1
|
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
|
2
|
Schmitz KS, Rennick LJ, Tilston-Lunel NL, Comvalius AD, Laksono BM, Geers D, van Run P, de Vries RD, de Swart RL, Duprex WP. Rational attenuation of canine distemper virus (CDV) to develop a morbillivirus animal model that mimics measles in humans. J Virol 2024; 98:e0185023. [PMID: 38415596 PMCID: PMC10949419 DOI: 10.1128/jvi.01850-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024] Open
Abstract
Morbilliviruses are members of the family Paramyxoviridae and are known for their ability to cause systemic disease in a variety of mammalian hosts. The prototypic morbillivirus, measles virus (MeV), infects humans and still causes morbidity and mortality in unvaccinated children and young adults. Experimental infection studies in non-human primates have contributed to the understanding of measles pathogenesis. However, ethical restrictions call for the development of new animal models. Canine distemper virus (CDV) infects a wide range of animals, including ferrets, and its pathogenesis shares many features with measles. However, wild-type CDV infection is almost always lethal, while MeV infection is usually self-limiting. Here, we made five recombinant CDVs, predicted to be attenuated, and compared their pathogenesis to the non-attenuated recombinant CDV in a ferret model. Three viruses were insufficiently attenuated based on clinical signs, fatality, and systemic infection, while one virus was too attenuated. The last candidate virus caused a self-limiting infection associated with transient viremia and viral dissemination to all lymphoid tissues, was shed transiently from the upper respiratory tract, and did not result in acute neurological signs. Additionally, an in-depth phenotyping of the infected white blood cells showed lower infection percentages in all lymphocyte subsets when compared to the non-attenuated CDV. In conclusion, infection models using this candidate virus mimic measles and can be used to study pathogenesis-related questions and to test interventions for morbilliviruses in a natural host species.IMPORTANCEMorbilliviruses are transmitted via the respiratory route but cause systemic disease. The viruses use two cellular receptors to infect myeloid, lymphoid, and epithelial cells. Measles virus (MeV) remains an important cause of morbidity and mortality in humans, requiring animal models to study pathogenesis or intervention strategies. Experimental MeV infections in non-human primates are restricted by ethical and practical constraints, and animal morbillivirus infections in natural host species have been considered as alternatives. Inoculation of ferrets with wild-type canine distemper virus (CDV) has been used for this purpose, but in most cases, the virus overwhelms the immune system and causes highly lethal disease. Introduction of an additional transcription unit and an additional attenuating point mutation in the polymerase yielded a candidate virus that caused self-limiting disease with transient viremia and virus shedding. This rationally attenuated CDV strain can be used for experimental morbillivirus infections in ferrets that reflect measles in humans.
Collapse
Affiliation(s)
| | - Linda J. Rennick
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Natasha L. Tilston-Lunel
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | - Daryl Geers
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Peter van Run
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Rik L. de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - W. Paul Duprex
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Gonzales-Viera O, Goldstein T, Duignan P, Eiamcharoen P, Keel MK. California sea lion ( Zalophus californianus) lymph-node explant reveals involvement and possible transcriptional regulation of SLAM and nectin-4 during phocine distemper virus infection. Vet Pathol 2024; 61:125-134. [PMID: 37458158 DOI: 10.1177/03009858231186189] [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: 07/18/2023]
Abstract
Phocine distemper virus (PDV) is a significant cause of mortality for phocid seals; however, the susceptibility of otariids to this virus is poorly understood. The authors used a lymph-node explant culture system from California sea lions (Zalophus californianus, CSL) to investigate: (1) the role of signaling lymphocyte activation molecule (SLAM) and nectin-4 in PDV infection and their cellular expression patterns, (2) if PDV induces transcriptional regulation of cell-entry receptors, and (3) the involvement of apoptosis in PDV infection. PDV replicated in the lymph-node explants with peak replication 3 days post-infection (dpi), but the replication was not sustained 4 to 5 dpi. The PDV+ cells co-localized SLAM and nectin-4. These cells expressed IBA1, indicating a histiocytic lineage. Comparison of receptor expression between infected and mock-infected lymph nodes suggested transcriptional downregulation of both receptors during the initial stage of infection and upregulation during the late stage of infection, but the values lack of statistical significance. Cleaved caspase-3+ cells were slightly increased in the infected lymph nodes compared with the mock-infected lymph node from 1 to 4 dpi, but without statistical significance, and a few apoptotic cells co-expressed PDV. The results suggest that lymph-node explants might be an important model to study PDV pathogenesis. CSLs have the potential to be infected with PDV, as they express both cell-entry receptors in histiocytes. The lack of statistical significance in the PDV replication, transcriptional regulation of viral receptors, and changes in apoptosis suggest that although CSL might be infected by PDV, they might be less susceptible than phocid species.
Collapse
|
4
|
Siering O, Sawatsky B, Pfaller CK. Canine Distemper Virus Pathogenesis in the Ferret Model. Methods Mol Biol 2024; 2808:197-208. [PMID: 38743372 DOI: 10.1007/978-1-0716-3870-5_15] [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: 05/16/2024]
Abstract
Canine distemper virus (CDV) is a highly contagious pathogen within the morbillivirus genus infecting a wide range of different carnivore species. The virus shares most biological features with other closely related morbilliviruses, including clinical signs, tissue tropism, and replication cycle in the respective host organisms.In the laboratory environment, experimental infections of ferrets with CDV were established as a potent surrogate model for the analysis of several aspects of the biology of the human morbillivirus, measles virus (MeV). The animals are naturally susceptible to CDV and display severe clinical signs resembling the disease seen in patients infected with MeV. As seen with MeV, CDV infects immune cells and is thus associated with a strong transient immunosuppression. Here we describe several methods to evaluate viral load and parameters of immunosuppression in blood-circulating immune cells isolated from CDV-infected animals.
Collapse
Affiliation(s)
- Oliver Siering
- 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, MN, USA.
| |
Collapse
|
5
|
Allen C, Ellis A, Liang R, Lim A, Newbury S. Prolonged persistence of canine distemper virus RNA, and virus isolation in naturally infected shelter dogs. PLoS One 2023; 18:e0280186. [PMID: 36662900 PMCID: PMC9858347 DOI: 10.1371/journal.pone.0280186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023] Open
Abstract
Canine distemper virus remains an important source of morbidity and mortality in animal shelters. RT-PCR is commonly used to aid diagnosis and has been used to monitor dogs testing positive over time to gauge the end of infectious potential. Many dogs excrete viral RNA for prolonged periods which has complicated disease management. The goal of this retrospective study was to describe the duration and characteristics of viral RNA excretion in shelter dogs with naturally occurring CDV and investigate the relationship between that viral RNA excretion and infectious potential using virus isolation data. Records from 98 different humane organizations with suspect CDV were reviewed. A total of 5,920 dogs were tested with 1,393; 4,452; and 75 found to be positive, negative, or suspect on RT-PCR respectively. The median duration of a positive test was 34 days (n = 325), and 25% (82/325) of the dogs still excreting viral RNA after 62 days of monitoring. Virus isolation was performed in six dogs who were RT-PCR positive for > 60 days. Infectious virus was isolated only within the first two weeks of monitoring at or around the peak viral RNA excretion (as detected by the lowest cycle threshold) reported for each dog. Our findings suggest that peak viral RNA excretion and the days surrounding it might be used as a functional marker to gauge the end of infectious risk. Clarifying the earliest point in time when dogs testing positive for canine distemper by RT-PCR can be considered non-contagious will improve welfare and lifesaving potential of shelters by enabling recovered dogs to be cleared more quickly for live release outcomes.
Collapse
Affiliation(s)
- Carolyn Allen
- Department of Medical Sciences, Shelter Medicine Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Alexandre Ellis
- Department of Medical Sciences, Shelter Medicine Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Ruibin Liang
- Wisconsin Veterinary Diagnostic Laboratory, Virology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Ailam Lim
- Wisconsin Veterinary Diagnostic Laboratory, Virology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sandra Newbury
- Department of Medical Sciences, Shelter Medicine Program, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
6
|
Seki F, Takeda M. Novel and classical morbilliviruses: Current knowledge of three divergent morbillivirus groups. Microbiol Immunol 2022; 66:552-563. [PMID: 36151905 DOI: 10.1111/1348-0421.13030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/30/2022] [Accepted: 09/23/2022] [Indexed: 12/24/2022]
Abstract
Currently, seven species of morbillivirus have been classified. Six of these species (Measles morbillivirus, Rinderpest morbillivirus, Small ruminant morbillivirus, Canine morbillivirus, Phocine morbillivirus, and Cetacean morbillivirus) are highly infectious and cause serious systemic diseases in humans, livestock, domestic dogs, and wild animals. These species commonly use the host proteins signaling lymphocytic activation molecule (SLAM) and nectin-4 as receptors, and this usage contributes to their virulence. The seventh species (Feline morbillivirus: FeMV) is phylogenetically divergent from the six SLAM-using species. FeMV differs from the SLAM-using morbillivirus group in pathogenicity and infectivity, and is speculated to use non-SLAM receptors. Recently, novel species of morbilliviruses have been discovered in bats, rodents, and domestic pigs. Because the ability to use SLAM and nectin-4 is closely related to the infectivity and pathogenicity of morbilliviruses, investigation of the potential usage of these receptors is useful for estimating infectivity and pathogenicity. The SLAM-binding sites in the receptor-binding protein show high similarity among the SLAM-using morbilliviruses. This feature may help to estimate whether novel morbillivirus species can use SLAM as a receptor. A novel morbillivirus species isolated from wild mice diverged from the classified morbilliviruses in the phylogenetic tree, forming a third group separate from the SLAM-using morbillivirus group and FeMV. This suggests that the novel rodent morbillivirus may exhibit a different risk from the SLAM-using morbillivirus group, and analyses of its viral pathogenicity and infectivity toward humans are warranted.
Collapse
Affiliation(s)
- Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| |
Collapse
|
7
|
Phenotypic and Transcriptional Changes of Pulmonary Immune Responses in Dogs Following Canine Distemper Virus Infection. Int J Mol Sci 2022; 23:ijms231710019. [PMID: 36077417 PMCID: PMC9456005 DOI: 10.3390/ijms231710019] [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: 08/10/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
Canine distemper virus (CDV), a morbillivirus within the family Paramyxoviridae, is a highly contagious infectious agent causing a multisystemic, devastating disease in a broad range of host species, characterized by severe immunosuppression, encephalitis and pneumonia. The present study aimed at investigating pulmonary immune responses of CDV-infected dogs in situ using immunohistochemistry and whole transcriptome analyses by bulk RNA sequencing. Spatiotemporal analysis of phenotypic changes revealed pulmonary immune responses primarily driven by MHC-II+, Iba-1+ and CD204+ innate immune cells during acute and subacute infection phases, which paralleled pathologic lesion development and coincided with high viral loads in CDV-infected lungs. CD20+ B cell numbers initially declined, followed by lymphoid repopulation in the advanced disease phase. Transcriptome analysis demonstrated an increased expression of transcripts related to innate immunity, antiviral defense mechanisms, type I interferon responses and regulation of cell death in the lung of CDV-infected dogs. Molecular analyses also revealed disturbed cytokine responses with a pro-inflammatory M1 macrophage polarization and impaired mucociliary defense in CDV-infected lungs. The exploratory study provides detailed data on CDV-related pulmonary immune responses, expanding the list of immunologic parameters potentially leading to viral elimination and virus-induced pulmonary immunopathology in canine distemper.
Collapse
|
8
|
Multiple Receptors Involved in Invasion and Neuropathogenicity of Canine Distemper Virus: A Review. Viruses 2022; 14:v14071520. [PMID: 35891500 PMCID: PMC9317347 DOI: 10.3390/v14071520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 12/04/2022] Open
Abstract
The canine distemper virus (CDV) is a morbillivirus that infects a broad range of terrestrial carnivores, predominantly canines, and is associated with high mortality. Similar to another morbillivirus, measles virus, which infects humans and nonhuman primates, CDV transmission from an infected host to a naïve host depends on two cellular receptors, namely, the signaling lymphocyte activation molecule (SLAM or CD150) and the adherens junction protein nectin-4 (also known as PVRL4). CDV can also invade the central nervous system by anterograde spread through olfactory nerves or in infected lymphocytes through the circulation, thus causing chronic progressive or relapsing demyelination of the brain. However, the absence of the two receptors in the white matter, primary cultured astrocytes, and neurons in the brain was recently demonstrated. Furthermore, a SLAM/nectin-4-blind recombinant CDV exhibits full cell-to-cell transmission in primary astrocytes. This strongly suggests the existence of a third CDV receptor expressed in neural cells, possibly glial cells. In this review, we summarize the recent progress in the study of CDV receptors, highlighting the unidentified glial receptor and its contribution to pathogenicity in the host nervous system. The reviewed studies focus on CDV neuropathogenesis, and neural receptors may provide promising directions for the treatment of neurological diseases caused by CDV. We also present an overview of other neurotropic viruses to promote further research and identification of CDV neural receptors.
Collapse
|
9
|
Gradauskaite V, Khosravi M, Plattet P. Selective SLAM/CD150 Receptor-Detargeting of Canine Distemper Virus. Virus Res 2022; 318:198841. [DOI: 10.1016/j.virusres.2022.198841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 11/29/2022]
|
10
|
Abstract
Canine distemper virus (CDV) is a highly contagious pathogen and is known to enter the host via the respiratory tract and disseminate to various organs. Current hypotheses speculate that CDV uses the homologous cellular receptors of measles virus (MeV), SLAM and nectin-4, to initiate the infection process. For validation, here, we established the well-differentiated air-liquid interface (ALI) culture model from primary canine tracheal airway epithelial cells. By applying the green fluorescent protein (GFP)-expressing CDV vaccine strain and recombinant wild-type viruses, we show that cell-free virus infects the airway epithelium mainly via the paracellular route and only after prior disruption of tight junctions by pretreatment with EGTA; this infection was related to nectin-4 but not to SLAM. Remarkably, when CDV-preinfected DH82 cells were cocultured on the basolateral side of canine ALI cultures grown on filter supports with a 1.0-μm pore size, cell-associated CDV could be transmitted via cell-to-cell contact from immunocytes to airway epithelial cultures. Finally, we observed that canine ALI cultures formed syncytia and started to release cell-free infectious viral particles from the apical surface following treatment with an inhibitor of the JAK/STAT signaling pathway (ruxolitinib). Our findings show that CDV can overcome the epithelial barrier through different strategies, including infection via immunocyte-mediated transmission and direct infection via the paracellular route when tight junctions are disrupted. Our established model can be adapted to other animals for studying the transmission routes and the pathogenicity of other morbilliviruses. IMPORTANCE Canine distemper virus (CDV) is not only an important pathogen of carnivores, but it also serves as a model virus for analyzing measles virus pathogenesis. To get a better picture of the different stages of infection, we used air-liquid interface cultures to analyze the infection of well-differentiated airway epithelial cells by CDV. Applying a coculture approach with DH82 cells, we demonstrated that cell-mediated infection from the basolateral side of well-differentiated epithelial cells is more efficient than infection via cell-free virus. In fact, free virus was unable to infect intact polarized cells. When tight junctions were interrupted by treatment with EGTA, cells became susceptible to infection, with nectin-4 serving as a receptor. Another interesting feature of CDV infection is that infection of well-differentiated airway epithelial cells does not result in virus egress. Cell-free virions are released from the cells only in the presence of an inhibitor of the JAK/STAT signaling pathway. Our results provide new insights into how CDV can overcome the barrier of the airway epithelium and reveal similarities and some dissimilarities compared to measles virus.
Collapse
|
11
|
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
|
12
|
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: 14] [Impact Index Per Article: 3.5] [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
|
13
|
C Protein is Essential for Canine Distemper Virus Virulence and Pathogenicity in Ferrets. J Virol 2021; 95:JVI.01840-20. [PMID: 33239455 PMCID: PMC7851556 DOI: 10.1128/jvi.01840-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Paramyxoviruses, including members of the genus Morbillivirus, express accessory proteins with ancillary functions during viral replication. One of these, the C protein, is expressed from an alternate open reading frame (ORF) located in the P gene. The measles virus (MeV) C protein has been implicated in modulation of interferon signaling, but has more recently been shown to play a vital role in regulation of viral transcription and replication, preventing the excessive production of double-stranded RNA. Failure to do so, as seen with C-deficient MeV, leads to early activation of innate immune responses resulting in restriction of viral replication and attenuation in the host. One puzzling aspect of morbillivirus C protein biology has been the finding that a C-deficient canine distemper virus (CDV) generated with a similar mutagenesis strategy displayed no attenuation in ferrets, an animal model commonly used to evaluate CDV pathogenesis. To resolve how virus lacking this protein could maintain virulence, we re-visited the CDV C protein and found that truncated C proteins are expressed from the CDV gene using alternative downstream start codons even when the first start codon was disrupted. We introduced an additional point mutation abrogating expression of these truncated C proteins. A new CDV with this mutation was attenuated in vitro and led to increased activation of protein kinase R. It was also strongly attenuated in ferrets, inducing only mild disease in infected animals, thus replicating the phenotype of C-deficient MeV. Our results demonstrate the crucial role of morbillivirus C proteins in pathogenesis.IMPORTANCE The measles (MeV) and canine distemper viruses (CDV) express accessory proteins that regulate the host immune response and enhance replication. The MeV C protein is critical in preventing the generation of excess immunostimulatory double-stranded RNA. C protein-deficient MeV is strongly attenuated compared to wild-type virus, whereas CDV with a similarly disrupted C open reading frame is fully pathogenic. Here we show that CDV can compensate the disrupting mutations by expression of truncated, but apparently functional C proteins from several alternative start codons. We generated a new recombinant CDV that does not express these truncated C protein. This virus was attenuated both in cell culture and in ferrets, and finally resolves the paradox of the MeV and CDV C proteins, showing that both in fact have similar functions important for viral pathogenesis.
Collapse
|
14
|
Weckworth JK, Davis BW, Roelke-Parker ME, Wilkes RP, Packer C, Eblate E, Schwartz MK, Mills LS. Identifying Candidate Genetic Markers of CDV Cross-Species Pathogenicity in African Lions. Pathogens 2020; 9:E872. [PMID: 33114123 PMCID: PMC7690837 DOI: 10.3390/pathogens9110872] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
Canine distemper virus (CDV) is a multi-host pathogen with variable clinical outcomes of infection across and within species. We used whole-genome sequencing (WGS) to search for viral markers correlated with clinical distemper in African lions. To identify candidate markers, we first documented single-nucleotide polymorphisms (SNPs) differentiating CDV strains associated with different clinical outcomes in lions in East Africa. We then conducted evolutionary analyses on WGS from all global CDV lineages to identify loci subject to selection. SNPs that both differentiated East African strains and were under selection were mapped to a phylogenetic tree representing global CDV diversity to assess if candidate markers correlated with documented outbreaks of clinical distemper in lions (n = 3). Of 54 SNPs differentiating East African strains, ten were under positive or episodic diversifying selection and 20 occurred in the clinical strain despite strong purifying selection at those loci. Candidate markers were in functional domains of the RNP complex (n = 19), the matrix protein (n = 4), on CDV glycoproteins (n = 5), and on the V protein (n = 1). We found mutations at two loci in common between sequences from three CDV outbreaks of clinical distemper in African lions; one in the signaling lymphocytic activation molecule receptor (SLAM)-binding region of the hemagglutinin protein and another in the catalytic center of phosphodiester bond formation on the large polymerase protein. These results suggest convergent evolution at these sites may have a functional role in clinical distemper outbreaks in African lions and uncover potential novel barriers to pathogenicity in this species.
Collapse
Affiliation(s)
- Julie K. Weckworth
- Wildlife Biology Program, Department of Ecosystem and Conservation Sciences, W. A. Franke College of Forestry and Conservation, University of Montana, Missoula, MT 59812, USA;
- United States Department of Agriculture, Forest Service, National Genomics Center for Wildlife and Fish Conservation, Rocky Mountain Research Station, Missoula, MT 59801, USA
| | - Brian W. Davis
- Department of Veterinary Integrative Biosciences, Texas A&M University College of Veterinary Medicine, College Station, TX 77843, USA;
| | - Melody E. Roelke-Parker
- Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA;
| | - Rebecca P. Wilkes
- Department of Comparative Pathobiology, Animal Disease Diagnostic Laboratory, Purdue University College of Veterinary Medicine, West Lafayette, IN 47907, USA;
| | - Craig Packer
- Department of Ecology, Evolution and Behavior, University of Minnesota, St. Paul, MN 55108, USA;
| | - Ernest Eblate
- Tanzania Wildlife Research Institute, Arusha, Tanzania;
| | - Michael K. Schwartz
- Wildlife Biology Program, Department of Ecosystem and Conservation Sciences, W. A. Franke College of Forestry and Conservation, University of Montana, Missoula, MT 59812, USA;
- United States Department of Agriculture, Forest Service, National Genomics Center for Wildlife and Fish Conservation, Rocky Mountain Research Station, Missoula, MT 59801, USA
| | - L. Scott Mills
- Fisheries, Wildlife, and Conservation Biology Program, Department of Forestry and Environmental Resources, North Carolina State University, Raleigh, NC 27695, USA;
| |
Collapse
|
15
|
Petrova VN, Sawatsky B, Han AX, Laksono BM, Walz L, Parker E, Pieper K, Anderson CA, de Vries RD, Lanzavecchia A, Kellam P, von Messling V, de Swart RL, Russell CA. Incomplete genetic reconstitution of B cell pools contributes to prolonged immunosuppression after measles. Sci Immunol 2020; 4:4/41/eaay6125. [PMID: 31672862 DOI: 10.1126/sciimmunol.aay6125] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022]
Abstract
Measles is a disease caused by the highly infectious measles virus (MeV) that results in both viremia and lymphopenia. Lymphocyte counts recover shortly after the disappearance of measles-associated rash, but immunosuppression can persist for months to years after infection, resulting in increased incidence of secondary infections. Animal models and in vitro studies have proposed various immunological factors underlying this prolonged immune impairment, but the precise mechanisms operating in humans are unknown. Using B cell receptor (BCR) sequencing of human peripheral blood lymphocytes before and after MeV infection, we identified two immunological consequences from measles underlying immunosuppression: (i) incomplete reconstitution of the naïve B cell pool leading to immunological immaturity and (ii) compromised immune memory to previously encountered pathogens due to depletion of previously expanded B memory clones. Using a surrogate model of measles in ferrets, we investigated the clinical consequences of morbillivirus infection and demonstrated a depletion of vaccine-acquired immunity to influenza virus, leading to a compromised immune recall response and increased disease severity after secondary influenza virus challenge. Our results show that MeV infection causes changes in naïve and memory B lymphocyte diversity that persist after the resolution of clinical disease and thus contribute to compromised immunity to previous infections or vaccinations. This work highlights the importance of MeV vaccination not only for the control of measles but also for the maintenance of herd immunity to other pathogens, which can be compromised after MeV infection.
Collapse
Affiliation(s)
| | - Bevan Sawatsky
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines and DZIF TTU Emerging Infections, Langen, Germany
| | - Alvin X Han
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Brigitta M Laksono
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Lisa Walz
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines and DZIF TTU Emerging Infections, Langen, Germany
| | - Edyth Parker
- Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Kathrin Pieper
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Carl A Anderson
- Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Paul Kellam
- Department of Medicine, Division of Infectious Diseases, Imperial College Faculty of Medicine, Wright Fleming Institute, St Mary's Campus, London, UK.,Kymab Ltd., The Bennet Building, Babraham Research Campus, Cambridge, UK
| | - Veronika von Messling
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines and DZIF TTU Emerging Infections, Langen, Germany
| | - Rik L de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Colin A Russell
- Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
16
|
Weckworth JK, Davis BW, Dubovi E, Fountain-Jones N, Packer C, Cleaveland S, Craft ME, Eblate E, Schwartz M, Mills LS, Roelke-Parker M. Cross-species transmission and evolutionary dynamics of canine distemper virus during a spillover in African lions of Serengeti National Park. Mol Ecol 2020; 29:4308-4321. [PMID: 32306443 DOI: 10.1111/mec.15449] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/05/2020] [Accepted: 04/08/2020] [Indexed: 01/01/2023]
Abstract
The outcome of pathogen spillover from a reservoir to a novel host population can range from a "dead-end" when there is no onward transmission in the recipient population, to epidemic spread and even establishment in new hosts. Understanding the evolutionary epidemiology of spillover events leading to discrete outcomes in novel hosts is key to predicting risk and can lead to a better understanding of the mechanisms of emergence. Here we use a Bayesian phylodynamic approach to examine cross-species transmission and evolutionary dynamics during a canine distemper virus (CDV) spillover event causing clinical disease and population decline in an African lion population (Panthera leo) in the Serengeti Ecological Region between 1993 and 1994. Using 21 near-complete viral genomes from four species we found that this large-scale outbreak was likely ignited by a single cross-species spillover event from a canid reservoir to noncanid hosts <1 year before disease detection and explosive spread of CDV in lions. Cross-species transmission from other noncanid species probably fuelled the high prevalence of CDV across spatially structured lion prides. Multiple lines of evidence suggest that spotted hyenas (Crocuta crocuta) could have acted as the proximate source of CDV exposure in lions. We report 13 nucleotide substitutions segregating CDV strains found in canids and noncanids. Our results are consistent with the hypothesis that virus evolution played a role in CDV emergence in noncanid hosts following spillover during the outbreak, suggest that host barriers to clinical infection can limit outcomes of CDV spillover in novel host species.
Collapse
Affiliation(s)
- Julie K Weckworth
- Wildlife Biology Program, Department of Ecosystem and Conservation Sciences, W. A. Franke College of Forestry and Conservation, University of Montana, Missoula, MT, USA
| | - Brian W Davis
- Department of Veterinary Integrative Biosciences, Texas A&M University College of Veterinary Medicine, TX, USA
| | - Edward Dubovi
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | - Craig Packer
- Department of Ecology, Evolution and Behavior, University of Minnesota, St. Paul, MN, USA
| | - Sarah Cleaveland
- The Boyd Orr Centre for Population and Ecosystem Health, Institute of Biodiversity, Animal Heal and Comparative Medicine, University of Glasgow, Glasgow, UK.,Nelson Mandela African Institution for Science and Technology, Arusha, Tanzania
| | - Meggan E Craft
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Ernest Eblate
- Tanzania Wildlife Research Institute, Arusha, Tanzania
| | - Michael Schwartz
- Wildlife Biology Program, Department of Ecosystem and Conservation Sciences, W. A. Franke College of Forestry and Conservation, University of Montana, Missoula, MT, USA.,United States Department of Agriculture, Forest Service, National Genomics Center for Wildlife and Fish Conservation, Rocky Mountain Research Station, Missoula, MT, USA
| | - L Scott Mills
- Fisheries, Wildlife, and Conservation Biology Program, Department of Forestry and Environmental Resources, North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|
17
|
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: 1.0] [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
|
18
|
Shi P, Cao Z, Cheng Y, Cheng S, Yi L. Identification of Linear B-Cell Epitopes on Hemagglutinin Protein of Canine Distemper Virus Using Two Monoclonal Antibodies. Front Vet Sci 2020; 7:47. [PMID: 32185182 PMCID: PMC7058631 DOI: 10.3389/fvets.2020.00047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 01/20/2020] [Indexed: 11/13/2022] Open
Abstract
Canine distemper virus (CDV) belongs to the Morbillivirus genus of the Paramyxoviridae family, which causes a threat to the domestic dog and fur-animal industry. Hemagglutinin protein is a major membrane protein of the vital molecular factor in CDV tropism, also known to induce hosts to produce neutralizing antibodies. In the current study, we prepared two monoclonal antibodies, 1A5 and 2B8, against the H protein of the CDV-PS strain. A series of partially overlapping synthetic peptides covering the hemagglutinin protein (amino acids 50–204) were screened to define the linear epitope identified by 1A5 and 2B8 mAbs. 120QKTNFFNPNREFDFR134 (F8) and 178ARGDIFPPY186 (F14-1) are minimal linear epitopes recognized by 1A5 and 2B8 mAbs, respectively. Further investigations revealed that F8 is conserved in different CDV strains; however, F14-1 contains mutant residues 178, 179, and 180. The epitopes F8 and F14-1 localized at the surface of hemagglutinin protein in a three-dimensional (3D) structure. CDV-infected dog serum can also recognize the identified B-cell epitopes.
Collapse
Affiliation(s)
- Pengfei Shi
- Institute of Special Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhigang Cao
- Institute of Special Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuening Cheng
- Institute of Special Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Shipeng Cheng
- Institute of Special Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Li Yi
- Institute of Special Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
19
|
Navaratnarajah CK, Generous AR, Yousaf I, Cattaneo R. Receptor-mediated cell entry of paramyxoviruses: Mechanisms, and consequences for tropism and pathogenesis. J Biol Chem 2020; 295:2771-2786. [PMID: 31949044 DOI: 10.1074/jbc.rev119.009961] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Research in the last decade has uncovered many new paramyxoviruses, airborne agents that cause epidemic diseases in animals including humans. Most paramyxoviruses enter epithelial cells of the airway using sialic acid as a receptor and cause only mild disease. However, others cross the epithelial barrier and cause more severe disease. For some of these viruses, the host receptors have been identified, and the mechanisms of cell entry have been elucidated. The tetrameric attachment proteins of paramyxoviruses have vastly different binding affinities for their cognate receptors, which they contact through different binding surfaces. Nevertheless, all input signals are converted to the same output: conformational changes that trigger refolding of trimeric fusion proteins and membrane fusion. Experiments with selectively receptor-blinded viruses inoculated into their natural hosts have provided insights into tropism, identifying the cells and tissues that support growth and revealing the mechanisms of pathogenesis. These analyses also shed light on diabolically elegant mechanisms used by morbilliviruses, including the measles virus, to promote massive amplification within the host, followed by efficient aerosolization and rapid spread through host populations. In another paradigm of receptor-facilitated severe disease, henipaviruses, including Nipah and Hendra viruses, use different members of one protein family to cause zoonoses. Specific properties of different paramyxoviruses, like neurotoxicity and immunosuppression, are now understood in the light of receptor specificity. We propose that research on the specific receptors for several newly identified members of the Paramyxoviridae family that may not bind sialic acid is needed to anticipate their zoonotic potential and to generate effective vaccines and antiviral compounds.
Collapse
Affiliation(s)
| | - Alex R Generous
- Mayo Clinic Graduate School of Biomedical Sciences, Virology and Gene Therapy Track, Mayo Clinic, Rochester, Minnesota 55905
| | - Iris Yousaf
- Mayo Clinic Graduate School of Biomedical Sciences, Virology and Gene Therapy Track, Mayo Clinic, Rochester, Minnesota 55905
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55905.
| |
Collapse
|
20
|
Jo WK, Peters M, Kydyrmanov A, van de Bildt MWG, Kuiken T, Osterhaus A, Ludlow M. The Canine Morbillivirus Strain Associated with An Epizootic in Caspian Seals Provides New Insights into the Evolutionary History of this Virus. Viruses 2019; 11:E894. [PMID: 31557833 PMCID: PMC6832514 DOI: 10.3390/v11100894] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/13/2019] [Accepted: 09/24/2019] [Indexed: 01/28/2023] Open
Abstract
Canine morbillivirus (canine distemper virus; CDV) is a worldwide distributed morbillivirus that causes sporadic cases and recurrent epizootics among an increasing number of wild, feral, and domestic animal species. We investigated the evolutionary history of CDV strains involved in the 1988 Lake Baikal (CDVPS88) and the 2000 Caspian Sea (CDVPC00) seal die-offs by recovery of full-length sequences from archived material using next-generation sequencing. Bayesian phylogenetic analyses indicated that CDVPC00 constitutes a novel strain in a separate clade (tentatively termed "Caspian") from the America-1 clade, which is comprised of older vaccine strains. The America-1/Caspian monophyletic group is positioned most basally with respect to other clades and is estimated to have separated from other CDV clades around 1832. Our results indicate that CDVPC00 recovered from the epizootic in the Caspian Sea in 2000 belongs to a previously undetected novel clade and constitutes the most ancestral wild-type CDV clade.
Collapse
Affiliation(s)
- Wendy K Jo
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| | - Martin Peters
- Chemisches und Veterinäruntersuchungsamt Westfalen, 59821 Arnsberg, Germany.
| | - Aidyn Kydyrmanov
- Laboratory of Viral Ecology, Institute of Microbiology and Virology, 050010 Almaty, Kazakhstan.
| | | | - Thijs Kuiken
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| | - Martin Ludlow
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| |
Collapse
|
21
|
Sanchez JN, Hudgens BR. Impacts of Heterogeneous Host Densities and Contact Rates on Pathogen Transmission in the Channel Island Fox ( Urocyon littoralis). BIOLOGICAL CONSERVATION 2019; 236:593-603. [PMID: 32831352 PMCID: PMC7441459 DOI: 10.1016/j.biocon.2019.05.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Diseases threaten wildlife populations worldwide and have caused severe declines resulting in host species being listed as threatened or endangered. The risk of a widespread epidemic is especially high when pathogens are introduced to naive host populations, often leading to high morbidity and mortality. Prevention and control of these epidemics is based on knowledge of what drives pathogen transmission among hosts. Previous disease outbreaks suggest the spread of directly transmitted pathogens is determined by host contact rates and local host density. While theoretical models of disease spread typically assume a constant host density, most wildlife populations occur at a variety of densities across the landscape. We explored how spatial heterogeneity in host density influences pathogen spread by simulating the introduction and spread of rabies and canine distemper in a spatially heterogeneous population of Channel Island foxes (Urocyon littoralis), coupling fox density and contact rates with probabilities of viral transmission. For both diseases, the outcome of pathogen introductions varied widely among simulation iterations and depended on the density of hosts at the site of pathogen introduction. Introductions into areas of higher fox densities resulted in more rapid pathogen transmission and greater impact on the host population than if the pathogen was introduced at lower densities. Both pathogens were extirpated in a substantial fraction of iterations. Rabies was over five times more likely to go locally extinct when introduced at low host density sites than at high host-density sites, leaving an average of >99% of foxes uninfected. Canine distemper went extinct in >98% of iterations regardless of introduction site, but only after >90% of foxes had become infected. Our results highlight the difficulty in predicting the course of an epidemic, in part due to complex interactions between pathogen biology and host behavior, exacerbated by the spatial variation of most host populations.
Collapse
Affiliation(s)
- Jessica N Sanchez
- Institute for Wildlife Studies, P.O. Box 1104, Arcata, California 95518, USA
| | - Brian R Hudgens
- Institute for Wildlife Studies, P.O. Box 1104, Arcata, California 95518, USA
| |
Collapse
|
22
|
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: 1.8] [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
|
23
|
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: 1.0] [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
|
24
|
Rendon-Marin S, da Fontoura Budaszewski R, Canal CW, Ruiz-Saenz J. Tropism and molecular pathogenesis of canine distemper virus. Virol J 2019; 16:30. [PMID: 30845967 PMCID: PMC6407191 DOI: 10.1186/s12985-019-1136-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/25/2019] [Indexed: 11/10/2022] Open
Abstract
Background Canine distemper virus (CDV), currently termed Canine morbillivirus, is an extremely contagious disease that affects dogs. It is identified as a multiple cell tropism pathogen, and its host range includes a vast array of species. As a member of Mononegavirales, CDV has a negative, single-stranded RNA genome, which encodes eight proteins. Main body Regarding the molecular pathogenesis, the hemagglutinin protein (H) plays a crucial role both in the antigenic recognition and the viral interaction with SLAM and nectin-4, the host cells’ receptors. These cellular receptors have been studied widely as CDV receptors in vitro in different cellular models. The SLAM receptor is located in lymphoid cells; therefore, the infection of these cells by CDV leads to immunosuppression, the severity of which can lead to variability in the clinical disease with the potential of secondary bacterial infection, up to and including the development of neurological signs in its later stage. Conclusion Improving the understanding of the CDV molecules implicated in the determination of infection, especially the H protein, can help to enhance the biochemical comprehension of the difference between a wide range of CDV variants, their tropism, and different steps in viral infection. The regions of interaction between the viral proteins and the identified host cell receptors have been elucidated to facilitate this understanding. Hence, this review describes the significant molecular and cellular characteristics of CDV that contribute to viral pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12985-019-1136-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Santiago Rendon-Marin
- Grupo de Investigación en Ciencias Animales - GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga, Colombia
| | - Renata da Fontoura Budaszewski
- Laboratório de Virologia, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Cláudio Wageck Canal
- Laboratório de Virologia, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales - GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga, Colombia.
| |
Collapse
|
25
|
Cattaneo R, Donohue RC, Generous AR, Navaratnarajah CK, Pfaller CK. Stronger together: Multi-genome transmission of measles virus. Virus Res 2019; 265:74-79. [PMID: 30853585 DOI: 10.1016/j.virusres.2019.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/29/2022]
Abstract
Measles virus (MeV) is an immunosuppressive, extremely contagious RNA virus that remains a leading cause of death among children. MeV is dual-tropic: it replicates first in lymphatic tissue, causing immunosuppression, and then in epithelial cells of the upper airways, accounting for extremely efficient contagion. Efficient contagion is counter-intuitive because the enveloped MeV particles are large and relatively unstable. However, MeV particles can contain multiple genomes, which can code for proteins with different functional characteristics. These proteins can cooperate to promote virus spread in tissue culture, prompting the question of whether multi-genome MeV transmission may promote efficient MeV spread also in vivo. Consistent with this hypothesis, in well-differentiated primary human airway epithelia large genome populations spread rapidly through intercellular pores. In another line of research, it was shown that distinct lymphocytic-adapted and epithelial-adapted genome populations exist; cyclical adaptation studies indicate that suboptimal variants in one environment may constitute a low frequency reservoir for adaptation to the other environment. Altogether, these observations suggest that, in humans, MeV spread relies on en bloc genome transmission, and that genomic diversity is instrumental for rapid MeV dissemination within hosts.
Collapse
Affiliation(s)
- Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, United States; Mayo Graduate School, Mayo Clinic, Rochester, MN, 55905, United States.
| | - Ryan C Donohue
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, United States; Mayo Graduate School, Mayo Clinic, Rochester, MN, 55905, United States
| | - Alex R Generous
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, United States; Mayo Graduate School, Mayo Clinic, Rochester, MN, 55905, United States
| | - Chanakha K Navaratnarajah
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, United States; Mayo Graduate School, Mayo Clinic, Rochester, MN, 55905, United States
| | - Christian K Pfaller
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, United States; Paul-Ehrlich-Institute, Division of Veterinary Medicine, Langen, 63225, Germany
| |
Collapse
|
26
|
Benhaiem S, Marescot L, East ML, Kramer-Schadt S, Gimenez O, Lebreton JD, Hofer H. Slow recovery from a disease epidemic in the spotted hyena, a keystone social carnivore. Commun Biol 2018; 1:201. [PMID: 30480102 PMCID: PMC6244218 DOI: 10.1038/s42003-018-0197-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
Predicting the impact of disease epidemics on wildlife populations is one of the twenty-first century's main conservation challenges. The long-term demographic responses of wildlife populations to epidemics and the life history and social traits modulating these responses are generally unknown, particularly for K-selected social species. Here we develop a stage-structured matrix population model to provide a long-term projection of demographic responses by a keystone social predator, the spotted hyena, to a virulent epidemic of canine distemper virus (CDV) in the Serengeti ecosystem in 1993/1994 and predict the recovery time for the population following the epidemic. Using two decades of longitudinal data from 625 known hyenas, we demonstrate that although the reduction in population size was moderate, i.e., the population showed high ecological 'resistance' to the novel CDV genotype present, recovery was slow. Interestingly, high-ranking females accelerated the population's recovery, thereby lessening the impact of the epidemic on the population.
Collapse
Affiliation(s)
- Sarah Benhaiem
- Department of Ecological Dynamics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Strasse 17, D-10315, Berlin, Germany.
| | - Lucile Marescot
- Department of Ecological Dynamics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Strasse 17, D-10315, Berlin, Germany
- CEFE, CNRS, University Montpellier, University Paul Valéry Montpellier 3, EPHE, IRD, Montpellier, 34090, France
| | - Marion L East
- Department of Ecological Dynamics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Strasse 17, D-10315, Berlin, Germany
| | - Stephanie Kramer-Schadt
- Department of Ecological Dynamics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Strasse 17, D-10315, Berlin, Germany
- Department of Ecology, Technische Universität Berlin, Rothenburgstr. 12, 12165, Berlin, Germany
| | - Olivier Gimenez
- CEFE, CNRS, University Montpellier, University Paul Valéry Montpellier 3, EPHE, IRD, Montpellier, 34090, France
| | - Jean-Dominique Lebreton
- CEFE, CNRS, University Montpellier, University Paul Valéry Montpellier 3, EPHE, IRD, Montpellier, 34090, France
| | - Heribert Hofer
- Department of Ecological Dynamics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Strasse 17, D-10315, Berlin, Germany
- Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, Berlin, 14163, Germany
- Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| |
Collapse
|
27
|
Sánchez D, Cesarman-Maus G, Amador-Molina A, Lizano M. Oncolytic Viruses for Canine Cancer Treatment. Cancers (Basel) 2018; 10:cancers10110404. [PMID: 30373251 PMCID: PMC6266482 DOI: 10.3390/cancers10110404] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/16/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022] Open
Abstract
Oncolytic virotherapy has been investigated for several decades and is emerging as a plausible biological therapy with several ongoing clinical trials and two viruses are now approved for cancer treatment in humans. The direct cytotoxicity and immune-stimulatory effects make oncolytic viruses an interesting strategy for cancer treatment. In this review, we summarize the results of in vitro and in vivo published studies of oncolytic viruses in different phases of evaluation in dogs, using PubMed and Google scholar as search platforms, without time restrictions (to date). Natural and genetically modified oncolytic viruses were evaluated with some encouraging results. The most studied viruses to date are the reovirus, myxoma virus, and vaccinia, tested mostly in solid tumors such as osteosarcomas, mammary gland tumors, soft tissue sarcomas, and mastocytomas. Although the results are promising, there are issues that need addressing such as ensuring tumor specificity, developing optimal dosing, circumventing preexisting antibodies from previous exposure or the development of antibodies during treatment, and assuring a reasonable safety profile, all of which are required in order to make this approach a successful therapy in dogs.
Collapse
Affiliation(s)
- Diana Sánchez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico.
| | - Gabriela Cesarman-Maus
- Department of Hematology, Instituto Nacional de Cancerología, Mexico City 14080, Mexico.
| | - Alfredo Amador-Molina
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico.
| | - Marcela Lizano
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico.
| |
Collapse
|
28
|
Phylogenetic analysis of canine distemper virus in South African wildlife. PLoS One 2018; 13:e0199993. [PMID: 30020954 PMCID: PMC6051617 DOI: 10.1371/journal.pone.0199993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 06/18/2018] [Indexed: 02/04/2023] Open
Abstract
Canine distemper virus (CDV) causes a severe contagious disease in a broad range of hosts. This is the first study to genetically characterise CDV strains from four different wildlife species in South Africa. The phylogenetic diversity of CDV is examined, using the haemagglutinin gene. The South African wildlife CDV isolates showed a high degree of similarity to CDV in South African domestic dogs. Phylogenetic analyses confirmed the presence of 12 geographical lineages with CDV strains from South African wildlife falling within the Southern African lineage. The study reveals two possible co-circulating sub-genotypes corresponding to the northern and southern regions of South Africa respectively. CDV strains from the non-canid species were distinct, but similar to CDV isolates from domestic dog and wild canids. Residues at amino acid sites of the SLAM binding region support the notion that CDV strains encoding 519I / 549H are better adapted to non-canid species than canid species. The amino acids present at site 530 are conserved regardless of host species. Strains from South African wild carnivores showed no difference between host species with all strains presenting 530N. All non-canid strains in this study presented the combination 519I/549H. No evidence of host adaptation or lineage grouping was observed for the Nectin-4 binding region. Further studies should include CDV strains isolated from various hosts from a wider geographical range in South Africa.
Collapse
|
29
|
Canine Distemper Virus Spread and Transmission to Naive Ferrets: Selective Pressure on Signaling Lymphocyte Activation Molecule-Dependent Entry. J Virol 2018; 92:JVI.00669-18. [PMID: 29793948 DOI: 10.1128/jvi.00669-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022] Open
Abstract
Upon infection, morbilliviruses such as measles virus, rinderpest virus, and canine distemper virus (CDV) initially target immune cells via the signaling lymphocyte activation molecule (SLAM) before spreading to respiratory epithelia through the adherens junction protein nectin-4. However, the roles of these receptors in transmission from infected to naive hosts have not yet been formally tested. To experimentally addressing this question, we established a model of CDV contact transmission between ferrets. We show here that transmission of wild-type CDV sometimes precedes the onset of clinical disease. In contrast, transmission was not observed in most animals infected with SLAM- or nectin-4-blind CDVs, even though all animals infected with the nectin-4-blind virus developed sustained viremia. There was an unexpected case of transmission of a nectin-4-blind virus, possibly due to biting. Another unprecedented event was transient viremia in an infection with a SLAM-blind virus. We identified three compensatory mutations within or near the SLAM-binding surface of the attachment protein. A recombinant CDV expressing the mutated attachment protein regained the ability to infect ferret lymphocytes in vitro, but its replication was not as efficient as that of wild-type CDV. Ferrets infected with this virus developed transient viremia and fever, but there was no transmission to naive contacts. Our study supports the importance of epithelial cell infection and of sequential CDV H protein interactions first with SLAM and then nectin-4 receptors for transmission to naive hosts. It also highlights the in vivo selection pressure on the H protein interactions with SLAM.IMPORTANCE Morbilliviruses such as measles virus, rinderpest virus, and canine distemper virus (CDV) are highly contagious. Despite extensive knowledge of how morbilliviruses interact with their receptors, little is known about how those interactions influence viral transmission to naive hosts. In a ferret model of CDV contact transmission, we showed that sequential use of the signaling lymphocytic activation molecule (SLAM) and nectin-4 receptors is essential for transmission. In one animal infected with a SLAM-blind CDV, we documented mild viremia due to the acquisition of three compensatory mutations within or near the SLAM-binding surface. The interaction, however, was not sufficient to cause disease or sustain transmission to naive contacts. This work confirms the sequential roles of SLAM and nectin-4 in morbillivirus transmission and highlights the selective pressure directed toward productive interactions with SLAM.
Collapse
|
30
|
Marescot L, Benhaiem S, Gimenez O, Hofer H, Lebreton J, Olarte‐Castillo XA, Kramer‐Schadt S, East ML. Social status mediates the fitness costs of infection with canine distemper virus in Serengeti spotted hyenas. Funct Ecol 2018; 32:1237-1250. [PMID: 32313354 PMCID: PMC7163977 DOI: 10.1111/1365-2435.13059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/26/2018] [Indexed: 02/06/2023]
Abstract
The extent to which the fitness costs of infection are mediated by key life-history traits such as age or social status is still unclear. Within populations, individual heterogeneity in the outcome of infection is the result of two successive processes; the degree of contact with the pathogen (exposure) and the immune response to infection. In social mammals, because individuals holding high social status typically interact more frequently with group members, they should be more often in contact with infected individuals than those of low social status. However, when access to resources is determined by social status, individuals with a high social status are often better nourished, have a greater opportunity to allocate resources to immune processes and therefore should have a smaller chance of succumbing to infection than individuals with low social status.We investigated the risk and fitness costs of infection during a virulent epidemic of canine distemper virus (CDV) in a social carnivore, the spotted hyena, in the Serengeti National Park. We analysed two decades of detailed life-history data from 625 females and 816 males using a multi-event capture-mark-recapture model that accounts for uncertainty in the assignment of individual infection states.Cubs of mothers with a high social status had a lower probability of CDV infection and were more likely to survive infection than those with low social status. Subadult and adult females with high social status had a higher infection probability than those with low social status. Subadult females and pre-breeder males that had recovered from CDV infection had a lower survival than susceptible ones.Our study disentangles the relative importance of individual exposure and resource allocation to immune processes, demonstrates fitness costs of infection for juveniles, particularly for those with low social status, shows that patterns of infection can be driven by different mechanisms among juveniles and adults and establishes a negative relationship between infection and fitness in a free-ranging mammal. A http://onlinelibrary.wiley.com/doi/10.1111/1365-2435.13059/suppinfo is available for this article.
Collapse
Affiliation(s)
- Lucile Marescot
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
| | - Sarah Benhaiem
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
| | - Olivier Gimenez
- CEFEUMR 5175CNRSUniversité de MontpellierUniversité Paul‐Valéry MontpellierEPHEMontpellier Cedex 5France
| | - Heribert Hofer
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
- Department of Veterinary MedicineFreie Universität BerlinBerlinGermany
- Department of Biology, Chemistry, PharmacyFreie Universität BerlinBerlinGermany
| | - Jean‐Dominique Lebreton
- CEFEUMR 5175CNRSUniversité de MontpellierUniversité Paul‐Valéry MontpellierEPHEMontpellier Cedex 5France
| | | | - Stephanie Kramer‐Schadt
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
| | - Marion L. East
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
| |
Collapse
|
31
|
Leon AJ, Borisevich V, Boroumand N, Seymour R, Nusbaum R, Escaffre O, Xu L, Kelvin DJ, Rockx B. Host gene expression profiles in ferrets infected with genetically distinct henipavirus strains. PLoS Negl Trop Dis 2018; 12:e0006343. [PMID: 29538374 PMCID: PMC5868854 DOI: 10.1371/journal.pntd.0006343] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/26/2018] [Accepted: 02/24/2018] [Indexed: 02/05/2023] Open
Abstract
Henipavirus infection causes severe respiratory and neurological disease in humans that can be fatal. To characterize the pathogenic mechanisms of henipavirus infection in vivo, we performed experimental infections in ferrets followed by genome-wide gene expression analysis of lung and brain tissues. The Hendra, Nipah-Bangladesh, and Nipah-Malaysia strains caused severe respiratory and neurological disease with animals succumbing around 7 days post infection. Despite the presence of abundant viral shedding, animal-to-animal transmission did not occur. The host gene expression profiles of the lung tissue showed early activation of interferon responses and subsequent expression of inflammation-related genes that coincided with the clinical deterioration. Additionally, the lung tissue showed unchanged levels of lymphocyte markers and progressive downregulation of cell cycle genes and extracellular matrix components. Infection in the brain resulted in a limited breadth of the host responses, which is in accordance with the immunoprivileged status of this organ. Finally, we propose a model of the pathogenic mechanisms of henipavirus infection that integrates multiple components of the host responses.
Collapse
Affiliation(s)
- Alberto J. Leon
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Viktoriya Borisevich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Nahal Boroumand
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Robert Seymour
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Rebecca Nusbaum
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Olivier Escaffre
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Luoling Xu
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - David J. Kelvin
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
- International Institute of Infection and Immunity, Shantou University Medical College, Shantou, PRC
- * E-mail: (DJK); (BR)
| | - Barry Rockx
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States of America
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
- * E-mail: (DJK); (BR)
| |
Collapse
|
32
|
Abstract
The critically endangered population of Far Eastern leopards ( Panthera pardus orientalis) may number as few as 60 individuals and is at risk from stochastic processes such as infectious disease. During May 2015, a case of canine distemper virus (CDV) was diagnosed in a wild leopard exhibiting severe neurologic disease in the Russian territory of Primorskii Krai. Amplified sequences of the CDV hemagglutinin gene and phosphoprotein gene aligned within the Arctic-like clade of CDV, which includes viruses from elsewhere in Russia, China, Europe, and North America. Histologic examination of cerebral tissue revealed perivascular lymphoid cuffing and demyelination of the white matter consistent with CDV infection. Neutralizing antibodies against CDV were detected in archived serum from two wild Far Eastern leopards sampled during 1993-94, confirming previous exposure in the population. This leopard population is likely too small to maintain circulation of CDV, suggesting that infections arise from spillover from more-abundant domestic or wild carnivore reservoirs. Increasing the population size and establishment of additional populations of leopards would be important steps toward securing the future of this subspecies and reducing the risk posed by future outbreaks of CDV or other infectious diseases.
Collapse
|
33
|
Nectin-4 Interactions Govern Measles Virus Virulence in a New Model of Pathogenesis, the Squirrel Monkey (Saimiri sciureus). J Virol 2017; 91:JVI.02490-16. [PMID: 28331086 DOI: 10.1128/jvi.02490-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/13/2017] [Indexed: 12/16/2022] Open
Abstract
In addition to humans, only certain nonhuman primates are naturally susceptible to measles virus (MeV) infection. Disease severity is species dependent, ranging from mild to moderate for macaques to severe and even lethal for certain New World monkey species. To investigate if squirrel monkeys (Saimiri sciureus), which are reported to develop a course of disease similar to humans, may be better suited than macaques for the identification of virulence determinants or the evaluation of therapeutics, we infected them with a green fluorescent protein-expressing MeV. Compared to cynomolgus macaques (Macaca fascicularis) infected with the same virus, the squirrel monkeys developed more-severe immunosuppression, higher viral load, and a broader range of clinical signs typical for measles. In contrast, infection with an MeV unable to interact with the epithelial receptor nectin-4, while causing immunosuppression, resulted in only a mild and transient rash and a short-lived elevation of the body temperature. Similar titers of the wild-type and nectin-4-blind MeV were detected in peripheral blood mononuclear cells and lymph node homogenates, but only the wild-type virus was found in tracheal lavage fluids and urine. Thus, our study demonstrates the importance of MeV interactions with nectin-4 for clinical disease in the new and better-performing S. sciureus model of measles pathogenesis.IMPORTANCE The characterization of mechanisms underlying measles virus clinical disease has been hampered by the lack of an animal model that reproduces the course of disease seen in human patients. Here, we report that infection of squirrel monkeys (Saimiri sciureus) fulfills these requirements. Comparative infection with wild-type and epithelial cell receptor-blind viruses demonstrated the importance of epithelial cell infection for clinical disease, highlighting the spread to epithelia as an attractive target for therapeutic strategies.
Collapse
|
34
|
Nikolin VM, Olarte‐Castillo XA, Osterrieder N, Hofer H, Dubovi E, Mazzoni CJ, Brunner E, Goller KV, Fyumagwa RD, Moehlman PD, Thierer D, East ML. Canine distemper virus in the Serengeti ecosystem: molecular adaptation to different carnivore species. Mol Ecol 2017; 26:2111-2130. [PMID: 27928865 PMCID: PMC7168383 DOI: 10.1111/mec.13902] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/25/2016] [Accepted: 09/02/2016] [Indexed: 12/24/2022]
Abstract
Was the 1993/1994 fatal canine distemper virus (CDV) epidemic in lions and spotted hyaenas in the Serengeti ecosystem caused by the recent spillover of a virulent domestic dog strain or one well adapted to these noncanids? We examine this question using sequence data from 13 'Serengeti' strains including five complete genomes obtained between 1993 and 2011. Phylogenetic and haplotype network analyses reveal that strains from noncanids during the epidemic were more closely related to each other than to those from domestic or wild canids. All noncanid 'Serengeti' strains during the epidemic encoded: (1) one novel substitution G134S in the CDV-V protein; and (2) the rare amino acid combination 519I/549H at two sites under positive selection in the region of the CDV-H protein that binds to SLAM (CD 150) host cell receptors. Worldwide, only a few noncanid strains in the America II lineage encode CDV-H 519I/549H. All canid 'Serengeti' strains during the epidemic coded CDV-V 134G, and CDV-H 519R/549Y, or 519R/549H. A functional assay of cell entry revealed the highest performance by CDV-H proteins encoding 519I/549H in cells expressing lion SLAM receptors, and the highest performance by proteins encoding 519R/549Y, typical of dog strains worldwide, in cells expressing dog SLAM receptors. Our findings are consistent with an epidemic in lions and hyaenas caused by CDV variants better adapted to noncanids than canids, but not with the recent spillover of a dog strain. Our study reveals a greater complexity of CDV molecular epidemiology in multihost environments than previously thought.
Collapse
Affiliation(s)
- Veljko M. Nikolin
- Leibniz Institute for Zoo and Wildlife ResearchAlfred‐Kowalke‐Str. 1710315BerlinGermany
- Institut für VirologieFreie Universität BerlinRobert‐von‐Ostertag‐Str. 7‐1314163BerlinGermany
- Present address:
Boehringer Ingelheim Veterinary Research CenterBemeroder Str. 3130559HannoverGermany
| | | | - Nikolaus Osterrieder
- Institut für VirologieFreie Universität BerlinRobert‐von‐Ostertag‐Str. 7‐1314163BerlinGermany
| | - Heribert Hofer
- Leibniz Institute for Zoo and Wildlife ResearchAlfred‐Kowalke‐Str. 1710315BerlinGermany
| | - Edward Dubovi
- Animal Health Diagnostic CentreCollege of Veterinary MedicineCornell UniversityIthacaNY14853USA
| | - Camila J. Mazzoni
- Leibniz Institute for Zoo and Wildlife ResearchAlfred‐Kowalke‐Str. 1710315BerlinGermany
- Berlin Center for Genomics in Biodiversity ResearchKönigin‐Luise‐Str. 6‐814195BerlinGermany
| | - Edgar Brunner
- Department of Medical StatisticsFaculty of MedicineUniversity of GöttingenHumboldtallee 3237073GöttingenGermany
| | - Katja V. Goller
- Leibniz Institute for Zoo and Wildlife ResearchAlfred‐Kowalke‐Str. 1710315BerlinGermany
- Present address:
Friedrich‐Loeffler‐InsitutBundesforschungsinstitut für TiergesundheitSüdufer 1017493Greifswald‐Insel RiemsGermany
| | | | | | - Dagmar Thierer
- Leibniz Institute for Zoo and Wildlife ResearchAlfred‐Kowalke‐Str. 1710315BerlinGermany
| | - Marion L. East
- Leibniz Institute for Zoo and Wildlife ResearchAlfred‐Kowalke‐Str. 1710315BerlinGermany
| |
Collapse
|
35
|
Morbillivirus Experimental Animal Models: Measles Virus Pathogenesis Insights from Canine Distemper Virus. Viruses 2016; 8:v8100274. [PMID: 27727184 PMCID: PMC5086610 DOI: 10.3390/v8100274] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 12/19/2022] Open
Abstract
Morbilliviruses share considerable structural and functional similarities. Even though disease severity varies among the respective host species, the underlying pathogenesis and the clinical signs are comparable. Thus, insights gained with one morbillivirus often apply to the other members of the genus. Since the Canine distemper virus (CDV) causes severe and often lethal disease in dogs and ferrets, it is an attractive model to characterize morbillivirus pathogenesis mechanisms and to evaluate the efficacy of new prophylactic and therapeutic approaches. This review compares the cellular tropism, pathogenesis, mechanisms of persistence and immunosuppression of the Measles virus (MeV) and CDV. It then summarizes the contributions made by studies on the CDV in dogs and ferrets to our understanding of MeV pathogenesis and to vaccine and drugs development.
Collapse
|
36
|
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: 6.6] [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
|
37
|
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: 4.3] [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
|
38
|
Sawatsky B, Bente DA, Czub M, von Messling V. Morbillivirus and henipavirus attachment protein cytoplasmic domains differently affect protein expression, fusion support and particle assembly. J Gen Virol 2016; 97:1066-1076. [PMID: 26813519 PMCID: PMC7482510 DOI: 10.1099/jgv.0.000415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The amino-terminal cytoplasmic domains of paramyxovirus attachment glycoproteins
include trafficking signals that influence protein processing and cell surface
expression. To characterize the role of the cytoplasmic domain in protein expression,
fusion support and particle assembly in more detail, we constructed chimeric Nipah
virus (NiV) glycoprotein (G) and canine distemper virus (CDV) haemagglutinin (H)
proteins carrying the respective heterologous cytoplasmic domain, as well as a series
of mutants with progressive deletions in this domain. CDV H retained fusion function
and was normally expressed on the cell surface with a heterologous cytoplasmic
domain, while the expression and fusion support of NiV G was dramatically decreased
when its cytoplasmic domain was replaced with that of CDV H. The cell surface
expression and fusion support functions of CDV H were relatively insensitive to
cytoplasmic domain deletions, while short deletions in the corresponding region of
NiV G dramatically decreased both. In addition, the first 10 residues of the CDV H
cytoplasmic domain strongly influence its incorporation into virus-like particles
formed by the CDV matrix (M) protein, while the co-expression of NiV M with NiV G had
no significant effect on incorporation of G into particles. The cytoplasmic domains
of both the CDV H and NiV G proteins thus contribute differently to the virus life
cycle.
Collapse
Affiliation(s)
- Bevan Sawatsky
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA.,INRS-Institut Armand-Frappier, University of Quebec, Laval, Quebec, Canada.,Veterinary Medicine Division, Paul-Ehrlich-Institute, Langen, Germany.,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Dennis A Bente
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Markus Czub
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Veronika von Messling
- INRS-Institut Armand-Frappier, University of Quebec, Laval, Quebec, Canada.,Veterinary Medicine Division, Paul-Ehrlich-Institute, Langen, Germany
| |
Collapse
|
39
|
Zhao J, Shi N, Sun Y, Martella V, Nikolin V, Zhu C, Zhang H, Hu B, Bai X, Yan X. Pathogenesis of canine distemper virus in experimentally infected raccoon dogs, foxes, and minks. Antiviral Res 2015. [PMID: 26210812 DOI: 10.1016/j.antiviral.2015.07.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Canine distemper virus (CDV) infects a broad range of carnivores and causes a highly contagious disease with severe immunosuppression. The disease severity markedly varies in different species. To investigate the pathogenesis of CDV in raccoon dog (Nyctereutes procyonoides), fox (Vulpes vulpes) and mink (Neovison vison) species, three groups of CDV sero-negative animals were infected with CDV strain LN(10)1. This CDV strain belongs to the Asia-1 genotype, which is epidemiologically predominant in carnivores in China. CDV infection provoked marked differences in virulence in the three species that were studied. Raccoon dogs developed fever, severe conjunctivitis, and pathological lesions, with 100% (5/5) mortality and with high viral RNA loads in organs within 15 days post infection (dpi). In infected foxes, the onset of the disease was delayed, with 40% (2/5) mortality by 21 dpi. Infected minks developed only mild clinical signs and pathological lesions, and mortality was not observed. Raccoon dogs and foxes showed more severe immune suppression (lymphopenia, decreased lymphocyte proliferation, viremia and low-level virus neutralizing antibodies) than minks. We also observed a distinct pattern of cytokine mRNA transcripts at different times after infection. Decreased IFN-γ and IL-4 mRNA responses were evident in the animals with fatal disease, while up-regulation of these cytokines was observed in the animals surviving the infection. Increased TNF-α response was detected in animals with mild or severe clinical signs. Based on the results, we could distinguish three different patterns of disease after experimental CDV infection, e.g. a mild form in minks, a moderate form in foxes and a severe disease in raccoon dogs. The observed differences in susceptibility to CDV could be related to distinct host cytokine profiles. Comparative evaluation of CDV pathogenesis in various animal species is pivotal to generate models suitable for the evaluation of CDV-host interactions and of vaccine response.
Collapse
Affiliation(s)
- Jianjun Zhao
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China; State Key Laboratory for Molecular Biology of Special Economic Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China.
| | - Ning Shi
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Yangang Sun
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Vito Martella
- Department of Veterinary Medicine, University of Bari, Bari, Italy
| | - Veljko Nikolin
- Boehringer Ingelheim Veterinary Research Center GmbH & Co. KG, Hannover, Germany
| | - Chunsheng Zhu
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Hailing Zhang
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Bo Hu
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Xue Bai
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China
| | - Xijun Yan
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China; State Key Laboratory for Molecular Biology of Special Economic Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, PR China.
| |
Collapse
|
40
|
Enkirch T, von Messling V. Ferret models of viral pathogenesis. Virology 2015; 479-480:259-70. [PMID: 25816764 PMCID: PMC7111696 DOI: 10.1016/j.virol.2015.03.017] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 01/28/2015] [Accepted: 03/02/2015] [Indexed: 11/26/2022]
Abstract
Emerging and well-known viral diseases remain one the most important global public health threats. A better understanding of their pathogenesis and mechanisms of transmission requires animal models that accurately reproduce these aspects of the disease. Here we review the role of ferrets as an animal model for the pathogenesis of different respiratory viruses with an emphasis on influenza and paramyxoviruses. We will describe the anatomic and physiologic characteristics that contribute to the natural susceptibility of ferrets to these viruses, and provide an overview of the approaches available to analyze their immune responses. Recent insights gained using this model will be highlighted, including the development of new prophylactic and therapeutic approaches. To provide decision criteria for the use of this animal model, its strengths and limitations will be discussed. Ferrets as models for respiratory virus pathogenesis. Ferrets as models for vaccine and drug efficacy assessment. Immunological tools for ferrets. Housing and handling of ferrets.
Collapse
Affiliation(s)
- T Enkirch
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - V von Messling
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany.
| |
Collapse
|
41
|
SLAM- and nectin-4-independent noncytolytic spread of canine distemper virus in astrocytes. J Virol 2015; 89:5724-33. [PMID: 25787275 DOI: 10.1128/jvi.00004-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/09/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Measles and canine distemper viruses (MeV and CDV, respectively) first replicate in lymphatic and epithelial tissues by using SLAM and nectin-4 as entry receptors, respectively. The viruses may also invade the brain to establish persistent infections, triggering fatal complications, such as subacute sclerosis pan-encephalitis (SSPE) in MeV infection or chronic, multiple sclerosis-like, multifocal demyelinating lesions in the case of CDV infection. In both diseases, persistence is mediated by viral nucleocapsids that do not require packaging into particles for infectivity but are directly transmitted from cell to cell (neurons in SSPE or astrocytes in distemper encephalitis), presumably by relying on restricted microfusion events. Indeed, although morphological evidence of fusion remained undetectable, viral fusion machineries and, thus, a putative cellular receptor, were shown to contribute to persistent infections. Here, we first showed that nectin-4-dependent cell-cell fusion in Vero cells, triggered by a demyelinating CDV strain, remained extremely limited, thereby supporting a potential role of nectin-4 in mediating persistent infections in astrocytes. However, nectin-4 could not be detected in either primary cultured astrocytes or the white matter of tissue sections. In addition, a bioengineered "nectin-4-blind" recombinant CDV retained full cell-to-cell transmission efficacy in primary astrocytes. Combined with our previous report demonstrating the absence of SLAM expression in astrocytes, these findings are suggestive for the existence of a hitherto unrecognized third CDV receptor expressed by glial cells that contributes to the induction of noncytolytic cell-to-cell viral transmission in astrocytes. IMPORTANCE While persistent measles virus (MeV) infection induces SSPE in humans, persistent canine distemper virus (CDV) infection causes chronic progressive or relapsing demyelination in carnivores. Common to both central nervous system (CNS) infections is that persistence is based on noncytolytic cell-to-cell spread, which, in the case of CDV, was demonstrated to rely on functional membrane fusion machinery complexes. This inferred a mechanism where nucleocapsids are transmitted through macroscopically invisible microfusion events between infected and target cells. Here, we provide evidence that CDV induces such microfusions in a SLAM- and nectin-4-independent manner, thereby strongly suggesting the existence of a third receptor expressed in glial cells (referred to as GliaR). We propose that GliaR governs intercellular transfer of nucleocapsids and hence contributes to viral persistence in the brain and ensuing demyelinating lesions.
Collapse
|
42
|
Abstract
UNLABELLED Paramyxoviruses, including the human pathogen measles virus (MV), enter host cells by fusing their viral envelope with the target cell membrane. This fusion process is driven by the concerted actions of the two viral envelope glycoproteins, the receptor binding protein (hemagglutinin [H]) and the fusion (F) protein. H attaches to specific proteinaceous receptors on host cells; once the receptor engages, H activates F to directly mediate lipid bilayer fusion during entry. In a recent MV outbreak in South Africa, several HIV-positive people died of MV central nervous system (CNS) infection. We analyzed the virus sequences from these patients and found that specific intrahost evolution of the F protein had occurred and resulted in viruses that are "CNS adapted." A mutation in F of the CNS-adapted virus (a leucine-to-tryptophan change present at position 454) allows it to promote fusion with less dependence on engagement of H by the two known wild-type (wt) MV cellular receptors. This F protein is activated independently of H or the receptor and has reduced thermal stability and increased fusion activity compared to those of the corresponding wt F. These functional effects are the result of the single L454W mutation in F. We hypothesize that in the absence of effective cellular immunity, such as HIV infection, MV variants bearing altered fusion machinery that enabled efficient spread in the CNS underwent positive selection. IMPORTANCE Measles virus has become a concern in the United States and Europe due to recent outbreaks and continues to be a significant global problem. While live immunization is available, there are no effective therapies or prophylactics to combat measles infection in unprotected people. Additionally, vaccination does not adequately protect immunocompromised people, who are vulnerable to the more severe CNS manifestations of disease. We found that strains isolated from patients with measles virus infection of the CNS have fusion properties different from those of strains previously isolated from patients without CNS involvement. Specifically, the viral entry machinery is more active and the virus can spread, even in the absence of H. Our findings are consistent with an intrahost evolution of the fusion machinery that leads to neuropathogenic MV variants.
Collapse
|
43
|
Duignan PJ, Van Bressem MF, Baker JD, Barbieri M, Colegrove KM, De Guise S, de Swart RL, Di Guardo G, Dobson A, Duprex WP, Early G, Fauquier D, Goldstein T, Goodman SJ, Grenfell B, Groch KR, Gulland F, Hall A, Jensen BA, Lamy K, Matassa K, Mazzariol S, Morris SE, Nielsen O, Rotstein D, Rowles TK, Saliki JT, Siebert U, Waltzek T, Wellehan JF. Phocine distemper virus: current knowledge and future directions. Viruses 2014; 6:5093-134. [PMID: 25533658 PMCID: PMC4276944 DOI: 10.3390/v6125093] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/05/2014] [Accepted: 12/11/2014] [Indexed: 11/16/2022] Open
Abstract
Phocine distemper virus (PDV) was first recognized in 1988 following a massive epidemic in harbor and grey seals in north-western Europe. Since then, the epidemiology of infection in North Atlantic and Arctic pinnipeds has been investigated. In the western North Atlantic endemic infection in harp and grey seals predates the European epidemic, with relatively small, localized mortality events occurring primarily in harbor seals. By contrast, PDV seems not to have become established in European harbor seals following the 1988 epidemic and a second event of similar magnitude and extent occurred in 2002. PDV is a distinct species within the Morbillivirus genus with minor sequence variation between outbreaks over time. There is now mounting evidence of PDV-like viruses in the North Pacific/Western Arctic with serological and molecular evidence of infection in pinnipeds and sea otters. However, despite the absence of associated mortality in the region, there is concern that the virus may infect the large Pacific harbor seal and northern elephant seal populations or the endangered Hawaiian monk seals. Here, we review the current state of knowledge on PDV with particular focus on developments in diagnostics, pathogenesis, immune response, vaccine development, phylogenetics and modeling over the past 20 years.
Collapse
Affiliation(s)
- Pádraig J. Duignan
- Department of Ecosystem and Public Health, University of Calgary, Calgary, AB T2N 4Z6, Canada; E-Mails: (P.D.); (K.L.)
| | - Marie-Françoise Van Bressem
- Cetacean Conservation Medicine Group (CMED), Peruvian Centre for Cetacean Research (CEPEC), Pucusana, Lima 20, Peru; E-Mail:
| | - Jason D. Baker
- Pacific Islands Fisheries Science Center, National Marine Fisheries Service, NOAA, 1845 WASP Blvd., Building 176, Honolulu, Hawaii 96818, USA; E-Mails: (J.D.B.); (M.B.)
| | - Michelle Barbieri
- Pacific Islands Fisheries Science Center, National Marine Fisheries Service, NOAA, 1845 WASP Blvd., Building 176, Honolulu, Hawaii 96818, USA; E-Mails: (J.D.B.); (M.B.)
- The Marine Mammal Centre, Sausalito, CA 94965, USA; E-Mail:
| | - Kathleen M. Colegrove
- Zoological Pathology Program, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Maywood, IL 60153, USA; E-Mail:
| | - Sylvain De Guise
- Department of Pathobiology and Veterinary Science, and Connecticut Sea Grant College Program, University of Connecticut, Storrs, CT 06269, USA; E-Mail:
| | - Rik L. de Swart
- Department of Viroscience, Erasmus MC, 3015 CN Rotterdam, The Netherlands; E-Mail:
| | - Giovanni Di Guardo
- Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy; E-Mail:
| | - Andrew Dobson
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544-2016, USA; E-Mails: (A.D.); (B.G.); (S.E.M.)
| | - W. Paul Duprex
- Department of Microbiology, Boston University School of Medicine, Boston University, 620 Albany Street, Boston, MA 02118, USA; E-Mail:
| | - Greg Early
- Greg Early, Integrated Statistics, 87 Water St, Woods Hole, MA 02543, USA; E-Mail:
| | - Deborah Fauquier
- National Marine Fisheries Service/National Oceanographic and Atmospheric Administration, Marine Mammal Health and Stranding Response Program, Silver Spring, MD 20910, USA; E-Mails: (D.F.); (T.K.R.)
| | - Tracey Goldstein
- One Health Institute, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; E-Mail:
| | - Simon J. Goodman
- School of Biology, University of Leeds, Leeds LS2 9JT, UK; E-Mail:
| | - Bryan Grenfell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544-2016, USA; E-Mails: (A.D.); (B.G.); (S.E.M.)
- Fogarty International Center, National Institutes of Health, Bethesda, MD 20892-2220, USA
| | - Kátia R. Groch
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil; E-Mail:
| | - Frances Gulland
- The Marine Mammal Centre, Sausalito, CA 94965, USA; E-Mail:
- Marine Mammal Commission, 4340 East-West Highway, Bethesda, MD 20814, USA
| | - Ailsa Hall
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St. Andrews, St. Andrews, Fife KY16 8LB, UK; E-Mail:
| | - Brenda A. Jensen
- Department of Natural Sciences, Hawai’i Pacific University, Kaneohe, HI 96744, USA; E-Mail:
| | - Karina Lamy
- Department of Ecosystem and Public Health, University of Calgary, Calgary, AB T2N 4Z6, Canada; E-Mails: (P.D.); (K.L.)
| | - Keith Matassa
- Keith Matassa, Pacific Marine Mammal Center, 20612 Laguna Canyon Road, Laguna Beach, CA 92651, USA; E-Mail:
| | - Sandro Mazzariol
- Department of Comparative Biomedicine and Food Science, University of Padua, 35020 Legnaro Padua, Italy; E-Mail:
| | - Sinead E. Morris
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544-2016, USA; E-Mails: (A.D.); (B.G.); (S.E.M.)
| | - Ole Nielsen
- Department of Fisheries and Oceans Canada, Central and Arctic Region, 501 University Crescent, Winnipeg, MB R3T 2N6, Canada; E-Mail:
| | - David Rotstein
- David Rotstein, Marine Mammal Pathology Services, 19117 Bloomfield Road, Olney, MD 20832, USA; E-Mail:
| | - Teresa K. Rowles
- National Marine Fisheries Service/National Oceanographic and Atmospheric Administration, Marine Mammal Health and Stranding Response Program, Silver Spring, MD 20910, USA; E-Mails: (D.F.); (T.K.R.)
| | - Jeremy T. Saliki
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, GA 30602, USA; E-Mail:
| | - Ursula Siebert
- Institute for Terrestrial and Aquatic Wildlife Research, University of Veterinary Medicine Hannover 30173, Germany; E-Mail:
| | - Thomas Waltzek
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, FL 32611, USA; E-Mail:
| | - James F.X. Wellehan
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, FL 32610, USA; E-Mail:
| |
Collapse
|
44
|
Canine distemper virus antigen detection in external epithelia of recently vaccinated, sick dogs by fluorescence microscopy is a valuable prognostic indicator. J Clin Microbiol 2014; 53:687-91. [PMID: 25428156 DOI: 10.1128/jcm.02741-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Currently, there are no reliable predictors of the clinical outcomes of domesticated dogs that have been recently vaccinated against canine distemper virus (CDV) and develop respiratory disease. In this study, vaccinated dogs from Oklahoma City that were showing clinical signs of respiratory disease were evaluated for CDV antigen using a direct fluorescent antibody test (FAT). Clinical outcomes after standard symptomatic therapy for respiratory disease were recorded, and a statistical analysis of the results was performed. We present our study showing that CDV FAT results were predictive of clinical recovery (prognostic indicator, prospects of clinical recovery) among vaccinated dogs showing clinical signs of respiratory disease. Negative CDV FAT results equated to 80% chances of recovery after symptomatic therapy, compared to 55% chances of recovery when the CDV FAT results were positive. Based on the results of this study, we show that veterinarians can make better informed decisions about the clinical outcomes of suspected CDV cases, with 2-h turnaround times, by using the CDV FAT. Thus, antemortem examination with the CDV FAT on external epithelia of recently vaccinated, sick dogs is a clinically useful diagnostic test and valuable prognostic indicator for veterinarians. Application of the CDV FAT to these samples avoids unnecessary euthanasia of dogs with suspected CDV.
Collapse
|
45
|
Prevention of measles virus infection by intranasal delivery of fusion inhibitor peptides. J Virol 2014; 89:1143-55. [PMID: 25378493 DOI: 10.1128/jvi.02417-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
UNLABELLED Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV H and the fusion (F) envelope glycoprotein; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad-repeat (HR) regions of F can potently inhibit MV infection at the entry stage. We show here that specific features of H's interaction with its receptors modulate the susceptibility of MV F to peptide fusion inhibitors. A higher concentration of inhibitory peptides is required to inhibit F-mediated fusion when H is engaged to its nectin-4 receptor than when H is engaged to its CD150 receptor. Peptide inhibition of F may be subverted by continued engagement of receptor by H, a finding that highlights the ongoing role of H-receptor interaction after F has been activated and that helps guide the design of more potent inhibitory peptides. Intranasal administration of these peptides results in peptide accumulation in the airway epithelium with minimal systemic levels of peptide and efficiently prevents MV infection in vivo in animal models. The results suggest an antiviral strategy for prophylaxis in vulnerable and/or immunocompromised hosts. IMPORTANCE Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that parenterally delivered fusion-inhibitory peptides protect mice from lethal CNS MV disease. Here we show, using established small-animal models of MV infection, that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. Since the fusion inhibitors are stable at room temperature, this intranasal strategy is feasible even outside health care settings, could be used to protect individuals and communities in case of MV outbreaks, and could complement global efforts to control measles.
Collapse
|
46
|
Delpeut S, Noyce RS, Richardson CD. The tumor-associated marker, PVRL4 (nectin-4), is the epithelial receptor for morbilliviruses. Viruses 2014; 6:2268-86. [PMID: 24892636 PMCID: PMC4074928 DOI: 10.3390/v6062268] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/14/2014] [Accepted: 05/15/2014] [Indexed: 01/25/2023] Open
Abstract
PVRL4 (nectin-4) was recently identified as the epithelial receptor for members of the Morbillivirus genus, including measles virus, canine distemper virus and peste des petits ruminants virus. Here, we describe the role of PVRL4 in morbillivirus pathogenesis and its promising use in cancer therapies. This discovery establishes a new paradigm for the spread of virus from lymphocytes to airway epithelial cells and its subsequent release into the environment. Measles virus vaccine strains have emerged as a promising oncolytic platform for cancer therapy in the last ten years. Given that PVRL4 is a well-known tumor-associated marker for several adenocarcinoma (lung, breast and ovary), the measles virus could potentially be used to specifically target, infect and destroy cancers expressing PVRL4.
Collapse
Affiliation(s)
- Sebastien Delpeut
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| | - Ryan S Noyce
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| | - Christopher D Richardson
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| |
Collapse
|
47
|
Identification of amino acid substitutions with compensational effects in the attachment protein of canine distemper virus. J Virol 2014; 88:8057-64. [PMID: 24807725 DOI: 10.1128/jvi.00454-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The hemagglutinin (H) gene of canine distemper virus (CDV) encodes the receptor-binding protein. This protein, together with the fusion (F) protein, is pivotal for infectivity since it contributes to the fusion of the viral envelope with the host cell membrane. Of the two receptors currently known for CDV (nectin-4 and the signaling lymphocyte activation molecule [SLAM]), SLAM is considered the most relevant for host susceptibility. To investigate how evolution might have impacted the host-CDV interaction, we examined the functional properties of a series of missense single nucleotide polymorphisms (SNPs) naturally accumulating within the H-gene sequences during the transition between two distinct but related strains. The two strains, a wild-type strain and a consensus strain, were part of a single continental outbreak in European wildlife and occurred in distinct geographical areas 2 years apart. The deduced amino acid sequence of the two H genes differed at 5 residues. A panel of mutants carrying all the combinations of the SNPs was obtained by site-directed mutagenesis. The selected mutant, wild type, and consensus H proteins were functionally evaluated according to their surface expression, SLAM binding, fusion protein interaction, and cell fusion efficiencies. The results highlight that the most detrimental functional effects are associated with specific sets of SNPs. Strikingly, an efficient compensational system driven by additional SNPs appears to come into play, virtually neutralizing the negative functional effects. This system seems to contribute to the maintenance of the tightly regulated function of the H-gene-encoded attachment protein. Importance: To investigate how evolution might have impacted the host-canine distemper virus (CDV) interaction, we examined the functional properties of naturally occurring single nucleotide polymorphisms (SNPs) in the hemagglutinin gene of two related but distinct strains of CDV. The hemagglutinin gene encodes the attachment protein, which is pivotal for infection. Our results show that few SNPs have a relevant detrimental impact and they generally appear in specific combinations (molecular signatures). These drastic negative changes are neutralized by compensatory mutations, which contribute to maintenance of an overall constant bioactivity of the attachment protein. This compensational mechanism might reflect the reaction of the CDV machinery to the changes occurring in the virus following antigenic variations critical for virulence.
Collapse
|
48
|
Delpeut S, Noyce RS, Richardson CD. The V domain of dog PVRL4 (nectin-4) mediates canine distemper virus entry and virus cell-to-cell spread. Virology 2014; 454-455:109-17. [PMID: 24725937 DOI: 10.1016/j.virol.2014.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 01/25/2014] [Accepted: 02/11/2014] [Indexed: 11/18/2022]
Abstract
The entry of canine distemper virus (CDV) is a multistep process that involves the attachment of CDV hemagglutinin (H) to its cellular receptor, followed by fusion between virus and cell membranes. Our laboratory recently identified PVRL4 (nectin-4) to be the epithelial receptor for measles and canine distemper viruses. In this study, we demonstrate that the V domain of PVRL4 is critical for CDV entry and virus cell-to-cell spread. Furthermore, four key amino acid residues within the V domain of dog PVRL4 and two within the CDV hemagglutinin were shown to be essential for receptor-mediated virus entry.
Collapse
Affiliation(s)
- Sebastien Delpeut
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5; IWK Health Centre, Canadian Center for Vaccinology, Goldbloom Pavilion, Halifax, Nova Scotia, Canada B3H 1X5
| | - Ryan S Noyce
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5; IWK Health Centre, Canadian Center for Vaccinology, Goldbloom Pavilion, Halifax, Nova Scotia, Canada B3H 1X5
| | - Christopher D Richardson
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5; IWK Health Centre, Canadian Center for Vaccinology, Goldbloom Pavilion, Halifax, Nova Scotia, Canada B3H 1X5; The Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
49
|
Measles vaccination of nonhuman primates provides partial protection against infection with canine distemper virus. J Virol 2014; 88:4423-33. [PMID: 24501402 DOI: 10.1128/jvi.03676-13] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Measles virus (MV) is being considered for global eradication, which would likely reduce compliance with MV vaccination. As a result, children will grow up without MV-specific immunity, creating a potential niche for closely related animal morbilliviruses such as canine distemper virus (CDV). Natural CDV infection causing clinical signs has never been reported in humans, but recent outbreaks in captive macaques have shown that CDV can cause disease in primates. We studied the virulence and tropism of recombinant CDV expressing enhanced green fluorescent protein in naive and measles-vaccinated cynomolgus macaques. In naive animals CDV caused viremia and fever and predominantly infected CD150(+) lymphocytes and dendritic cells. Virus was reisolated from the upper and lower respiratory tracts, but infection of epithelial or neuronal cells was not detectable at the time points examined, and the infections were self-limiting. This demonstrates that CDV readily infects nonhuman primates but suggests that additional mutations are necessary to achieve full virulence in nonnatural hosts. Partial protection against CDV was observed in measles-vaccinated macaques, as demonstrated by accelerated control of virus replication and limited shedding from the upper respiratory tract. While neither CDV infection nor MV vaccination induced detectable cross-reactive neutralizing antibodies, MV-specific neutralizing antibody levels of MV-vaccinated macaques were boosted by CDV challenge infection, suggesting that cross-reactive VN epitopes exist. Rapid increases in white blood cell counts in MV-vaccinated macaques following CDV challenge suggested that cross-reactive cellular immune responses were also present. This study demonstrates that zoonotic morbillivirus infections can be controlled by measles vaccination. IMPORTANCE Throughout history viral zoonoses have had a substantial impact on human health. Given the drive toward global eradication of measles, it is essential to understand the zoonotic potential of animal morbilliviruses. Morbilliviruses are thought to have evolved from a common ancestral virus that jumped species and adapted to new hosts. Recently, canine distemper virus (CDV), a morbillivirus normally restricted to carnivores, caused disease outbreaks in nonhuman primates. Here, we report that experimental CDV infection of monkeys resulted in fever and leukopenia. The virus replicated to high levels in lymphocytes but did not spread to epithelial cells or the central nervous system. Importantly, like measles virus in macaques, the infections were self-limiting. In measles-vaccinated macaques CDV was cleared more rapidly, resulting in limited virus shedding from the upper respiratory tract. These studies demonstrate that although CDV can readily infect primates, measles immunity is protective, and CDV infection is self-limiting.
Collapse
|
50
|
Ludlow M, Rennick LJ, Nambulli S, de Swart RL, Duprex WP. Using the ferret model to study morbillivirus entry, spread, transmission and cross-species infection. Curr Opin Virol 2013; 4:15-23. [PMID: 24525290 DOI: 10.1016/j.coviro.2013.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 01/26/2023]
Abstract
Canine distemper virus (CDV) is an animal morbillivirus with a worldwide circulation that infects carnivores, including domestic dogs and an assortment of wildlife hosts. The development of reverse genetics systems for wild-type strains of CDV and the use of the resulting recombinant (r) viruses to infect ferrets by a natural route has shed new light on the temporal pathogenesis of distemper. Combining fluorescent protein expressing recombinant viruses and multimodal, macroscopic and microscopic imaging modalities has highlighted the differential role of the cellular receptors CD150 and PVRL4 in disease progression. This in turn has enabled pathways of viral spread, including multiple routes of entry into the central nervous system, to be mapped with unparalleled sensitivity.
Collapse
Affiliation(s)
- Martin Ludlow
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Linda J Rennick
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Sham Nambulli
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Rik L de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - W Paul Duprex
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA; School of Medicine, Dentistry and Biomedical Sciences, The Queen's University of Belfast, Belfast, Northern Ireland, UK.
| |
Collapse
|