies with 88 meeting inclusion criteria. The studies spanned from 1989 to 2023, with a peak in publications observed in 2020. Most of the included studies were retrospective, providing valuable insights into historical trends. The United States (U.S.) reported the highest number of mortality events followed by Spain, Italy, Brazil and the United Kingdom. Harbor seals were the most impacted species, particularly in regions like Anholt, Denmark and the New England Coast, U.S. Analysis revealed six main viruses responsible for mortality events, with Morbillivirus causing the highest proportion of deaths. Notably, the occurrence of these viral events varied geographically, with distinct patterns observed in different regions. Immunohistochemistry emerged as the most employed detection method. This study underscores the importance of global surveillance efforts in understanding and mitigating the impact of viral infections on marine mammal populations, thereby emphasizing the necessity of collaborative One Health approaches to address emerging threats at the human-animal-environment interface. Additionally, the potential transfer of zoonotic viruses to aquatic organisms used in food production, such as fish and shellfish, highlights the broader implications for food safety, food security and public health.
Collapse
Affiliation(s)
- Katie Vigil
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Huiyun Wu
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Tiong Gim Aw
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| |
Collapse
|
2
|
Siering O, Langbein M, Herrmann M, Wittwer K, von Messling V, Sawatsky B, Pfaller CK. Genetic diversity accelerates canine distemper virus adaptation to ferrets. J Virol 2024; 98:e0065724. [PMID: 39007615 PMCID: PMC11334482 DOI: 10.1128/jvi.00657-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
RNA viruses adapt rapidly to new host environments by generating highly diverse genome sets, so-called "quasispecies." Minor genetic variants promote their rapid adaptation, allowing for the emergence of drug-resistance or immune-escape mutants. Understanding these adaptation processes is highly relevant to assessing the risk of cross-species transmission and the safety and efficacy of vaccines and antivirals. We hypothesized that genetic memory within a viral genome population facilitates rapid adaptation. To test this, we investigated the adaptation of the Morbillivirus canine distemper virus to ferrets and compared an attenuated, Vero cell-adapted virus isolate with its recombinant derivative over consecutive ferret passages. Although both viruses adapted to the new host, the reduced initial genetic diversity of the recombinant virus resulted in delayed disease onset. The non-recombinant virus gradually increased the frequencies of beneficial mutations already present at very low frequencies in the input virus. In contrast, the recombinant virus first evolved de novo mutations to compensate for the initial fitness impairments. Importantly, while both viruses evolved different sets of mutations, most mutations found in the adapted non-recombinant virus were identical to those found in a previous ferret adaptation experiment with the same isolate, indicating that mutations present at low frequency in the original virus stock serve as genetic memory. An arginine residue at position 519 in the carboxy terminus of the nucleoprotein shared by all adapted viruses was found to contribute to pathogenesis in ferrets. Our work illustrates the importance of genetic diversity for adaptation to new environments and identifies regions with functional relevance.IMPORTANCEWhen viruses encounter a new host, they can rapidly adapt to this host and cause disease. How these adaptation processes occur remains understudied. Morbilliviruses have high clinical and veterinary relevance and are attractive model systems to study these adaptation processes. The canine distemper virus is of particular interest, as it exhibits a broader host range than other morbilliviruses and frequently crosses species barriers. Here, we compared the adaptation of an attenuated virus and its recombinant derivative to that of ferrets. Pre-existing mutations present at low frequency allowed faster adaptation of the non-recombinant virus compared to the recombinant virus. We identified a common point mutation in the nucleoprotein that affected the pathogenesis of both viruses. Our study shows that genetic memory facilitates environmental adaptation and that erasing this genetic memory by genetic engineering results in delayed and different adaptation to new environments, providing an important safety aspect for the generation of live-attenuated vaccines.
Collapse
Affiliation(s)
- Oliver Siering
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Mareike Langbein
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Maike Herrmann
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Kevin Wittwer
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | | | - Bevan Sawatsky
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Christian K. Pfaller
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
3
|
Liang J, Wang T, Wang Q, Wang X, Fan X, Hu T, Leng X, Shi K, Li J, Gong Q, Du R. Prevalence of canine distemper in minks, foxes and raccoon dogs from 1983 to 2023 in Asia, North America, South America and Europe. Front Vet Sci 2024; 11:1394631. [PMID: 39193367 PMCID: PMC11348944 DOI: 10.3389/fvets.2024.1394631] [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: 03/01/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Canine distemper (CD) is a virulent disease caused by the canine distemper virus (CDV) in canines and mustelidaes with high mortality. The incidence of CDV is worldwide distribution and it has caused huge economic losses to multiple industries around the world. There are many studies investigating the prevalence of CD infection, but no comprehensive analysis of CDV infection in minks, foxes and raccoon dogs worldwide has therefore been carried out. The aim of this meta is to provide a comprehensive assessment of the prevalence of CDV infection in minks, foxes and raccoon dogs dogs through a meta-analysis of articles published from around the world. Data from 8,582 small carnivores in 12 countries were used to calculate the combined prevalence of CD. A total of 22.6% (1,937/8,582) of minks, foxes and raccoon dogs tested positive for CD. The prevalence was higher in Asia (13.8, 95% CI: 22.2-45.6), especially in South Korea (65.8, 95% CI: 83.3-95.8). Our study found that the incidence of CD was also associated with geographic climate, population size, health status, and breeding patterns. CD is more commonly transmitted in minks, foxes and raccoon dogs. However, the concentrated breeding as an economic animal has led to an increase in the prevalence rate. The difference analysis study recommended that countries develop appropriate preventive and control measures based on the prevalence in the minks, foxes, and raccoon dogs industries, and that reducing stocking density is important to reduce the incidence of CDV. In addition, CDV is more common in winter, so vaccination in winter should be strengthened and expanded to reduce the incidence of CD in minks, foxes and raccoon dogs.
Collapse
Affiliation(s)
- Jian Liang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Tingting Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Qi Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xiaolin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xinying Fan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Tingting Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xue Leng
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Kun Shi
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Jianming Li
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Qinglong Gong
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Rui Du
- Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin, China
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Srikrishna D. Pentagon Found Daily, Metagenomic Detection of Novel Bioaerosol Threats to Be Cost-Prohibitive: Can Virtualization and AI Make It Cost-Effective? Health Secur 2024; 22:108-129. [PMID: 38625036 DOI: 10.1089/hs.2023.0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
In 2022, the Pentagon Force Protection Agency found threat agnostic detection of novel bioaerosol threats to be "not feasible for daily operations" due to the cost of reagents used for metagenomics, cost of sequencing instruments, and cost of labor for subject matter experts to analyze bioinformatics. Similar operational difficulties might extend to many of the 280,000 buildings (totaling 2.3 billion square feet) at 5,000 secure US Department of Defense military sites, 250 Navy ships, as well as many civilian buildings. These economic barriers can still be addressed in a threat agnostic manner by dynamically pooling samples from dry filter units, called spike-triggered virtualization, whereby pooling and sequencing depth are automatically modulated based on novel biothreats in the sequencing output. By running at a high average pooling factor, the daily and annual cost per dry filter unit can be reduced by 10 to 100 times depending on the chosen trigger thresholds. Artificial intelligence can further enhance the sensitivity of spike-triggered virtualization. The risk of infection during the 12- to 24-hour window between a bioaerosol incident and its detection remains, but in some cases it can be reduced by 80% or more with high-speed indoor air cleaning exceeding 12 air changes per hour, which is similar to the rate of air cleaning in passenger airplanes in flight. That level of air changes per hour or higher is likely to be cost-prohibitive using central heating ventilation and air conditioning systems, but it can be achieved economically by using portable air filtration in rooms with typical ceiling heights (less than 10 feet) for a cost of approximately $0.50 to $1 per square foot for do-it-yourself units and $2 to $5 per square foot for high-efficiency particulate air filters.
Collapse
|
7
|
Vigil K, Aw TG. Comparison of de novo assembly using long-read shotgun metagenomic sequencing of viruses in fecal and serum samples from marine mammals. Front Microbiol 2023; 14:1248323. [PMID: 37808316 PMCID: PMC10556685 DOI: 10.3389/fmicb.2023.1248323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Viral diseases of marine mammals are difficult to study, and this has led to a limited knowledge on emerging known and unknown viruses which are ongoing threats to animal health. Viruses are the leading cause of infectious disease-induced mass mortality events among marine mammals. Methods In this study, we performed viral metagenomics in stool and serum samples from California sea lions (Zalophus californianus) and bottlenose dolphins (Tursiops truncates) using long-read nanopore sequencing. Two widely used long-read de novo assemblers, Canu and Metaflye, were evaluated to assemble viral metagenomic sequencing reads from marine mammals. Results Both Metaflye and Canu assembled similar viral contigs of vertebrates, such as Parvoviridae, and Poxviridae. Metaflye assembled viral contigs that aligned with one viral family that was not reproduced by Canu, while Canu assembled viral contigs that aligned with seven viral families that was not reproduced by Metaflye. Only Canu assembled viral contigs from dolphin and sea lion fecal samples that matched both protein and nucleotide RefSeq viral databases using BLASTx and BLASTn for Anelloviridae, Parvoviridae and Circoviridae families. Viral contigs assembled with Canu aligned with torque teno viruses and anelloviruses from vertebrate hosts. Viruses associated with invertebrate hosts including densoviruses, Ambidensovirus, and various Circoviridae isolates were also aligned. Some of the invertebrate and vertebrate viruses reported here are known to potentially cause mortality events and/or disease in different seals, sea stars, fish, and bivalve species. Discussion Canu performed better by producing the most viral contigs as compared to Metaflye with assemblies aligning to both protein and nucleotide databases. This study suggests that marine mammals can be used as important sentinels to surveil marine viruses that can potentially cause diseases in vertebrate and invertebrate hosts.
Collapse
Affiliation(s)
| | - Tiong Gim Aw
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| |
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: 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
|
9
|
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: 2.0] [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
|
10
|
Van PD, Mai NTA, Nguyen VT, Nguyen TTH, Van Dong H, Le PN, Lai TNH, Thi PN, Pham NT, Nguyen LT, Anh DBT, Le VP. Detection and genetic characterization of canine distemper virus isolated in civets in Vietnam. Res Vet Sci 2023; 154:97-101. [PMID: 36521201 DOI: 10.1016/j.rvsc.2022.12.004] [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: 11/03/2022] [Revised: 11/22/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Canine distemper (CD), caused by the canine distemper virus (CDV), is a lethal systemic disease to a wide range of wild and domestic carnivorous hosts, including civets. In this study, a possible CD outbreak in a backyard farm with 32 diseased civets (Viverricula indica) in Hanoi, Vietnam, was investigated. The sick civets showed CD-like clinical signs such as anorexia, sedentary behavior, diarrhea, dermatitis, nasal, and footpad hyperkeratosis. Various tissue samples collected from the dead civets were utilized for molecular screening of CDV and histopathological examination. The genetic detection and characterization confirmed that samples collected from dead civets tested positive for CDV. The phylogenetic analysis based on the full-length H gene sequences indicated that all CDV strains isolated from civets belonged to the Asia-1 lineage and were closely related to the CDV strains previously reported from dogs in Thailand, China, and Vietnam. Histopathological examination showed severe interstitial pneumonia, hemorrhagic alveolar septa, necrotic alveolar epithelial cells, necrotic, degenerated, or lost Purkinje cells, eosinophilic intracytoplasmic inclusion bodies, edema, and perivascular cuff. This study confirmed the detection of CDV in civets for the first time in Vietnam.
Collapse
Affiliation(s)
- Phai Dam Van
- Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Trau Quy Town, Gia Lam District, Hanoi 131000, Viet Nam
| | | | - Van Tam Nguyen
- Institute of Veterinary Science and Technology (IVST), Hanoi, Viet Nam
| | | | - Hieu Van Dong
- Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Trau Quy Town, Gia Lam District, Hanoi 131000, Viet Nam
| | - Phuong Nam Le
- Institute of Veterinary Science and Technology (IVST), Hanoi, Viet Nam
| | - Thi Ngoc Ha Lai
- Institute of Veterinary Science and Technology (IVST), Hanoi, Viet Nam
| | - Phuong Nguyen Thi
- Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Trau Quy Town, Gia Lam District, Hanoi 131000, Viet Nam
| | - Ngoc Thach Pham
- Institute of Veterinary Science and Technology (IVST), Hanoi, Viet Nam
| | - Lan Thi Nguyen
- Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Trau Quy Town, Gia Lam District, Hanoi 131000, Viet Nam
| | - Dao Bui Tran Anh
- Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Trau Quy Town, Gia Lam District, Hanoi 131000, Viet Nam
| | - Van Phan Le
- Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Trau Quy Town, Gia Lam District, Hanoi 131000, Viet Nam; Institute of Veterinary Science and Technology (IVST), Hanoi, Viet Nam.
| |
Collapse
|
11
|
FeMV is a cathepsin-dependent unique morbillivirus infecting the kidneys of domestic cats. Proc Natl Acad Sci U S A 2022; 119:e2209405119. [PMID: 36251995 PMCID: PMC9618091 DOI: 10.1073/pnas.2209405119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Feline morbillivirus (FeMV) is a recently discovered pathogen of domestic cats and has been classified as a morbillivirus in the Paramyxovirus family. We determined the complete sequence of FeMVUS5 directly from an FeMV-positive urine sample without virus isolation or cell passage. Sequence analysis of the viral genome revealed potential divergence from characteristics of archetypal morbilliviruses. First, the virus lacks the canonical polybasic furin cleavage signal in the fusion (F) glycoprotein. Second, conserved amino acids in the hemagglutinin (H) glycoprotein used by all other morbilliviruses for binding and/or fusion activation with the cellular receptor CD150 (signaling lymphocyte activation molecule [SLAM]/F1) are absent. We show that, despite this sequence divergence, FeMV H glycoprotein uses feline CD150 as a receptor and cannot use human CD150. We demonstrate that the protease responsible for cleaving the FeMV F glycoprotein is a cathepsin, making FeMV a unique morbillivirus and more similar to the closely related zoonotic Nipah and Hendra viruses. We developed a reverse genetics system for FeMVUS5 and generated recombinant viruses expressing Venus fluorescent protein from an additional transcription unit located either between the phospho-protein (P) and matrix (M) genes or the H and large (L) genes of the genome. We used these recombinant FeMVs to establish a natural infection and demonstrate that FeMV causes an acute morbillivirus-like disease in the cat. Virus was shed in the urine and detectable in the kidneys at later time points. This opens the door for long-term studies to address the postulated role of this morbillivirus in the development of chronic kidney disease.
Collapse
|
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
|
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: 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: 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
|
14
|
Multiple Receptors Involved in Invasion and Neuropathogenicity of Canine Distemper Virus: A Review. Viruses 2022; 14:v14071520. [PMID: 35891500 PMCID: PMC9317347 DOI: 10.3390/v14071520] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 12/04/2022] Open
Abstract
The canine distemper virus (CDV) is a morbillivirus that infects a broad range of terrestrial carnivores, predominantly canines, and is associated with high mortality. Similar to another morbillivirus, measles virus, which infects humans and nonhuman primates, CDV transmission from an infected host to a naïve host depends on two cellular receptors, namely, the signaling lymphocyte activation molecule (SLAM or CD150) and the adherens junction protein nectin-4 (also known as PVRL4). CDV can also invade the central nervous system by anterograde spread through olfactory nerves or in infected lymphocytes through the circulation, thus causing chronic progressive or relapsing demyelination of the brain. However, the absence of the two receptors in the white matter, primary cultured astrocytes, and neurons in the brain was recently demonstrated. Furthermore, a SLAM/nectin-4-blind recombinant CDV exhibits full cell-to-cell transmission in primary astrocytes. This strongly suggests the existence of a third CDV receptor expressed in neural cells, possibly glial cells. In this review, we summarize the recent progress in the study of CDV receptors, highlighting the unidentified glial receptor and its contribution to pathogenicity in the host nervous system. The reviewed studies focus on CDV neuropathogenesis, and neural receptors may provide promising directions for the treatment of neurological diseases caused by CDV. We also present an overview of other neurotropic viruses to promote further research and identification of CDV neural receptors.
Collapse
|
15
|
Shi N, Zhang L, Yu X, Zhu X, Zhang S, Zhang D, Duan M. Insight Into an Outbreak of Canine Distemper Virus Infection in Masked Palm Civets in China. Front Vet Sci 2021; 8:728238. [PMID: 34805333 PMCID: PMC8595205 DOI: 10.3389/fvets.2021.728238] [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: 06/21/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
In August 2019, a suspected outbreak of canine distemper was observed in a masked palm civet farm that also received stray civets and rescued wild civets in Henan Province of China. A virulent canine distemper virus (CDV) strain, named HN19, from vaccinated masked palm civets was the etiologic agent identified in this outbreak using RT-PCR and sequencing of the complete genome. Serological analysis indicated a lower positive rate of CDV-neutralizing antibody in wild civets than in captive civets. Phylogenetic analysis of viral hemagglutinin (H) and the complete genome showed high identities with Rockborn-like strains at the nucleotide (98.7~99.72%) and the closest nucleotide similarity with a strain that killed lesser pandas in China in 1997, but low identities with America-1 strains (vaccine strains). Most importantly, one distinct amino acid exchange in the H protein at position 540 Asp → Gly (D540G), which confers CDV with an improved ability to adapt and utilize the human receptor, was observed in HN19. This study represents the first reported outbreak of a Rockborn-like CDV strain infection in masked palm civets in China. Based on this report, the existence of Rockborn-like strains in Chinese wild animals may not only cause immune failure in captive animals, but may also confer increased zoonotic potential.
Collapse
Affiliation(s)
- Ning Shi
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Le Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiuhua Yu
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Xiangyu Zhu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Shu Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Daining Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ming Duan
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
16
|
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: 1.0] [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
|
17
|
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: 0.7] [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
|
18
|
Gallo G, Conceicao C, Tsirigoti C, Willett B, Graham SC, Bailey D. Application of error-prone PCR to functionally probe the morbillivirus Haemagglutinin protein. J Gen Virol 2021; 102. [PMID: 33739251 PMCID: PMC8290269 DOI: 10.1099/jgv.0.001580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The enveloped morbilliviruses utilise conserved proteinaceous receptors to enter host cells: SLAMF1 or Nectin-4. Receptor binding is initiated by the viral attachment protein Haemagglutinin (H), with the viral Fusion protein (F) driving membrane fusion. Crystal structures of the prototypic morbillivirus measles virus H with either SLAMF1 or Nectin-4 are available and have served as the basis for improved understanding of this interaction. However, whether these interactions remain conserved throughout the morbillivirus genus requires further characterisation. Using a random mutagenesis approach, based on error-prone PCR, we targeted the putative receptor binding site for SLAMF1 interaction on peste des petits ruminants virus (PPRV) H, identifying mutations that inhibited virus-induced cell-cell fusion. These data, combined with structural modelling of the PPRV H and ovine SLAMF1 interaction, indicate this region is functionally conserved across all morbilliviruses. Error-prone PCR provides a powerful tool for functionally characterising functional domains within viral proteins.
Collapse
Affiliation(s)
- Giulia Gallo
- The Pirbright Institute, Guildford, Surrey, GU24 0NF, UK
| | | | | | - Brian Willett
- MRC University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Stephen C Graham
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Dalan Bailey
- The Pirbright Institute, Guildford, Surrey, GU24 0NF, UK
| |
Collapse
|
19
|
Benfield CTO, Hill S, Shatar M, Shiilegdamba E, Damdinjav B, Fine A, Willett B, Kock R, Bataille A. Molecular epidemiology of peste des petits ruminants virus emergence in critically endangered Mongolian saiga antelope and other wild ungulates. Virus Evol 2021; 7:veab062. [PMID: 34754511 PMCID: PMC8570150 DOI: 10.1093/ve/veab062] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 01/06/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) causes disease in domestic and wild ungulates, is the target of a Global Eradication Programme, and threatens biodiversity. Understanding the epidemiology and evolution of PPRV in wildlife is important but hampered by the paucity of wildlife-origin PPRV genomes. In this study, full PPRV genomes were generated from three Mongolian saiga antelope, one Siberian ibex, and one goitered gazelle from the 2016-2017 PPRV outbreak. Phylogenetic analysis showed that for Mongolian and Chinese PPRV since 2013, the wildlife and livestock-origin genomes were closely related and interspersed. There was strong phylogenetic support for a monophyletic group of PPRV from Mongolian wildlife and livestock, belonging to a clade of lineage IV PPRV from livestock and wildlife from China since 2013. Discrete diffusion analysis found strong support for PPRV spread into Mongolia from China, and phylogeographic analysis indicated Xinjiang Province as the most likely origin, although genomic surveillance for PPRV is poor and lack of sampling from other regions could bias this result. Times of most recent common ancestor (TMRCA) were June 2015 (95 per cent highest posterior density (HPD): August 2014 to March 2016) for all Mongolian PPRV genomes and May 2016 (95 per cent HPD: October 2015 to October 2016) for Mongolian wildlife-origin PPRV. This suggests that PPRV was circulating undetected in Mongolia for at least 6 months before the first reported outbreak in August 2016 and that wildlife were likely infected before livestock vaccination began in October 2016. Finally, genetic variation and positively selected sites were identified that might be related to PPRV emergence in Mongolian wildlife. This study is the first to sequence multiple PPRV genomes from a wildlife outbreak, across several host species. Additional full PPRV genomes and associated metadata from the livestock-wildlife interface are needed to enhance the power of molecular epidemiology, support PPRV eradication, and safeguard the health of the whole ungulate community.
Collapse
Affiliation(s)
- Camilla T O Benfield
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, AL9 7TA UK
| | - Sarah Hill
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, AL9 7TA UK
| | - Munkduuren Shatar
- Department of Veterinary Services of Dundgobi province, General Authority for Veterinary Services of Mongolia (GAVS), Mandalgobi, Dundgobi Province 4800 Mongolia
| | - Enkhtuvshin Shiilegdamba
- Wildlife Conservation Society, Mongolia Program, Post Office 20A, PO Box 21 Ulaanbaatar 14200, Mongolia
| | | | - Amanda Fine
- Health Program, Wildlife Conservation Society, Bronx, New York 10460, USA
| | - Brian Willett
- MRC-University of Glasgow Centre for Virus Research, Henry Wellcome Building, Garscube Glasgow, G61 1QH UK
| | - Richard Kock
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, AL9 7TA UK
| | - Arnaud Bataille
- CIRAD, UMR ASTRE, F-34398 Montpellier, France
- ASTRE, University of Montpellier, CIRAD, INRAE, F-34398 Montpellier, France
| |
Collapse
|
20
|
Yadav AK, Rajak KK, Kumar A, Bhatt M, Chakravarti S, Muthu S, Dubal ZB, Khulape S, Yousuf RW, Rai V, Kumar B, Muthuchelvan D, Gupta PK, Singh RP, Singh R. Replication competence of canine distemper virus in cell lines expressing signaling lymphocyte activation molecule (SLAM) of goat, sheep and dog origin. Microb Pathog 2021; 156:104940. [PMID: 33962006 DOI: 10.1016/j.micpath.2021.104940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 11/30/2022]
Abstract
Cellular receptors play an important role in entry and cell to cell spread of morbillivirus infections. The cells expressing SLAM and Nectin-4 have been used for successful and efficient isolation of canine distemper virus (CDV) in high titre. There are several methods for generation of cells expressing receptor molecules. Here, we have used a comparatively cheaper and easily available method, pcDNA 3.1 (+) for engineering Vero cells to express SLAM gene of goat, sheep and dog origin (Vero/Goat/SLAM (VGS), Vero/Sheep/SLAM (VSS) and Vero/Dog/SLAM (VDS), respectively). The generated cell lines were then compared to test their efficacy to support CDV replication. CDV could be grown in high titre in the cells expressing SLAM and a difference of log two could be recorded in virus titre between VDS and native Vero cells. Also, CDV could be grown in a higher titre in VDS as compared to VGS and VSS. The finding of this study supports the preferential use of SLAM expressing cells over the native Vero cells by CDV. Further, the higher titre of CDV in cells expressing dog-SLAM as compared to the cells expressing SLAM of non-CDV hosts (i.e. goat and sheep) points towards the preferential use of dog SLAM by the CDV and may be a plausible reason for differential susceptibility of small ruminants and Canines to CDV.
Collapse
Affiliation(s)
- Ajay Kumar Yadav
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India; ICAR -National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Kaushal Kishor Rajak
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India.
| | - Ashok Kumar
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Mukesh Bhatt
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India; ICAR -National Organic Farming Research Institute, Tadong, Gangtok, Sikkim, 737102, India
| | - Soumendu Chakravarti
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Sankar Muthu
- Division of Parasitology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Z B Dubal
- Division of Veterinary Public Health, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Sagar Khulape
- ICAR-D-FMD, Indian Veterinary Research Institute (IVRI), Mukteswar, 263138, Nainital, Uttarakhand, India
| | - Raja Wasim Yousuf
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Vishal Rai
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Bablu Kumar
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Dhanavelu Muthuchelvan
- Division of Virology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Mukteswar, 263138, Nainital, Uttrakhand, India
| | - Praveen Kumar Gupta
- Division of Animal Biotechnology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Rabindra Prasad Singh
- Division of Biological Products, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | - Rajkumar Singh
- Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India.
| |
Collapse
|
21
|
Sylvatic Canine Morbillivirus in Captive Panthera Highlights Viral Promiscuity and the Need for Better Prevention Strategies. Pathogens 2021; 10:pathogens10050544. [PMID: 33946447 PMCID: PMC8147164 DOI: 10.3390/pathogens10050544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022] Open
Abstract
Canine Distemper Virus (CDV) is a multi-host morbillivirus that infects virtually all Carnivora and a few non-human primates. Here we describe a CDV outbreak in an exotic felid rescue center that led to the death of eight felids in the genus Panthera. Similar to domestic dogs and in contrast to previously described CDV cases in Panthera, severe pneumonia was the primary lesion and no viral antigens or CDV-like lesions were detected in the central nervous system. Four tigers succumbed to opportunistic infections. Viral hemagglutinin (H)-gene sequence was up to 99% similar to strains circulating contemporaneously in regional wildlife. CDV lesions in raccoons and skunk were primarily encephalitis. A few affected felids had at least one previous vaccination for CDV, while most felids at the center were vaccinated during the outbreak. Panthera sharing a fence or enclosure with infected conspecifics had significantly higher chances of getting sick or dying, suggesting tiger-tiger spread was more likely than recurrent spillover. Prior vaccination was incomplete and likely not protective. This outbreak highlights the need for further understanding of CDV epidemiology for species conservation and public health.
Collapse
|
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: 1.0] [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
|
Sialic Acid Receptors: The Key to Solving the Enigma of Zoonotic Virus Spillover. Viruses 2021; 13:v13020262. [PMID: 33567791 PMCID: PMC7915228 DOI: 10.3390/v13020262] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Emerging viral diseases are a major threat to global health, and nearly two-thirds of emerging human infectious diseases are zoonotic. Most of the human epidemics and pandemics were caused by the spillover of viruses from wild mammals. Viruses that infect humans and a wide range of animals have historically caused devastating epidemics and pandemics. An in-depth understanding of the mechanisms of viral emergence and zoonotic spillover is still lacking. Receptors are major determinants of host susceptibility to viruses. Animal species sharing host cell receptors that support the binding of multiple viruses can play a key role in virus spillover and the emergence of novel viruses and their variants. Sialic acids (SAs), which are linked to glycoproteins and ganglioside serve as receptors for several human and animal viruses. In particular, influenza and coronaviruses, which represent two of the most important zoonotic threats, use SAs as cellular entry receptors. This is a comprehensive review of our current knowledge of SA receptor distribution among animal species and the range of viruses that use SAs as receptors. SA receptor tropism and the predicted natural susceptibility to viruses can inform targeted surveillance of domestic and wild animals to prevent the future emergence of zoonotic viruses.
Collapse
|
24
|
Muñoz-Alía MÁ, Nace RA, Tischer A, Zhang L, Bah ES, Auton M, Russell SJ. MeV-Stealth: A CD46-specific oncolytic measles virus resistant to neutralization by measles-immune human serum. PLoS Pathog 2021; 17:e1009283. [PMID: 33534834 PMCID: PMC7886131 DOI: 10.1371/journal.ppat.1009283] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 02/16/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
The frequent overexpression of CD46 in malignant tumors has provided a basis to use vaccine-lineage measles virus (MeV) as an oncolytic virotherapy platform. However, widespread measles seropositivity limits the systemic deployment of oncolytic MeV for the treatment of metastatic neoplasia. Here, we report the development of MeV-Stealth, a modified vaccine MeV strain that exhibits oncolytic properties and escapes antimeasles antibodies in vivo. We engineered this virus using homologous envelope glycoproteins from the closely-related but serologically non-cross reactive canine distemper virus (CDV). By fusing a high-affinity CD46 specific single-chain antibody fragment (scFv) to the CDV-Hemagglutinin (H), ablating its tropism for human nectin-4 and modifying the CDV-Fusion (F) signal peptide we achieved efficient retargeting to CD46. A receptor binding affinity of ~20 nM was required to trigger CD46-dependent intercellular fusion at levels comparable to the original MeV H/F complex and to achieve similar antitumor efficacy in myeloma and ovarian tumor-bearing mice models. In mice passively immunized with measles-immune serum, treatment of ovarian tumors with MeV-Stealth significantly increased overall survival compared with treatment with vaccine-lineage MeV. Our results show that MeV-Stealth effectively targets and lyses CD46-expressing cancer cells in mouse models of ovarian cancer and myeloma, and evades inhibition by human measles-immune serum. MeV-Stealth could therefore represent a strong alternative to current oncolytic MeV strains for treatment of measles-immune cancer patients. Vaccine strains of the measles virus (MeV) have been shown to be promising anti-cancer agents because of the frequent overexpression of the host-cell receptor CD46 in human malignancies. However, anti-MeV antibodies in the human population severely restrict the use of MeV as an oncolytic agent. Here, we engineered a neutralization-resistant MeV vaccine, MeV-Stealth, by replacing its envelope glycoproteins with receptor-targeted glycoproteins from wild-type canine distemper virus. By fully-retargeting the new envelope to the receptor CD46, we found that in mouse models of ovarian cancer and myeloma MeV-Stealth displayed oncolytic properties similar to the parental MeV vaccine. Furthermore, we found that passive immunization with measles-immune human serum did not eliminate the oncolytic potency of the MeV-Stealth, whereas it did destroy the potency of the parental MeV strain. The virus we here report may be considered a suitable oncolytic agent for the treatment of MeV-immune patients.
Collapse
Affiliation(s)
- Miguel Ángel Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (MÁM-A); (SJR)
| | - Rebecca A. Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alexander Tischer
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Eugene S. Bah
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Matthew Auton
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (MÁM-A); (SJR)
| |
Collapse
|
25
|
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: 1.3] [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
|
26
|
Takeda M, Seki F, Yamamoto Y, Nao N, Tokiwa H. Animal morbilliviruses and their cross-species transmission potential. Curr Opin Virol 2020; 41:38-45. [PMID: 32344228 DOI: 10.1016/j.coviro.2020.03.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
Like measles virus (MV), whose primary hosts are humans, non-human animal morbilliviruses use SLAM (signaling lymphocytic activation molecule) and PVRL4 (nectin-4) expressed on immune and epithelial cells, respectively, as receptors. PVRL4's amino acid sequence is highly conserved across species, while that of SLAM varies significantly. However, non-host animal SLAMs often function as receptors for different morbilliviruses. Uniquely, human SLAM is somewhat specific for MV, but canine distemper virus, which shows the widest host range among morbilliviruses, readily gains the ability to use human SLAM. The host range for morbilliviruses is also modulated by their ability to counteract the host's innate immunity, but the risk of cross-species transmission of non-human animal morbilliviruses to humans could occur if MV is successfully eradicated.
Collapse
Affiliation(s)
- Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Nishi-Ikebukuro 3-34-1, Toshima-ku, Tokyo 171-8501, Japan
| | - Naganori Nao
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| | - Hiroaki Tokiwa
- Department of Chemistry, Rikkyo University, Nishi-Ikebukuro 3-34-1, Toshima-ku, Tokyo 171-8501, Japan
| |
Collapse
|
27
|
Viral Pathogenesis, Recombinant Vaccines, and Oncolytic Virotherapy: Applications of the Canine Distemper Virus Reverse Genetics System. Viruses 2020; 12:v12030339. [PMID: 32244946 PMCID: PMC7150803 DOI: 10.3390/v12030339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/26/2022] Open
Abstract
Canine distemper virus (CDV) is a highly contagious pathogen transmissible to a broad range of terrestrial and aquatic carnivores. Despite the availability of attenuated vaccines against CDV, the virus remains responsible for outbreaks of canine distemper (CD) with significant morbidity and mortality in domesticated and wild carnivores worldwide. CDV uses the signaling lymphocytic activation molecule (SLAM, or CD150) and nectin-4 (PVRL4) as entry receptors, well-known tumor-associated markers for several lymphadenomas and adenocarcinomas, which are also responsible for the lysis of tumor cells and apparent tumor regression. Thus, CDV vaccine strains have emerged as a promising platform of oncolytic viruses for use in animal cancer therapy. Recent advances have revealed that use of the CDV reverse genetic system (RGS) has helped increase the understanding of viral pathogenesis and explore the development of recombinant CDV vaccines. In addition, genetic engineering of CDV based on RGS approaches also has the potential of enhancing oncolytic activity and selectively targeting tumors. Here, we reviewed the host tropism and pathogenesis of CDV, and current development of recombinant CDV-based vaccines as well as their use as oncolytic viruses against cancers.
Collapse
|
28
|
Ng WM, Stelfox AJ, Bowden TA. Unraveling virus relationships by structure-based phylogenetic classification. Virus Evol 2020; 6:veaa003. [PMID: 32064119 PMCID: PMC7015158 DOI: 10.1093/ve/veaa003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Delineation of the intricacies of protein function from macromolecular structure constitutes a continual obstacle in the study of cell and pathogen biology. Structure-based phylogenetic analysis has emerged as a powerful tool for addressing this challenge, allowing the detection and quantification of conserved architectural properties between proteins, including those with low or no detectable sequence homology. With a focus on viral protein structure, we highlight how a number of investigations have utilized this powerful method to infer common functionality and ancestry.
Collapse
Affiliation(s)
- Weng M Ng
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Alice J Stelfox
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Thomas A Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
29
|
GENETIC CHARACTERISTICS OF CANINE DISTEMPER VIRUSES CIRCULATING IN WILDLIFE IN THE UNITED STATES. J Zoo Wildl Med 2020; 50:790-797. [PMID: 31926508 DOI: 10.1638/2019-0052] [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] [Accepted: 09/03/2019] [Indexed: 11/21/2022] Open
Abstract
Canine distemper virus (CDV) is a highly contagious disease of wild and domestic mammals. Maintenance of CDV among wildlife plays an important role in the disease epidemiology. Wild animals, including raccoons (Procyon lotor) and gray foxes (Urocyon cinereoargenteus), serve as reservoirs of CDV and hamper the control of the disease. Recently, we discovered that at least three different CDV lineages (America-3 [Edomex], America-4, and America-5] that are genetically different from the available vaccine strains are circulating in domestic dogs in the United States. Because wildlife serve as a reservoir for the virus, it is important to determine if wildlife play a role in the maintenance and spread of these lineages. To determine the genetic characteristics of circulating strains of CDV in wildlife in various geographic regions in the United States, we studied the nucleotide sequences of the hemagglutinin (H) gene of 25 CDV strains detected in nondomestic species. The species included were free-ranging wildlife: three fishers (Martes pennanti), six foxes, one skunk (Mephitis mephitis), 10 raccoons, two wolves (Canis lupus), and one mink (Neovison vison). Strains from two species in managed care, one sloth (Choloepus didactylus) and one red panda (Ailurus fulgens), were also evaluated. Phylogenetic analysis of the H genes indicated that in addition to America-3, America-4, and America-5 lineages, there are at least two other lineages circulating in US wildlife. One of these includes CDV nucleotide sequences that grouped with that of a single CDV isolate previously detected in a raccoon from Rhode Island in 2012. The other lineage is independent and genetically distinct from other CDV strains included in the analysis. Additional genetically variable strains were detected, mainly in raccoons, suggesting that this species may be the host responsible for the genetic variability of newly detected strains in the domestic dog population.
Collapse
|
30
|
Phylodynamic analysis of two amino acid substitutions in the hemagglutinin protein of canine distemper virus strains detected in fur-bearing animals in China. Virus Genes 2019; 56:58-66. [PMID: 31802380 DOI: 10.1007/s11262-019-01720-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/29/2019] [Indexed: 02/05/2023]
Abstract
Canine distemper virus (CDV) causes a highly contagious disease in a wide range of carnivores. The hemagglutinin (H) protein of viruses shows the highest variability and plays an important role in modulation of viral antigenicity, virulence, and receptor recognition. Since 2012, canine distemper (CD) outbreaks in fur-bearing animals (minks, foxes, raccoon dogs) caused by CDV variants with I542N and Y549H substitutions in the H protein have been frequently reported in China. To characterize the molecular evolutionary dynamics and epidemiological dynamics of CDV, 235 H gene sequences of CDV wild-type strains collected from 22 countries between 1975 and 2015, including 44 strains predominant in fur-bearing animals in China, were analyzed. The phylogenetic relationships and evolutionary rates of the CDV strains were determined by Bayesian phylogenetics. The CDV strains clustered into distinct geographic genotypes, irrespective of the species of isolation. All the variant strains formed a distinct monophyletic cluster and belonged to the F sub-genotype within the Asia-1 genotype-currently the predominant sub-genotype in fur-bearing animals in China. Evolutionary analysis suggested that the variant strains originated in 2006. Furthermore, the selection pressure analysis revealed that the Y549H substitution was under positive selection pressure for adaptation toward the fur-bearing animals. The residue at position 549 also showed structural interaction with the V domain of the mink signaling lymphocyte-activation molecule (SLAM) receptor based on the homology modeling of the H-SLAM complex. Our results suggested that the Y549H substitution contributed to the molecular adaptation of CDV variants in the fur-bearing animals during the viral evolutionary phase in China.
Collapse
|
31
|
Kennedy JM, Earle JP, Omar S, Abdullah H, Nielsen O, Roelke-Parker ME, Cosby SL. Canine and Phocine Distemper Viruses: Global Spread and Genetic Basis of Jumping Species Barriers. Viruses 2019; 11:E944. [PMID: 31615092 PMCID: PMC6833027 DOI: 10.3390/v11100944] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
Canine distemper virus (CDV) and phocine distemper (PDV) are closely-related members of the Paramyxoviridae family, genus morbillivirus, in the order Mononegavirales. CDV has a broad host range among carnivores. PDV is thought to be derived from CDV through contact between terrestrial carnivores and seals. PDV has caused extensive mortality in Atlantic seals and other marine mammals, and more recently has spread to the North Pacific Ocean. CDV also infects marine carnivores, and there is evidence of morbillivirus infection of seals and other species in Antarctica. Recently, CDV has spread to felines and other wildlife species in the Serengeti and South Africa. Some CDV vaccines may also have caused wildlife disease. Changes in the virus haemagglutinin (H) protein, particularly the signaling lymphocyte activation molecule (SLAM) receptor binding site, correlate with adaptation to non-canine hosts. Differences in the phosphoprotein (P) gene sequences between disease and non-disease causing CDV strains may relate to pathogenicity in domestic dogs and wildlife. Of most concern are reports of CDV infection and disease in non-human primates raising the possibility of zoonosis. In this article we review the global occurrence of CDV and PDV, and present both historical and genetic information relating to these viruses crossing species barriers.
Collapse
Affiliation(s)
- Judith M. Kennedy
- Wellcome Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7BL, UK; (J.M.K.); (S.O.); (H.A.)
| | - J.A. Philip Earle
- Wellcome Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7BL, UK; (J.M.K.); (S.O.); (H.A.)
| | - Shadia Omar
- Wellcome Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7BL, UK; (J.M.K.); (S.O.); (H.A.)
| | - Hani’ah Abdullah
- Wellcome Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7BL, UK; (J.M.K.); (S.O.); (H.A.)
| | - Ole Nielsen
- Department of Fisheries and Oceans Canada, Winnipeg, Manitoba R3T 2N6, Canada;
| | | | - S. Louise Cosby
- Wellcome Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7BL, UK; (J.M.K.); (S.O.); (H.A.)
- Virology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT4 3SD, UK
| |
Collapse
|
32
|
Quintero-Gil C, Rendon-Marin S, Martinez-Gutierrez M, Ruiz-Saenz J. Origin of Canine Distemper Virus: Consolidating Evidence to Understand Potential Zoonoses. Front Microbiol 2019; 10:1982. [PMID: 31555226 PMCID: PMC6722215 DOI: 10.3389/fmicb.2019.01982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/12/2019] [Indexed: 11/15/2022] Open
Affiliation(s)
- Carolina Quintero-Gil
- Grupo de Investigación en Ciencias Animales-GRICA, Universidad Cooperativa de Colombia, Bucaramanga, Colombia
| | - Santiago Rendon-Marin
- Grupo de Investigación en Ciencias Animales-GRICA, Universidad Cooperativa de Colombia, Bucaramanga, Colombia
| | - Marlen Martinez-Gutierrez
- Grupo de Investigación en Ciencias Animales-GRICA, Universidad Cooperativa de Colombia, Bucaramanga, Colombia.,Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales-GRICA, Universidad Cooperativa de Colombia, Bucaramanga, Colombia.,Asociación Colombiana de Virología, Bogotá, Colombia
| |
Collapse
|
33
|
Fukuhara H, Ito Y, Sako M, Kajikawa M, Yoshida K, Seki F, Mwaba MH, Hashiguchi T, Higashibata MA, Ose T, Kuroki K, Takeda M, Maenaka K. Specificity of Morbillivirus Hemagglutinins to Recognize SLAM of Different Species. Viruses 2019; 11:v11080761. [PMID: 31430904 PMCID: PMC6722581 DOI: 10.3390/v11080761] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/15/2022] Open
Abstract
Measles virus (MV) and canine distemper virus (CDV) are highly contagious and deadly, forming part of the morbillivirus genus. The receptor recognition by morbillivirus hemagglutinin (H) is important for determining tissue tropism and host range. Recent reports largely urge caution as regards to the potential expansion of host specificities of morbilliviruses. Nonetheless, the receptor-binding potential in different species of morbillivirus H proteins is largely unknown. Herein, we show that the CDV-H protein binds to the dog signaling lymphocyte activation molecule (SLAM), but not to the human, tamarin, or mouse SLAM. In contrast, MV-H can bind to human, tamarin and dog SLAM, but not to that of mice. Notably, MV binding to dog SLAM showed a lower affinity and faster kinetics than that of human SLAM, and MV exhibits a similar entry activity in dog SLAM- and human SLAM-expressing Vero cells. The mutagenesis study using a fusion assay, based on the MV-H–SLAM complex structure, revealed differences in tolerance for the receptor specificity between MV-H and CDV-H. These results provide insights into H-SLAM specificity related to potential host expansion.
Collapse
Affiliation(s)
- Hideo Fukuhara
- Center for Research and Education on Drug Discovery, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yuri Ito
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Miyuki Sako
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Mizuho Kajikawa
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Koki Yoshida
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Mwila Hilton Mwaba
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takao Hashiguchi
- Department of Virology, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | - Masa-Aki Higashibata
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Toyoyuki Ose
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Kimiko Kuroki
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Katsumi Maenaka
- Center for Research and Education on Drug Discovery, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
34
|
Muñoz-Alía MA, Russell SJ. Probing Morbillivirus Antisera Neutralization Using Functional Chimerism between Measles Virus and Canine Distemper Virus Envelope Glycoproteins. Viruses 2019; 11:E688. [PMID: 31357579 PMCID: PMC6722617 DOI: 10.3390/v11080688] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/23/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023] Open
Abstract
Measles virus (MeV) is monotypic. Live virus challenge provokes a broadly protective humoral immune response that neutralizes all known measles genotypes. The two surface glycoproteins, H and F, mediate virus attachment and entry, respectively, and neutralizing antibodies to H are considered the main correlate of protection. Herein, we made improvements to the MeV reverse genetics system and generated a panel of recombinant MeVs in which the globular head domain or stalk region of the H glycoprotein or the entire F protein, or both, were substituted with the corresponding protein domains from canine distemper virus (CDV), a closely related morbillivirus that resists neutralization by measles-immune sera. The viruses were tested for sensitivity to human or guinea pig neutralizing anti-MeV antisera and to ferret anti-CDV antisera. Virus neutralization was mediated by antibodies to both H and F proteins, with H being immunodominant in the case of MeV and F being so in the case of CDV. Additionally, the globular head domains of both MeV and CDV H proteins were immunodominant over their stalk regions. These data shed further light on the factors constraining the evolution of new morbillivirus serotypes.
Collapse
Affiliation(s)
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
35
|
Ohishi K, Maruyama T, Seki F, Takeda M. Marine Morbilliviruses: Diversity and Interaction with Signaling Lymphocyte Activation Molecules. Viruses 2019; 11:E606. [PMID: 31277275 PMCID: PMC6669707 DOI: 10.3390/v11070606] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 01/08/2023] Open
Abstract
Epidemiological reports of phocine distemper virus (PDV) and cetacean morbillivirus (CeMV) have accumulated since their discovery nearly 30 years ago. In this review, we focus on the interaction between these marine morbilliviruses and their major cellular receptor, the signaling lymphocyte activation molecule (SLAM). The three-dimensional crystal structure and homology models of SLAMs have demonstrated that 35 residues are important for binding to the morbillivirus hemagglutinin (H) protein and contribute to viral tropism. These 35 residues are essentially conserved among pinnipeds and highly conserved among the Caniformia, suggesting that PDV can infect these animals, but are less conserved among cetaceans. Because CeMV can infect various cetacean species, including toothed and baleen whales, the CeMV-H protein is postulated to have broader specificity to accommodate more divergent SLAM interfaces and may enable the virus to infect seals. In silico analysis of viral H protein and SLAM indicates that each residue of the H protein interacts with multiple residues of SLAM and vice versa. The integration of epidemiological, virological, structural, and computational studies should provide deeper insight into host specificity and switching of marine morbilliviruses.
Collapse
Affiliation(s)
- Kazue Ohishi
- Faculty of Engineering, Tokyo Polytechnic University, 1583, Iiyama, Atsugi, Kanagawa 243-0297, Japan.
| | - Tadashi Maruyama
- School of Marine Biosciences, Kitasato University, 1-15-1, Kitazato, Minami, Sagamihara, Kanagawa 252-0373, Japan
| | - Fumio Seki
- Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Makoto Takeda
- Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| |
Collapse
|
36
|
Duque-Valencia J, Sarute N, Olarte-Castillo XA, Ruíz-Sáenz J. Evolution and Interspecies Transmission of Canine Distemper Virus-An Outlook of the Diverse Evolutionary Landscapes of a Multi-Host Virus. Viruses 2019; 11:v11070582. [PMID: 31247987 PMCID: PMC6669529 DOI: 10.3390/v11070582] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/13/2019] [Accepted: 05/18/2019] [Indexed: 12/17/2022] Open
Abstract
Canine distemper virus (CDV) is a worldwide distributed virus which belongs to the genus Morbillivirus within the Paramyxoviridae family. CDV spreads through the lymphatic, epithelial, and nervous systems of domestic dogs and wildlife, in at least six orders and over 20 families of mammals. Due to the high morbidity and mortality rates and broad host range, understanding the epidemiology of CDV is not only important for its control in domestic animals, but also for the development of reliable wildlife conservation strategies. The present review aims to give an outlook of the multiple evolutionary landscapes and factors involved in the transmission of CDV by including epidemiological data from multiple species in urban, wild and peri-urban settings, not only in domestic animal populations but at the wildlife interface. It is clear that different epidemiological scenarios can lead to the presence of CDV in wildlife even in the absence of infection in domestic populations, highlighting the role of CDV in different domestic or wild species without clinical signs of disease mainly acting as reservoirs (peridomestic and mesocarnivores) that are often found in peridomestic habits triggering CDV epidemics. Another scenario is driven by mutations, which generate genetic variation on which random drift and natural selection can act, shaping the genetic structure of CDV populations leading to some fitness compensations between hosts and driving the evolution of specialist and generalist traits in CDV populations. In this scenario, the highly variable protein hemagglutinin (H) determines the cellular and host tropism by binding to signaling lymphocytic activation molecule (SLAM) and nectin-4 receptors of the host; however, the multiple evolutionary events that may have facilitated CDV adaptation to different hosts must be evaluated by complete genome sequencing. This review is focused on the study of CDV interspecies transmission by examining molecular and epidemiological reports based on sequences of the hemagglutinin gene and the growing body of studies of the complete genome; emphasizing the importance of long-term multidisciplinary research that tracks CDV in the presence or absence of clinical signs in wild species, and helping to implement strategies to mitigate the infection. Integrated research incorporating the experience of wildlife managers, behavioral and conservation biologists, veterinarians, virologists, and immunologists (among other scientific areas) and the inclusion of several wild and domestic species is essential for understanding the intricate epidemiological dynamics of CDV in its multiple host infections.
Collapse
Affiliation(s)
- July Duque-Valencia
- Grupo de Investigación en Ciencias Animales-GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, sede Medellín 050012, Colombia
| | - Nicolás Sarute
- Sección Genética Evolutiva, Facultad de Ciencias, Universidad de la Republica, Montevideo 11200, Uruguay
- Department of Microbiology and Immunology, UIC College of Medicine, Chicago, IL 60612, USA
| | - Ximena A Olarte-Castillo
- Facultad de Ciencias Exactas, Naturales y Agropecuarias. Universidad de Santander (UDES), sede Bucaramanga 680002, Colombia
| | - Julián Ruíz-Sáenz
- Grupo de Investigación en Ciencias Animales-GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, sede Medellín 050012, Colombia.
| |
Collapse
|
37
|
Yadav AK, Rajak KK, Bhatt M, Kumar A, Chakravarti S, Sankar M, Muthuchelvan D, Kumar R, Khulape S, Singh RP, Singh RK. Comparative sequence analysis of morbillivirus receptors and its implication in host range expansion. Can J Microbiol 2019; 65:783-794. [PMID: 31238018 DOI: 10.1139/cjm-2019-0008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
SLAM (CD150) and nectin-4 are the major morbillivirus receptors responsible for virus pathogenesis and host range expansion. Recently, morbillivirus infections have been reported in unnatural hosts, including endangered species, posing a threat to their conservation. To understand the host range expansion of morbilliviruses, we generated the full-length sequences of morbillivirus receptors (goat, sheep, and dog SLAM, and goat nectin-4) and tried to correlate their role in determining host tropism. A high level of amino acid identity was observed between the sequences of related species, and phylogenetic reconstruction showed that the receptor sequences of carnivores, marine mammals, and small ruminants grouped separately. Analysis of the ligand binding region (V region; amino acid residues 52-136) of SLAM revealed high amino acid identity between small ruminants and bovine SLAMs. Comparison of canine SLAM with ruminants and non-canids SLAM revealed appreciable changes, including charge alterations. Significant differences between feline SLAM and canine SLAM have been reported. The binding motifs of nectin-4 genes (FPAG motif and amino acid residues 60, 62, and 63) were found to be conserved in sheep, goat, and dog. The differences reported in the binding region may be responsible for the level of susceptibility or resistance of a species to a particular morbillivirus.
Collapse
Affiliation(s)
- Ajay Kumar Yadav
- Division of Biological Products, Indian Council of Agricultural Research (ICAR) - Indian Veterinary Research Institute (IVRI), Izatnagar-243122, Bareilly, Uttar Pradesh, India.,ICAR - National Research Centre on Pig, Rani, Guwahati, Assam-781131, India
| | - Kaushal Kishor Rajak
- Division of Biological Products, Indian Council of Agricultural Research (ICAR) - Indian Veterinary Research Institute (IVRI), Izatnagar-243122, Bareilly, Uttar Pradesh, India
| | - Mukesh Bhatt
- Division of Biological Products, Indian Council of Agricultural Research (ICAR) - Indian Veterinary Research Institute (IVRI), Izatnagar-243122, Bareilly, Uttar Pradesh, India.,ICAR - National Organic Farming Research Institute, Tadong, Gangtok, Sikkim-737102, India
| | - Ashok Kumar
- Division of Biological Products, Indian Council of Agricultural Research (ICAR) - Indian Veterinary Research Institute (IVRI), Izatnagar-243122, Bareilly, Uttar Pradesh, India
| | - Soumendu Chakravarti
- Division of Biological Products, Indian Council of Agricultural Research (ICAR) - Indian Veterinary Research Institute (IVRI), Izatnagar-243122, Bareilly, Uttar Pradesh, India
| | - Muthu Sankar
- Temperate Animal Husbandry Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Mukteswar-263138, Nainital, Uttarakhand, India
| | - Dhanavelu Muthuchelvan
- Division of Virology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Mukteswar-263138, Nainital, Uttarakhand, India
| | - Ravi Kumar
- Department of Biotechnology, Indian Institute of Technology, Roorkee-247667, Uttarakhand, India
| | - Sagar Khulape
- ICAR-D-FMD, Indian Veterinary Research Institute (IVRI), Mukteswar-263138, Nainital, Uttarakhand, India
| | - Rabindra Prasad Singh
- Division of Biological Products, Indian Council of Agricultural Research (ICAR) - Indian Veterinary Research Institute (IVRI), Izatnagar-243122, Bareilly, Uttar Pradesh, India
| | - Raj Kumar Singh
- Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Izatnagar-243122, Bareilly, Uttar Pradesh, India
| |
Collapse
|
38
|
Uhl EW, Kelderhouse C, Buikstra J, Blick JP, Bolon B, Hogan RJ. New world origin of canine distemper: Interdisciplinary insights. INTERNATIONAL JOURNAL OF PALEOPATHOLOGY 2019; 24:266-278. [PMID: 30743216 DOI: 10.1016/j.ijpp.2018.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 11/29/2018] [Accepted: 12/31/2018] [Indexed: 06/09/2023]
Abstract
OBJECTIVE Canine distemper virus (CDV), human measles virus (HMV), and rinderpest virus (RPV) of cattle are morbilliviruses that have caused devastating outbreaks for centuries. This paper seeks to reconstruct the evolutionary history of CDV. MATERIALS AND METHODS An interdisciplinary approach is adopted, synthesizing paleopathological analysis of 96 Pre-Columbian dogs (750-1470 CE) from the Weyanoke Old Town, Virginia site, with historical reports, molecular analysis and morbilliviral epidemiology. RESULTS Both measles (c.900CE) and rinderpest (c. 376 BCE) were first reported in Eurasia, while canine distemper was initially described in South America much later (1735 CE); there are no paleopathological indications of CDV in Weyanoke Old Town dogs. Molecularly, CDV is closely related to HMV, while viral codon usage indicates CDV may have previously infected humans; South American measles epidemics occurred prior to the emergence of canine distemper and would have facilitated HMV transmission and adaptation to dogs. CONCLUSIONS The measles epidemics that decimated indigenous South American populations in the 1500-1700 s likely facilitated the establishment of CDV as a canine pathogen, which eventually spread to Europe and beyond. SIGNIFICANCE Understanding the historical and environmental conditions that have driven morbilliviral evolution provides important insights into potential future threats of animal/human cross-species infections. LIMITATIONS Interpreting historical disease descriptions is difficult and the archaeological specimens are limited. Molecular sequence data and codon usage analyses rely on modern viruses. SUGGESTIONS FOR FURTHER RESEARCH Interdisciplinary approaches are increasingly needed to understand diseases of the past and present, as critical information and knowledge is scattered in different disciplines.
Collapse
Affiliation(s)
- Elizabeth W Uhl
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602-7388, USA.
| | - Charles Kelderhouse
- Augusta University/University of Georgia Medical Partnership, Athens, GA, 30602-7388, USA.
| | - Jane Buikstra
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ 85287-2402, USA.
| | - Jeffrey P Blick
- Department of Government and Sociology, Georgia College and State University, Milledgeville, GA 31061-0490, USA
| | - Brad Bolon
- Department of Government and Sociology, Georgia College and State University, Milledgeville, GA 31061-0490, USA.
| | - Robert J Hogan
- Department of Veterinary Biosciences and Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602-7388, USA.
| |
Collapse
|
39
|
Histopathological Characteristics and Expression of CDV-NP Antigen in the Brain of Serologically Positive Spontaneously Infected Red Foxes ( Vulpes Vulpes) In Western Serbia. ACTA VET-BEOGRAD 2019. [DOI: 10.2478/acve-2018-0035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Canine distemper virus (CDV) is a worldwide distributed RNA virus that can cause severe disease in carnivore and non-carnivore species. Red foxes are highly susceptible and may act as a reservoir of the virus. As in other wild species, distemper in red foxes can manifest as acute, systemic and chronic nervous form. In the present study, we detected antibodies against CDV among red foxes in Western Serbia, and analyzed histopathologically and immunohistochemically for CDV nuclear protein antigen (CDV-NP) brain samples derived from seropositive animals. Seroprevalence of CDV antibodies was 36.8%. Histopathological changes included gliosis, neuronal degeneration, satellitosis, mononuclear inflammation, demyelination and presence of inclusion bodies. Immunostaining showed a diffuse presence of CDV-NP antigen, mainly in the cytoplasm of astrocytes and neurons. Results of this work contribute to the opinion that red foxes act as a potential reservoir of CDV and underline the importance of routine vaccination of dogs that could come in close contact with these animals. Potential active surveillance program would give a better insight in the degree of CDV infection in wildlife.
Collapse
|
40
|
Structure-Guided Identification of a Nonhuman Morbillivirus with Zoonotic Potential. J Virol 2018; 92:JVI.01248-18. [PMID: 30232185 PMCID: PMC6232486 DOI: 10.1128/jvi.01248-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023] Open
Abstract
Morbilliviruses infect a broad range of mammalian hosts, including ruminants, carnivores, and humans. The recent eradication of rinderpest virus (RPV) and the active campaigns for eradication of the human-specific measles virus (MeV) have raised significant concerns that the remaining morbilliviruses may emerge in so-called vacated ecological niches. Seeking to assess the zoonotic potential of nonhuman morbilliviruses within human populations, we found that peste des petits ruminants virus (PPRV)-the small-ruminant morbillivirus-is restricted at the point of entry into human cells due to deficient interactions with human SLAMF1-the immune cell receptor for morbilliviruses. Using a structure-guided approach, we characterized a single amino acid change, mapping to the receptor-binding domain in the PPRV hemagglutinin (H) protein, which overcomes this restriction. The same mutation allowed escape from some cross-protective, human patient, anti-MeV antibodies, raising concerns that PPRV is a pathogen with zoonotic potential. Analysis of natural variation within human and ovine SLAMF1 also identified polymorphisms that could correlate with disease resistance. Finally, the mechanistic nature of the PPRV restriction was also investigated, identifying charge incompatibility and steric hindrance between PPRV H and human SLAMF1 proteins. Importantly, this research was performed entirely using surrogate virus entry assays, negating the requirement for in situ derivation of a human-tropic PPRV and illustrating alternative strategies for identifying gain-of-function mutations in viral pathogens.IMPORTANCE A significant proportion of viral pandemics occur following zoonotic transmission events, where animal-associated viruses jump species into human populations. In order to provide forewarnings of the emergence of these viruses, it is necessary to develop a better understanding of what determines virus host range, often at the genetic and structural levels. In this study, we demonstrated that the small-ruminant morbillivirus, a close relative of measles, is unable to use human receptors to enter cells; however, a change of a single amino acid in the virus is sufficient to overcome this restriction. This information will be important for monitoring this virus's evolution in the field. Of note, this study was undertaken in vitro, without generation of a fully infectious virus with this phenotype.
Collapse
|
41
|
De Luca E, Crisi PE, Di Domenico M, Malatesta D, Vincifori G, Di Tommaso M, Di Guardo G, Di Francesco G, Petrini A, Savini G, Boari A, Lorusso A. A real-time RT-PCR assay for molecular identification and quantitation of feline morbillivirus RNA from biological specimens. J Virol Methods 2018; 258:24-28. [PMID: 29730392 DOI: 10.1016/j.jviromet.2018.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/09/2018] [Accepted: 05/02/2018] [Indexed: 01/03/2023]
Abstract
The aim of this study was to develop a real-time RT-PCR to detect and quantitate feline morbillivirus (FeMV) RNA in biological samples. Primers and probe were targeted on a conserved region of FeMV P/V/C gene. To validate the assay with field samples, a total number of specimens of cats have been recruited including 264 urine and blood samples and compared with a generic RT-PCR targeting the L protein encoding gene of morbilliviruses. In addition, 385 tissue samples from 35 carcasses of cats have been also employed. RNA titres were low in all tested samples. Results also indicated the absence of cross-reaction with related morbilliviruses and existing pathogens of cats. In tissues with low levels of FeMV RNA, the presence of viral antigen was also evidenced by immunohistochemistry targeting the N viral protein. This newly described assay allows for a rapid, accurate and reliable quantitative detection of FeMV RNA that can be applied for diagnostics and research studies.
Collapse
Affiliation(s)
- Eliana De Luca
- Faculty of Veterinary Medicine, University of Teramo, Italy; Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Teramo, Italy
| | | | - Marco Di Domenico
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Teramo, Italy
| | - Daniela Malatesta
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Teramo, Italy
| | - Giacomo Vincifori
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Teramo, Italy
| | | | | | | | - Antonio Petrini
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Teramo, Italy
| | - Giovanni Savini
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Teramo, Italy
| | - Andrea Boari
- Faculty of Veterinary Medicine, University of Teramo, Italy
| | - Alessio Lorusso
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Teramo, Italy.
| |
Collapse
|
42
|
Baron MD, Diop B, Njeumi F, Willett BJ, Bailey D. Future research to underpin successful peste des petits ruminants virus (PPRV) eradication. J Gen Virol 2017; 98:2635-2644. [PMID: 29022862 PMCID: PMC5845661 DOI: 10.1099/jgv.0.000944] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is a significant pathogen of small ruminants and is prevalent in much of Africa, the Near and Middle East and Asia. Despite the availability of an efficacious and cheap live-attenuated vaccine, the virus has continued to spread, with its range stretching from Morocco in the west to China and Mongolia in the east. Some of the world's poorest communities rely on small ruminant farming for subsistence and the continued endemicity of PPRV is a constant threat to their livelihoods. Moreover, PPRV's effects on the world's population are felt broadly across many economic, agricultural and social situations. This far-reaching impact has prompted the Food and Agriculture Organization of the United Nations (FAO) and the World Organisation for Animal Health (OIE) to develop a global strategy for the eradication of this virus and its disease. PPRV is a morbillivirus and, given the experience of these organizations in eradicating the related rinderpest virus, the eradication of PPRV should be feasible. However, there are many critical areas where basic and applied virological research concerning PPRV is lacking. The purpose of this review is to highlight areas where new research could be performed in order to guide and facilitate the eradication programme. These areas include studies on disease transmission and epidemiology, the existence of wildlife reservoirs and the development of next-generation vaccines and diagnostics. With the support of the international virology community, the successful eradication of PPRV can be achieved.
Collapse
Affiliation(s)
- Michael D Baron
- The Pirbright Institute, Ash Rd Pirbright, Surrey GU24 0NF, UK
| | - Bouna Diop
- Food and Agriculture Organization of the United Nation, FAO, 00153 Rome, Italy
| | - Felix Njeumi
- Food and Agriculture Organization of the United Nation, FAO, 00153 Rome, Italy
| | - Brian J Willett
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow, G61 1QH, UK
| | - Dalan Bailey
- College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,The Pirbright Institute, Ash Rd Pirbright, Surrey GU24 0NF, UK
| |
Collapse
|
43
|
Affiliation(s)
- Dalan Bailey
- Centre for Human Virology, Institute of Immunology & Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
44
|
mosGCTL-7, a C-Type Lectin Protein, Mediates Japanese Encephalitis Virus Infection in Mosquitoes. J Virol 2017; 91:JVI.01348-16. [PMID: 28250133 DOI: 10.1128/jvi.01348-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/30/2017] [Indexed: 01/08/2023] Open
Abstract
Japanese encephalitis virus (JEV) is an arthropod-borne flavivirus prevalent in Asia and the Western Pacific and is the leading cause of viral encephalitis. JEV is maintained in a transmission cycle between mosquitoes and vertebrate hosts, but the molecular mechanisms by which the mosquito vector participates in transmission are unclear. We investigated the expression of all C-type lectins during JEV infection in Aedes aegypti The C-type lectin mosquito galactose-specific C-type lectin 7 (mosGCTL-7) (VectorBase accession no. AAEL002524) was significantly upregulated by JEV infection and facilitated infection in vivo and in vitro mosGCTL-7 bound to the N-glycan at N154 on the JEV envelope protein. This recognition of viral N-glycan by mosGCTL-7 is required for JEV infection, and we found that this interaction was Ca2+ dependent. After mosGCTL-7 bound to the glycan, mosPTP-1 bound to mosGCTL-7, promoting JEV entry. The viral burden in vivo and in vitro was significantly decreased by mosPTP-1 double-stranded RNA (dsRNA) treatment, and infection was abolished by anti-mosGCTL-7 antibodies. Our results indicate that the mosGCTL-7/mosPTP-1 pathway plays a key role in JEV infection in mosquitoes. An improved understanding of the mechanisms underlying flavivirus infection in mosquitoes will provide further opportunities for developing new strategies to control viral dissemination in nature.IMPORTANCE Japanese encephalitis virus is a mosquito-borne flavivirus and is the primary cause of viral encephalitis in the Asia-Pacific region. Twenty-four countries in the WHO Southeast Asia and Western Pacific regions have endemic JEV transmission, which exposes >3 billion people to the risks of infection, although JEV primarily affects children. C-type lectins are host factors that play a role in flavivirus infection in humans, swine, and other mammals. In this study, we investigated C-type lectin functions in JEV-infected Aedes aegypti and Culex pipiens pallens mosquitoes and cultured cells. JEV infection changed the expression of almost all C-type lectins in vivo and in vitro, and mosGCTL-7 bound to the JEV envelope protein via an N-glycan at N154. Cell surface mosPTP-1 interacted with the mosGCTL-7-JEV complex to facilitate virus infection in vivo and in vitro Our findings provide further opportunities for developing new strategies to control arbovirus dissemination in nature.
Collapse
|
45
|
Thibault PA, Watkinson RE, Moreira-Soto A, Drexler JF, Lee B. Zoonotic Potential of Emerging Paramyxoviruses: Knowns and Unknowns. Adv Virus Res 2017; 98:1-55. [PMID: 28433050 PMCID: PMC5894875 DOI: 10.1016/bs.aivir.2016.12.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The risk of spillover of enzootic paramyxoviruses and the susceptibility of recipient human and domestic animal populations are defined by a broad collection of ecological and molecular factors that interact in ways that are not yet fully understood. Nipah and Hendra viruses were the first highly lethal zoonotic paramyxoviruses discovered in modern times, but other paramyxoviruses from multiple genera are present in bats and other reservoirs that have unknown potential to spillover into humans. We outline our current understanding of paramyxovirus reservoir hosts and the ecological factors that may drive spillover, and we explore the molecular barriers to spillover that emergent paramyxoviruses may encounter. By outlining what is known about enzootic paramyxovirus receptor usage, mechanisms of innate immune evasion, and other host-specific interactions, we highlight the breadth of unexplored avenues that may be important in understanding paramyxovirus emergence.
Collapse
Affiliation(s)
| | - Ruth E Watkinson
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Jan F Drexler
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Benhur Lee
- Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
46
|
Peste des petits ruminants (PPR): A neglected tropical disease in Maghreb region of North Africa and its threat to Europe. PLoS One 2017; 12:e0175461. [PMID: 28426782 PMCID: PMC5398521 DOI: 10.1371/journal.pone.0175461] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/27/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Peste des petits ruminants (PPR) is a contagious disease listed by the World Organisation for Animal health (OIE) as being a specific hazard. It affects sheep, goats, and wild ungulates, and is prevalent throughout the developing world particularly Asia, the Middle East, and Africa. PPR has been targeted for eradication by 2030 by the Food and Agriculture Organization of the United Nations (FAO) and the OIE, after the successful eradication of the related disease, rinderpest in cattle. PPR was first reported in 1942 in the Ivory Coast in Western Africa and has since extended its range in Asia, the Middle East, and Africa posing an immediate threat of incursion into Europe, South East Asia and South Africa. Although robust vaccines are available, the use of these vaccines in a systematic and rational manner is not widespread, resulting in this devastating disease becoming an important neglected tropical disease in the developing world. METHODOLOGY We isolated and characterized the PPR virus from an outbreak in Cheraga, northern Algeria, during October 2015 by analyzing the partial N-gene sequence in comparison with other viruses from the Maghreb region. As well as sequencing the full length viral genome and performing real-time RT-PCR on clinical samples. Maximum-likelihood and Bayesian temporal and phylogeographic analyses were performed to assess the persistence and spread of PPRV circulation from Eastern Africa in the Maghreb region of North Africa. CONCLUSIONS Recent PPR outbreaks in Cheraga, in the northern part of Algiers (October 2015) and North-West Morocco (June, 2015) highlight that PPRV has spread to the northern border of North Africa and may pose a threat of introduction to Europe. Phylogeographic analysis suggests that lineage IV PPRV has spread from Eastern Africa, most likely from the Sudan 2000 outbreak, into Northern Africa resulting in the 2008 Moroccan outbreak. Maximum-likelihood and Bayesian analysis shows that these North African viruses cluster closely together suggesting the existence of continual regional circulation. Considering the same virus is circulating in Algeria, Morocco and Tunisia, implementation of a common Maghreb PPR eradication strategy would be beneficial for the region.
Collapse
|
47
|
Lin LT, Richardson CD. The Host Cell Receptors for Measles Virus and Their Interaction with the Viral Hemagglutinin (H) Protein. Viruses 2016; 8:v8090250. [PMID: 27657109 PMCID: PMC5035964 DOI: 10.3390/v8090250] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/29/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022] Open
Abstract
The hemagglutinin (H) protein of measles virus (MeV) interacts with a cellular receptor which constitutes the initial stage of infection. Binding of H to this host cell receptor subsequently triggers the F protein to activate fusion between virus and host plasma membranes. The search for MeV receptors began with vaccine/laboratory virus strains and evolved to more relevant receptors used by wild-type MeV. Vaccine or laboratory strains of measles virus have been adapted to grow in common cell lines such as Vero and HeLa cells, and were found to use membrane cofactor protein (CD46) as a receptor. CD46 is a regulator that normally prevents cells from complement-mediated self-destruction, and is found on the surface of all human cells, with the exception of erythrocytes. Mutations in the H protein, which occur during adaptation and allow the virus to use CD46 as a receptor, have been identified. Wild-type isolates of measles virus cannot use the CD46 receptor. However, both vaccine/laboratory and wild-type strains can use an immune cell receptor called signaling lymphocyte activation molecule family member 1 (SLAMF1; also called CD150) and a recently discovered epithelial receptor known as Nectin-4. SLAMF1 is found on activated B, T, dendritic, and monocyte cells, and is the initial target for infections by measles virus. Nectin-4 is an adherens junction protein found at the basal surfaces of many polarized epithelial cells, including those of the airways. It is also over-expressed on the apical and basal surfaces of many adenocarcinomas, and is a cancer marker for metastasis and tumor survival. Nectin-4 is a secondary exit receptor which allows measles virus to replicate and amplify in the airways, where the virus is expelled from the body in aerosol droplets. The amino acid residues of H protein that are involved in binding to each of the receptors have been identified through X-ray crystallography and site-specific mutagenesis. Recombinant measles “blind” to each of these receptors have been constructed, allowing the virus to selectively infect receptor specific cell lines. Finally, the observations that SLAMF1 is found on lymphomas and that Nectin-4 is expressed on the cell surfaces of many adenocarcinomas highlight the potential of measles virus for oncolytic therapy. Although CD46 is also upregulated on many tumors, it is less useful as a target for cancer therapy, since normal human cells express this protein on their surfaces.
Collapse
Affiliation(s)
- Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Christopher D Richardson
- Department of Microbiology and Immunology, Dalhousie University, 5850 College St., Halifax, NS B3H 4R2, Canada.
- Department of Pediatrics and Canadian Center for Vaccinology, Izaak Walton Killam Health Centre, Halifax, NS B3K 6R8, Canada.
| |
Collapse
|
48
|
Organization, Function, and Therapeutic Targeting of the Morbillivirus RNA-Dependent RNA Polymerase Complex. Viruses 2016; 8:v8090251. [PMID: 27626440 PMCID: PMC5035965 DOI: 10.3390/v8090251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/02/2016] [Accepted: 09/05/2016] [Indexed: 12/16/2022] Open
Abstract
The morbillivirus genus comprises major human and animal pathogens, including the highly contagious measles virus. Morbilliviruses feature single stranded negative sense RNA genomes that are wrapped by a plasma membrane-derived lipid envelope. Genomes are encapsidated by the viral nucleocapsid protein forming ribonucleoprotein complexes, and only the encapsidated RNA is transcribed and replicated by the viral RNA-dependent RNA polymerase (RdRp). In this review, we discuss recent breakthroughs towards the structural and functional understanding of the morbillivirus polymerase complex. Considering the clinical burden imposed by members of the morbillivirus genus, the development of novel antiviral therapeutics is urgently needed. The viral polymerase complex presents unique structural and enzymatic properties that can serve as attractive candidates for druggable targets. We evaluate distinct strategies for therapeutic intervention and examine how high-resolution insight into the organization of the polymerase complex may pave the path towards the structure-based design and optimization of next-generation RdRp inhibitors.
Collapse
|
49
|
Nambulli S, Sharp CR, Acciardo AS, Drexler JF, Duprex WP. Mapping the evolutionary trajectories of morbilliviruses: what, where and whither. Curr Opin Virol 2016; 16:95-105. [PMID: 26921570 PMCID: PMC7102722 DOI: 10.1016/j.coviro.2016.01.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/27/2016] [Indexed: 12/15/2022]
Abstract
Morbilliviruses are important human and animal pathogens. Measles virus is the prototype and is the most infectious human pathogen on earth. Live attenuated vaccines have been used to control the infections. Rinderpest virus is the second virus to be eradicated from earth. New morbilliviruses have been identified in cats and vampire bats.
Morbilliviruses are pathogens of humans and other animals. Live attenuated morbillivirus vaccines have been used to end endemic transmission of measles virus (MV) in many parts of the developed world and to eradicate rinderpest virus. Entry is mediated by two different receptors which govern virus lymphotropism and epitheliotropism. Morbillivirus transmissibility is unparalleled and MV represents the most infectious human pathogen on earth. Their evolutionary origins remain obscure and their potential for adaption to new hosts is poorly understood. It has been suggested that MV could be eradicated. Therefore it is imperative to dissect barriers which restrict cross species infections. This is important as ecological studies identify novel morbilliviruses in a vast number of small mammals and carnivorous predators.
Collapse
Affiliation(s)
- Sham Nambulli
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Claire R Sharp
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Andrew S Acciardo
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - J Felix Drexler
- Institute of Virology, University of Bonn Medical Centre, Bonn, 53127, Germany; German Centre for Infection Research, Bonn-Cologne, Germany
| | - W Paul Duprex
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
50
|
Affiliation(s)
- Antra Zeltina
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- * E-mail: (TAB); (BL)
| | - Benhur Lee
- Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail: (TAB); (BL)
| |
Collapse
|